Transforming growth factor-β (TGF-β) is a key player in malignant disease through its actions on host tissues and cells. Malignant cells often secrete large amounts of TGF-β that act on nontransformed cells present in the tumor mass as well as distal cells in the host to suppress antitumor immune responses creating an environment of immune tolerance, augmenting angiogenesis, invasion and metastasis, and increasing tumor extracellular matrix deposition. Cells of the innate immune system contribute to the high concentrations of TGF-β found in tumor masses. In addition, dendritic cell subpopulations secreting TGF-β contribute to the generation of regulatory T cells that actively inhibit the activity of other T cells. Elevated levels of plasma TGF-β are associated with advanced stage disease and may separate patients into prognostically high-risk populations. Anti–TGF-β therapy could reverse the immunosuppressive effects of this cytokine on the host as well as decrease extracellular matrix formation, decrease angiogenesis, decrease osteolytic activity, and increase the sensitivity of the malignant cells to cytotoxic therapies and immunotherapies. Phase I clinical trials of an inhibitor of TGF-β receptor type I kinase activity and a TGF-β neutralizing antibody are under way.

Despite all that we have learned about genetic alterations that occur in malignant cells, the development of highly effective anticancer therapeutics remains a stunningly difficult task because malignant cells are so similar to normal cells. This statement is true and yet untrue. The untruth comes from our knowledge that the immune system can recognize and eliminate cells that are slightly abnormal through immune surveillance. The truth comes from our knowledge that frankly malignant cells suppress the immune system and survive and thrive by coopting varied nearby and distal host cells to support the malignant disease (14). An effective immune system recognizes and destroys a wide diversity of pathogens, recognizes and destroys altered or damaged self, including apoptotic and necrotic cells, exhausted cells and malignant cells, and is tolerant to self. The theory of immune surveillance is that the immune system continuously recognizes and eliminates tumor cells. When a tumor escapes immune surveillance, it is frankly malignant and can grow to become lethal to the host.

Transforming growth factor-β (TGF-β) is a key player in malignant disease through its actions on host tissues and cells. Aberrant expression of TGF-β results in profound changes in the genetic stability of cells leading to alteration of both the differentiation state of the cells, altered interaction of the cells with the host environment, and the generation of therapy-resistant disease. Malignant cell resistance to TGF-β is frequently due to loss, silencing, or mutational inactivation of genes in the TGF-β signaling pathway including the type I and type II receptors and receptor-associated and common-mediator Smads (5, 6).

In cells with intact canonical TGF-β signaling, The binding of a TGF-β (isoform-1, -2, or -3) to the TGF-β type II receptor enables the formation of a heteromeric complex between TGF-β type I and type II receptors. The type I receptor is phosphorylated by the type II receptor serine/threonine kinase. The activated type I receptor phosphorylates selected receptor-activated Smads that then complex with Smad4. The Smad complexes translocate into the nucleus. Activated Smad complexes regulate the transcription of target genes through direct or indirect interaction with DNA-binding transcription factors or coactivators. Activation of receptor-activated Smads can be inhibited by Smad6 or Smad7 (5, 6).

TGF-β knockout mice suffer from a lethal multifocal inflammatory disease that shows the importance of TGF-β in maintaining immune system homeostasis (79). The blockade of TGF-β signaling in T cells by transfection with a dominant-negative type II receptor or in bone marrow by conditional knockout of the TGF-β type II receptor, results in similar multifocal inflammatory responses (10, 11). On the other hand, transgenic expression of a dominant-negative type II receptor specifically in T cells of C57BL6 mice prevented the growth of the syngeneic tumors EL-4 thymoma (i.p.) and B16F10 melanoma (i.v.) in the animals (12).

Malignant cells often secrete large amounts of TGF-β that act on nontransformed cells present in the tumor mass as well as distal cells in the host to suppress antitumor immune responses creating an environment of immune tolerance, augmenting angiogenesis, invasion and metastasis, and increasing tumor extracellular matrix deposition. In addition, within the tumor and tumor vicinity, TGF-β may be released from the extracellular matrix or secreted by mesenchymal cells, resident leukocytes, or by monocytes and macrophages recruited to the tumor (1316).

