1
|
Evaluating the efficacy of multiple myeloma cell lines as models for patient tumors via transcriptomic correlation analysis. Leukemia 2020; 34:2754-2765. [PMID: 32123307 PMCID: PMC7483300 DOI: 10.1038/s41375-020-0785-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023]
Abstract
Multiple myeloma (MM) cell lines are routinely used to model the disease. However, a long-standing question is how well these cell lines truly represent tumor cells in patients. Here, we employ a recently described method of transcriptional correlation profiling to compare similarity of 66 MM cell lines to 779 newly diagnosed MM patient tumors. We found that individual MM lines differ significantly with respect to patient tumor representation, with median R ranging from 0.35 to 0.54. ANBL-6 was the “best” line, markedly exceeding all others (p < 2.2e−16). Notably, some widely used cell lines (RPMI-8226, U-266) scored poorly in our patient similarity ranking (48 and 52 of 66, respectively). Lines cultured with interleukin-6 showed significantly improved correlations with patient tumor (p = 9.5e−4). When common MM genomic features were matched between cell lines and patients, only t(4;14) and t(14;16) led to increased transcriptional correlation. To demonstrate the utility of our top-ranked line for preclinical studies, we showed that intravenously implanted ANBL-6 proliferates in hematopoietic organs in immunocompromised mice. Overall, our large-scale quantitative correlation analysis, utilizing emerging datasets, provides a resource informing the MM community of cell lines that may be most reliable for modeling patient disease while also elucidating biological differences between cell lines and tumors.
Collapse
|
2
|
Hawley TS, Linsley PS, Hawley RG. Co-expression of B7–1 with Interleukin-12 Enhances Vaccine-induced Antitumour Immunity in Experimental Myeloma. Hematology 2016; 3:365-74. [DOI: 10.1080/10245332.1998.11746410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Teresa S. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| | - Peter S. Linsley
- Bristol-Meyers Squibb Pharmaceutical Research Institute, Seattle, Washington, USA
| | - Robert G. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Oncolytic Virus and Anti–4-1BB Combination Therapy Elicits Strong Antitumor Immunity against Established Cancer. Cancer Res 2012; 72:1651-60. [DOI: 10.1158/0008-5472.can-11-2788] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
4
|
Kumar R, Sharma RK, Bansal DD, Patel DD, Mishra S, Miteva L, Dobreva Z, Gadjeva V, Stanilova S. Induction of immunostimulatory cytokine genes expression in human PBMCs by a novel semiquinone glucoside derivative (SQGD) isolated from a Bacillus sp. INM-1. Cell Immunol 2011; 267:67-75. [DOI: 10.1016/j.cellimm.2010.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 11/12/2010] [Accepted: 11/15/2010] [Indexed: 01/13/2023]
|
5
|
Wang H, Wei H, Zhang R, Hou S, Li B, Qian W, Zhang D, Kou G, Dai J, Guo Y. Genetically targeted T cells eradicate established breast cancer in syngeneic mice. Clin Cancer Res 2009; 15:943-50. [PMID: 19188165 DOI: 10.1158/1078-0432.ccr-08-2381] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of the present study was to evaluate the capacity and mechanisms of genetically modified erbB2-specific T cells to eradicate erbB2+ tumors in syngeneic mice. EXPERIMENTAL DESIGN Primary mouse T cells were modified to target the breast tumor-associated antigen erbB2 through retroviral-mediated transfer of a chimeric antigen receptor, termed single-chain antibody (scFv)-CD28-zeta. Antitumor efficacy of scFv-CD28-zeta-modified T cells was analyzed in mice bearing D2F2/E2 breast tumors. RESULTS The scFv-CD28-zeta-modified T cells were shown to specifically secrete T cytotoxic-1 cytokines and lyse erbB2+ breast tumor cells following receptor stimulation in vitro. Treatment with scFv-CD28-zeta-modified T cells was able to lead to long-term, tumor-free survival in mice bearing erbB2+ D2F2/E2 breast tumors. Importantly, the surviving mice developed a host memory response to D2F2/E2 tumor cells, and this host response was able to protect against a rechallenge with erbB2+ D2F2/E2 tumor cells and parental erbB2(-) D2F2 tumor cells. In addition, scFv-CD28-zeta T-cell expression of perforin and interferon-gamma were essential for complete antitumor efficacy. CONCLUSIONS Treatment with scFv-CD28