TGF-β is a potent suppressor of the immune system. With broad activity over natural killer (NK) cells, T cells, monocytes/macrophages, and dendritic cells, TGF-β can affect the initiation and stimulation of both primary and secondary immune responses as well as suppress antitumor effector cells (1725). TGF-β in lung and colorectal cancer patient plasma samples directly suppressed NK cell activity, a defect that could be reversed with anti–TGF-β antibodies (24).

Cells of the innate immune system including neutrophils, NK cells, monocytes, dendritic cells, and monocyte-derived macrophages contribute to the high concentrations of TGF-β found in tumor masses (Table 1). Tumor-infiltrating macrophages may be particularly instrumental to malignancy because they adopt a trophic role that results in extracellular matrix breakdown, angiogenesis, and tumor cell motility, thus facilitating tumor growth and metastasis (26). Active TGF-β binds to the TGF-β receptors in tumor-infiltrating lymphocytes including CD4+, CD8+, or NK cells and alters their phenotype, proliferation, and cytokine secretion (ref. 27; Fig. 1). Tumor-infiltrating dendritic cells secrete TGF-β and respond to TGF-β and interleukin 10 with markedly down-regulated expression of the costimulatory molecules CD80, CD86, and CD40, and markedly decreased secretion of tumor necrosis factor-α, interleukin 12, and CCL5/RANTES (28). Dendritic cell subpopulations (CD4CD8 dendritic cells) secreting TGF-β have been implicated in the generation of CD4+ regulatory T cells (29). Regulatory T cell differentiation can be driven by immature or “tolerogenic” dendritic cells (30). Regulatory T cells actively inhibit the activity of other T cells. There are two classes of regulatory T cells: natural Tregs differentiate in the thymus and induced or “adaptive” Tregs that arise in the periphery. The most common phenotypes for Tregs are CD4+CD25+Foxp3+ and CD8+CD25+Foxp3+. However, this is a very active area of investigation and other potential phenotypes involve CTLA4 and TGF-β. The action of natural (innate) Tregs may be different from the mechanism of action of induced (adaptive) Tregs with one involving cell contact and the other involving secretion of TGF-β or other cytokines. TGF-β is important in both the induction of the regulatory phenotype in peripheral Tregs and in the effector function of Tregs. The presence of Tregs in a tumor produces an environment of immune privilege (15). By direct cell-to-cell contact, Tregs deliver inhibitory signals to CD4+, CD8+, and NK cells to produce host tolerance to tumors (31). Tregs migrate to and are retained in tumor tissue and may account for >20% of tumor-infiltrating T cells; thus, providing immune privilege for the malignancy (32, 33). Treg and TGF-β alter the innate immune response through direct interaction with NK cells and Th17 cells (15, 34). Similarly, suppression of CD8+ cytolytic T cell activity by TGF-β from T regulatory cells or from the local tumor environment can be reversed by neutralizing TGF-β or by genetically rendering the effector T cells insensitive to its effects (3537). Finally, anti–TGF-β antibodies could enhance immune responses to vaccine therapies (22, 35). All these effects combine to make TGF-β a key factor in the suppression of the immune system and make it an ideal target for cancers in which antitumor immunity seems to play an important role in the control of the disease.

Table 1.

Local and distal TGF-β–secreting cells involved in maintaining the tolerogenic sanctuary and immune privilege environment that occurs in malignant disease

Local tolerogenic sanctuary and immune privilege tumor microenvironment (TGF-β producing cells)
1. Malignant cells 
2. Mesenchymal cells 
3. Treg regulatory T cells 
4. Macrophage 
5. Neutrophils 
6. NK T cells 
7. Monocytes 
8. Dendritic cells 
9. Mast cells 
10. Platelets 
Local tolerogenic sanctuary and immune privilege tumor microenvironment (TGF-β producing cells)
1. Malignant cells 
2. Mesenchymal cells 
3. Treg regulatory T cells 
4. Macrophage 
5. Neutrophils 
6. NK T cells 
7. Monocytes 
8. Dendritic cells 
9. Mast cells 
10. Platelets 
Fig. 1.

Alterations in behavior and secreted proteins that occur in immune system cells under the influence of a TGF-β–rich environment.

Fig. 1.

Alterations in behavior and secreted proteins that occur in immune system cells under the influence of a TGF-β–rich environment.

Close modal

The majority of tumors from patients with advanced breast cancer as well as several other malignancies have been reported to be refractory to TGF-β–induced growth inhibition and many produce large amounts of this cytokine (refs. 38, 39; Table 2). In addition, elevated levels of plasma TGF-β have been associated with advanced stage disease and might separate patients into prognostically high-risk populations (4042). It is believed that active TGF-β produced by the tumor and local stroma contributes to the progression and metastatic potential of this cancer through autocrine and paracrine effects (43). As with breast cancer, TGF-β plasma levels are elevated in patients with prostate cancer, and these levels correlate with advanced stage, metastases, and poorer clinical outcome (4447). Increased TGF-β expression has been observed in both tumor cells and in tumor stroma (4850). Similarly, TGF-β plasma levels are elevated in patients with pancreatic cancer and these levels correlate with advanced stage, metastases, and poorer clinical outcome (51).

Table 2.

Human malignant diseases in which elevated levels of TGF-β have been implicated as contributing to poor outcome

Patients/tumor typeReferences
Breast cancer (38–43) 
Prostate cancer (44–50, 76) 
Renal cell cancer (52, 53) 
Melanoma (58) 
Pancreatic cancer (51) 
Multiple myeloma (61, 62) 
Non–Hodgkin's Lymphoma (63–65) 
Non–small cell lung cancer (77) 
Small cell lung cancer (78, 79) 
Colorectal cancer (80) 
Ovarian cancer (81) 
Cervical cancer (82) 
Bladder cancer (83, 84) 
Kaposi sarcoma (85) 
Glioma (86) 
Head and neck cancer (87) 
Thyroid cancer (88 
Esophageal cancer (89) 
Gastric cancer (90) 
Hepatocellular cancer (91, 92) 
Patients/tumor typeReferences
Breast cancer (38–43) 
Prostate cancer (44–50, 76) 
Renal cell cancer (52, 53) 
Melanoma (58) 
Pancreatic cancer (51) 
Multiple myeloma (61, 62) 
Non–Hodgkin's Lymphoma (63–65) 
Non–small cell lung cancer (77) 
Small cell lung cancer (78, 79) 
Colorectal cancer (80) 
Ovarian cancer (81) 
Cervical cancer (82) 
Bladder cancer (83, 84) 
Kaposi sarcoma (85) 
Glioma (86) 
Head and neck cancer (87) 
Thyroid cancer (88 
Esophageal cancer (89) 
Gastric cancer (90) 
Hepatocellular cancer (91, 92) 

TGF-β plasma levels are elevated in patients with renal cell cancer (52, 53). TGF-β neutralization can be an effective therapy in animal models of renal cell cancer. In some animal models, the benefits from TGF-β neutralization may be additive or synergistic when combined with chemotherapy (54, 55). In addition, renal cell carcinomas are sensitive to antitumor immunotherapy including immunomodulators such as interleukin 2, lymphocyte-activated killer cells, tumor-infiltrating lymphocyte therapy, and vaccine approaches. It remains to be determined if neutralization of TGF-β could enhance immunity and be effective in combination with immunotherapeutic approaches (56, 57).

Malignant melanoma has increased TGF-β expression in tumor cells, which is not observed in benign or in situ lesions (58). Higher expression of TGF-β is associated with metastatic lesions and deeper invasion (worse prognosis, Clark's level 3 and higher) in this disease (58). In transgenic models in which T cells are rendered insensitive to TGF-β, animals are able to completely eradicate tumors such as B16 melanoma. Similarly, anti–TGF-β antibodies enhance tumor-specific immune responses, but have not been successful in completely eradicating tumors (12, 59, 60). Gorelik and Flavell suggested that this might be due to the incomplete neutralization of TGF-β (12).

Elevated serum levels of TGF-β have been observed in patients with myeloma and have correlated with higher levels of serum β2-microglobulin, which is an adverse prognostic marker of this disease (61). Sorted CD38+CD45RA− myeloma cells secrete significantly more TGF-β than peripheral blood mononuclear cells, splenic B cells, or CD40 ligand-activated B cells. In addition, TGF-β secretion by myeloma bone marrow stromal/mononuclear cells was significantly greater than by normal bone marrow mononuclear cells. Therefore, in patients with myeloma, the source of TGF-β is clearly from the malignant cells as well as bone marrow stromal cells (62). TGF-β levels are elevated in non–Hodgkin's lymphoma and are markedly elevated in high-grade lymphomas, cutaneous T cell lymphomas with a T-regulatory phenotype, and in splenic marginal zone lymphomas presenting as myelofibrosis (6365). Non–Hodgkin's lymphomas are sensitive to antitumor immunotherapy including antibody therapies and cell therapies, as well as vaccine approaches. The idiotype proteins on B cell non–Hodgkin's lymphomas are tumor-specific antigens, which are effective targets for monoclonal antibodies as well as for vaccine approaches (6670). Unmaintained remissions of >10 years have been seen in patients treated with specific anti-idiotype antibodies or idiotype vaccines (67, 68). The reversal of TGF-β–induced immunosuppression might improve both the induction of primary immune responses and the maintenance of effective cytotoxic T cell activity against these malignancies.

Malignant disease grips the host influencing the behavior of cells in the vicinity of the tumor and distal cells and tissues as well. TGF-β, a secreted protein, is a key player in the malignant process. Anti–TGF-β therapy could reverse the immunosuppressive effects of this cytokine on the host as well as decrease extracellular matrix formation, decrease angiogenesis, decrease osteolytic activity, and increase the sensitivity of the malignant cells to cytotoxic therapies and immunotherapies (7175). Phase I clinical trials are under way, a small molecule inhibitor of TGF-β receptor type I kinase activity and with a fully human monoclonal antibody that neutralizes the three isoforms of TGF-β.

1
Teicher BA, Herman TS, Holden SA, et al. Tumor resistance to alkylating agents conferred by mechanisms operative only in vivo.
Science
1990
;
247
:
1457
–61.
2
Teicher BA. A systems approach to cancer therapy (antiangiogenics + standard cytotoxics' mechanism(s) of interaction).
Cancer Metastasis Rev
1996
;
15
:
247
–72.
3
Teicher BA, Maehara, Kakeji Y, et al. Reversal of in vivo drug resistance by the transforming growth factor-β inhibitor decorin.
Int J Cancer
1997
;
71
:
49
–58.
4
Teicher BA. Malignant cells, directors of the malignant process: role of transforming growth factor-β.
Cancer Metastasis Rev
2001
;
20
:
133
–43.
5
ten Dijke P, Hill CS. New insights into TGF-β-Smad signaling.
Trends Biochem Sci
2004
;
29
:
265
–73.
6
Janssens K, ten Dijke P, Janssens S, Van Hul W. Transforming growth factor-β1 to the bone.
Endocr Rev
2005
;
26
:
743
–74.
7
Letterio JJ, Roberts AB. Regulation of immune responses by TGF-β.
Annu Rev Immunol
1998
;
16
:
137
–61.
8
Kulkarni AB, Huh C, Becker D, et al. Transforming growth factor β1 null mutation in mice causes excessive inflammatory response and early death.
Proc Natl Acad Sci U S A
1993
;
90
:
770
–4.
9
Shull MM, Ormsby I, Kier AB, et al. Targeted disruption of the mouse transforming growth factor-β gene results in multifocal inflammatory disease.
Nature
1992
;
359
:
693
–9.
10
Gorelik L, Flavell RA. Abrogation of TGF-β signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease.
Immunity
2000
;
12
:
171
–81.
11
Leveen P, Larsson, Ehinger M, et al. Induced disruption of the transforming growth factor β type II receptor gene in mice causes a lethal inflammatory disorder that is transplantable.
Blood
2002
;
100
:
560
–8.
12
Gorelik L, Flavell RA. Immune-related eradication of tumors through the blockade of transforming growth factor-β signaling in T cells.
Nat Med
2001
;
7
:
1118
–22.
13
Yingling JM, Blanchard KL, Sawyer JS. Development of TGF-β signaling inhibitors for cancer therapy.
Nat Rev Drug Discov
2004
;
3
:
1011
–22.
14
Biswas S, Criswell TL, Wang SE, Arteaga CL. Inhibition of transforming growth factor-β signaling in human cancer: targeting a tumor suppressor network as a therapeutic strategy.
Clin Cancer Res
2006
;
12
:
4142
–6.
15
Wahl SM, Wen J, Moutsopoulos N. TGF-β: a mobile purveyor of immune privilege.
Immunol Rev
2006
;
213
:
213
–27.
16
Wahl SM. Transforming growth factor-β: innately bipolar.
Curr Opin Immunol
2007
;
19
:
55
–62.
17
Arteaga CL, Hurd SD, Winnier AR, Johnson MD, Fendly BM, Forbes JT. Anti-transforming growth factor (TGF)-β antibodies inhibit breast cancer cell tumorigenicity and increase mouse spleen natural killer cell activity. Implications for a possible role of tumor cell/host TGF-β interactions in human breast cancer progression.
J Clin Invest
1993
;
92
:
2569
–76.
18
Ludviksson BR, Seegers D, Resnick AS, Strober W. The effect of TGF-β1 on immune responses of naive versus memory CD4+ Th1/Th2 T cells.
Eur J Immunol
2000
;
30
:
2101
–11.
19
Nakamura K, Kitani A, Strober W. Cell contact-dependent immunosuppression by CD4(+)CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor β.
J Exp Med
2001
;
194
:
629
–44.
20
Woo EY, Chu CS, Goletz TJ, et al. Regulatory CD4(+)CD25(+) T cells in tumors from patients with early-stage non-small cell lung cancer and late-stage ovarian cancer.
Cancer Res
2001
;
61
:
4766
–72.
21
Kao JY, Gong Y, Chen CM, Zheng QD, Chen JJ. Tumor-derived TGF-β reduces the efficacy of dendritic cell/tumor fusion vaccine.
J Immunol
2003
;
170
:
3806
–11.
22
Kobie JJ, Wu RS, Kurt RA, Lou S, Adelman MK, Whitesell LJ. Transforming growth factor β inhibits the antigen-presenting functions and antitumor activity of dendritic cell vaccines.
Cancer Res
2003
;
63
:
1860
–4.
23
Terabe M, Matsui S, Park J-M, Mamura, et al. Transforming growth factor-β production and myeloid cells are an effector mechanism through which CD1d-restricted T cells block cytotoxic T lymphocyte-mediated tumor immunosurveillance: abrogation prevents tumor recurrence.
J Exp Med
2003
;
198
:
1741
–52.
24
Lee JC, Lee KM, Kim DW, Heo DS. Elevated TGF-β1 secretion and down-modulation of NKG2D underlies impaired NK cytotoxicity in cancer patients.
J Immunol
2004
;
172
:
7335
–40.
25
Terabe M, Berzofsky JA. Immunoregulatory T cells in tumor immunity.
Curr Opin Immunol
2004
;
16
:
157
–62.
26
Pollard JW. Tumor-educated macrophages promote tumor progression and metastasis.
Nat Rev Cancer
2004
;
4
:
71
–8.
27
Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA. Transforming growth factor-β regulation of immune responses.
Annu Rev Immunol
2006
;
24
:
99
–146.
28
Larmonier N, Marron M, Zeng Y, et al. Tumor-derived CD4(+)CD25(+)regulatory T cell suppression of dendritic cell function involves TGF-β and IL-10.
Cancer Immunol Immunother
2007
;
56
:
48
–59.
29
Zhang X, et al. CD4-8− dendritic cells prime CD4+ T regulatory 1 cells to suppress anti-tumor immunity.
J Immunol
2005
;
175
:
2931
–7.
30
Roncarolo MG, Levings MK, Traversari C. Differentiation of T regulatory cells by immature dendritic cells.
J Exp Med
2001
;
193
:
F5
–9.
31
Khazaie K, von Boehmer H. The impact of CD4+CD25+ Treg on tumor specific CD8+ T cell cytotoxicity and cancer.
Semin Cancer Biol
2006
;
16
:
124
–36.
32
Beyer M, Schultze JL. Regulatory T cells in cancer.
Blood
2006
;
108
:
804
–11.
33
Curiel TJ, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival.
Nat Med
2004
;
10
:
942
–9.
34
Mangan PR, Harrington LE, O'Quinn DB, et al. Transforming growth factor-β induces development of the Th17 lineage.
Nature
2006
;
441
:
231
–4.
35
Tzai Ts, Shiau Al, Lin CS, Wu Cl, Lin JS. Modulation of the immunostimulating effect of autologous tumor vaccine by anti-TGF-β antibody and interfereon-α on murine MBT-2 bladder cancer.
Anticancer Res
1997
;
17
:
1073
–8.
36
Zhang Q, Yang X, Pins M, et al. Adoptive transfer of tumor-reactive transforming growth factor-β-insensitive CD8+ T cells: eradication of autologous mouse prostate cancer.
Cancer Res
2005
;
65
:
1761
–9.
37
Ahmadzadeh M, Rosenberg SA. TGF-{beta}1 attenuates the acquisition and expression of effector function by tumor antigen-specific human memory CD8 T cells.
J Immunol
2005
;
174
:
5215
–23.
38
Reiss M, Barcellos-Hoff MH. Transforming growth factor-β in breast cancer: a working hypothesis.
Breast Cancer Res Treat
1997
;
45
:
81
–95.
39
Baillie R, Coombes RC, Smith J. Multiple forms of TGF-β1 in breast tissues: a biologically active form of the small latent complex of TGF-β1.
Eur J Cancer
1996
;
32A
:
1566
–73.
40
Ivanovic V, Todorovic-Rakovic N, Demajo M, et al. Elevated plasma levels of transforming growth factor-β1 (TGF-β1) in patients with advanced breast cancer: association with disease progression.
Eur J Cancer
2003
;
39
:
454
–61.
41
Nikolic-Vukosavljevic D, Todorovic-Rakovic N, Demajo M, et al. Plasma TGF-β1-related survival of postmenopausal metastatic breast cancer patients.
Clin Exp Metastasis
2004
;
21
:
581
–5.
42
Buck MB, Fritz P, Dippon J, Zugmaier G, Knabbe C. Prognostic significance of transforming growth factor β receptor II in estrogen receptor-negative breast cancer patients.
Clin Cancer Res
2004
;
10
:
491
–8.
43
Dumont N, Arteaga CL. Transforming growth factor-β and breast cancer: tumor promoting effects of transforming growth factor-β.
Breast Cancer Res
2000
;
2
:
125
–32.
44
Ivanovic V, Melman A, Davis-Joseph B, Valcic M, Geliebter J. Elevated plasma levels of TGF-β1 in patients with invasive prostate cancer.
Nat Med
1995
;
1
:
282
–4.
45
Wikstrom P, Stattin P, Franck-Lissbrant I, Damber JE, Bergh A. Transforming growth factor β1 is associated with angiogenesis, metastasis, and poor clinical outcome in prostate cancer.
Prostate
1998
;
37
:
19
–29.
46
Sinnreich O, Kratzsch J, Reichenbach A, Glaser C, Huse K, Birkenmeier G. Plasma levels of transforming growth factor-1β and α2-macroglobulin before and after radical prostatectomy: association to clinicopathological parameters.
Prostate
2004
;
61
:
201
–8.
47
Shariat SF, Lamb DJ, Kattan MW, et al. Association of preoperative plasma levels of insulin-like growth factor I and insulin-like growth factor binding proteins-2 and -3 with prostate cancer invasion, progression, and metastasis.
J Clin Oncol
2002
;
20
:
833
–41.
48
Eastham JA, Truong LD, Rogers E, et al. Transforming growth factor-β1: comparative immunohistochemical localization in human primary and metastatic prostate cancer.
Lab Invest
1995
;
73
:
628
–35.
49
Gerdes MJ, Larsen M, McBride L, Dang TD, Lu B, Rowley DR. Localization of transforming growth factor-β1 and type II receptor in developing normal human prostate and carcinoma tissues.
J Histochem Cytochem
1998
;
46
:
379
–88.
50
Steiner MS, Zhou ZZ, Tonb DC, Barrack ER. Expression of transforming growth factor-β1 in prostate cancer.
Endocrinology
1994
;
135
:
2240
–7.
51
Friess H, Yamanaka Y, Buchler M, et al. Enhanced expression of transforming growth factor β isoforms in pancreatic cancer correlates with decreased survival.
Gastroenterology
1993
;
105
:
1846
–56.
52
Junker U, Knoefel B, Nuske K, et al. Transforming growth factor β1 is significantly elevated in plasma of patients suffering from renal cell carcinoma.
Cytokine
1996
;
8
:
794
–8.
53
Wunderlich H, Steiner T, Kosmehl H, et al. Increased transforming growth factor β1 plasma level in patients with renal cell carcinoma: a tumor-specific marker?
Urol Int
1998
;
60
:
205
–7.
54
Yagasaki H, Kawata N, Takimoto Y, Nemoto N. Histopathological analysis of angiogenic factors in renal cell carcinoma.
Int J Urol
2003
;
10
:
220
–7.
55
Ananth S, Knebelmann B, Gruning W, et al. Transforming growth factor β1 is a target for the von Hippel-Lindau tumor suppressor and a critical growth factor for clear cell renal carcinoma.
Cancer Res
1999
;
59
:
2210
–6.
56
Avigan D. Dendritic cell-tumor fusion vaccines for renal cell carcinoma.
Clin Cancer Res
2004
;
10
:
6347
–52S.
57
Avigan D, Vasir B, Gong J, et al. Fusion cell vaccination of patients with metastatic breast and renal cancer induces immunological and clinical responses.
Clin Cancer Res
2004
;
10
:
4699
–708.
58
Reed JA, Mcnutt NS, Prieto VG, Albino AP. Expression of transforming growth factor-β2 in malignant melanoma correlates with the depth of tumor invasion. implications for tumor progression.
Am J Pathol
1994
;
145
:
97
–104.
59
Shah AH, Tabayoyong WB, Kundu SD, et al. Suppression of tumor metastasis by blockade of transforming growth factor β signaling in bone marrow cells through a retroviral-mediated gene therapy in mice.
Cancer Res
2002
;
62
:
7135
–8.
60
Maeda H, Shiraishi A. TGF-β contributes to the shift toward Th2-type responses through direct and IL-10-mediated pathways in tumor-bearing mice.
J Immunol
1996
;
156
:
73
–8.
61
Urba Ska-Rys H, Wierzbowska A, Robak T. Circulating angiogenic cytokines in multiple myeloma and related disorders.
Eur Cytokine Netw
2003
;
14
:
40
–51.
62
Urashima M, Ogata A, Chauhan D, et al. Transforming growth factor-β1: differential effects on multiple myeloma versus normal B cells.
Blood
1996
;
87
:
1928
–38.
63
Woszczyk D, Gola J, Jurzak M, Mazurek U, Mykala-Ciesla J, Wilczok T. Expression of TGF β1 genes and their receptor types I, II, and III in low- and high-grade malignancy non-Hodgkin's lymphomas.
Med Sci Monit
2004
;
10
:
CR33
–7.
64
Berger CL, Tigelaar R, Cohen J, et al. Cutaneous T-cell lymphoma: malignant proliferation of T-regulatory cells.
Blood
2005
;
105
:
1640
–7.
65
Matsunaga T, Takemoto N, Miyajima N, et al. Splenic marginal zone lymphoma presenting as myelofibrosis associated with bone marrow involvement of lymphoma cells which secrete a large amount of TGF-β.
Ann Hematol
2004
;
83
:
322
–5.
66
Hsu FJ, Benike C, Fagnoni F, et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells.
Nat Med
1996
;
2
:
52
–8.
67
Hsu FJ, Caspar CB, Czerwinski D, et al. Tumor-specific idiotype vaccines in the treatment of patients with B-cell lymphoma-long-term results of a clinical trial.
Blood
1997
;
89
:
3129
–35.
68
Davis TA, Maloney DG, Czerwinski DK, Liles TM, Levy R. Anti-idiotype antibodies can induce long-term complete remissions in non-Hodgkin's lymphoma without eradicating the malignant clone.
Blood
1998
;
92
:
1184
–90.
69
Maloney DG, Brown S, Czerwinski DK, et al. Monoclonal anti-idiotype antibody therapy of B-cell lymphoma: the addition of a short course of chemotherapy does not interfere with the antitumor effect nor prevent the emergence of idiotype-negative variant cells.
Blood
1992
;
80
:
1502
–10.
70
Miller RA, Maloney DG, Warnke R, Levy R. Treatment of B-cell lymphoma with monoclonal anti-idiotype antibody.
N Engl J Med
1982
;
306
:
517
–22.
71
Wojtowicz-Praga S. Reversal of tumor-induced immunosuppression by TGF-β inhibitors.
Invest New Drugs
2003
;
21
:
21
–32.
72
Biswas S, Guix M, Rinehart C, et al. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression.
J Clin Invest
2007
;
117
:
1305
–13.
73
Hayashi T, Hideshima T, Nguyen AN, et al. Transforming growth factor β receptor I kinase inhibitor down-regulates cytokine secretion and multiple myeloma cell growth in the bone marrow microenvironment.
Clin Cancer Res
2004
;
10
:
7540
–6.
74
Iyer S, Wang Z-G, Akhtari M, Zhao W, Seth P. Targeting TGF-β signaling for cancer therapy.
Cancer Biol Ther
2005
;
4
:
261
–6.
75
Stoika R, Yakmovych M, Souchelnytskyi S, Yakymovych I. Potential role of transforming growth factor β1 in drug resistance of tumor cells.
Acta Biochim Pol
2003
;
50
:
497
–508.
76
Truong LD, Kadmon D, Mccune BK, Flanders KC, Scardino PT, Thompson TC. Association of transforming growth factor-β1 with prostate cancer: an immunohistochemical study.
Hum Pathol
1993
;
24
:
4
–9.
77
Takanami I, Imamura T, Hashizume T, Kikuchi K, Yamamoto Y, Kodaira S. Transforming growth factor β1 as a prognostic factor in pulmonary adenocarcinoma.
J Clin Pathol
1994
;
47
:
1098
–100.
78
Fischer JR, Darjes H, Lahm H, Schindel M, Drings P, Krammer PH. Constitutive secretion of bioactive transforming growth factor β1 by small cell lung cancer cell lines.
Eur J Cancer
1994
;
30a
:
2125
–9.
79
Damstrup L, Rygaard K, Spang-Thomsen M, Skovgaard Puolsen H. Expression of transforming growth factor β (TGF-β) receptors and expression of TGF-β1, TGF-β2 and TGF-β3 in human small cell lung cancer cell lines.
Br J Cancer
1993
;
67
:
1015
–21.
80
Friedman E, Gold Li, Klimstra D, Zeng ZS, Winawer S, Cohen A. High levels of transforming growth factor β1 correlate with disease progression in human colon cancer.
Cancer Epidemiol Biomarkers Prev
1995
;
4
:
549
–54.
81
Bristrow RE, Baldwin RL, Yamada SD, Korc M, Karlan BY. Altered expression of transforming growth factor-β ligands and receptors in primary and recurrent ovarian carcinoma.
Cancer
1999
;
85
:
658
–68.
82
Hazelbag S, Fleuren GJ, Baelde JJ, Schuuring E, Kenter GG, Gorter A. Cytokine profile of cervical cancer cells.
Gynecol Oncol
2001
;
83
:
235
–43.
83
Miyamoto H, Kubota Y, Shuin T, Torigoe S, Dobashi Y, Hosaka M. Expression of transforming growth factor-β1 in humans bladder cancer.
Cancer
1995
;
75
:
2565
–70.
84
Shariat SF, Kim JH, Andrews B, et al. Preoperative plasma levels of transforming growth factor β(1) strongly predict clinical outcome in patients with bladder carcinoma.
Cancer
2001
;
92
:
2985
–92.
85
Williams AO, Ward JM, Li JF, Jackson MA, Flanders KC. Immunohistochemical localization of transforming growth factor-β1 Kaposi's sarcoma.
Hum Pathol
1995
;
26
:
469
–73.
86
Sasaki A, Naganuma H, Satoh, et al. Secretion of transforming growth factor-β1 and β2 by malignant glioma cells.
Neurol Med Chir (Tokyo)
1995
;
35
:
423
–30.
87
Pasini FS, Brentani MM, Kowalski LP, Federico MH. Transforming growth factor β1, urokinase-type plasminogen activator and plasminogen activator activator inhibitor-1 mRNA expression in head and neck squamous carcinoma and normal adjacent mucosa.
Head Neck
2001
;
23
:
725
–32.
88
Matoba H, Sugano S, Yamaguchi N, Miyachi Y. Expression of transforming growth factor-β1 and transforming growth factor-β type II receptor mRNA in papillary thyroid carcinoma.
Horm Metab Res
1998
;
30
:
624
–8.
89
Yoshida K, Kuniyasu H, Yasui W, Kitadai Y, Toge T, Tahara E. Expression of growth factors and their receptors in human esophageal carcinomas: regulation of expression by epidermal growth factor and transforming growth factor α.
J Cancer Res Clin Oncol
1993
;
119
:
401
–7.
90
Liu P, Menon K, Alvarez E, Lu K, Teicher BA. Transforming growth factor-β and response to anticancer therapies in human liver and gastric tumors in vitro and in vivo.
Int J Oncol
2000
;
16
:
599
–610.
91
Matsuzaki K, Date M, Furukawa F, et al. Autocrine stimulatory mechanism by transforming growth factor β in human hepatocellular carcinoma.
Cancer Res
2000
;
60
:
1394
–402.
92
Tsushima H, Ito N, Tamura S, et al. Circulating transforming growth factor β1 as a predictor of liver metastasis after resection in colorectal cancer.
Clin Cancer Res
2001
;
7
:
1258
–62.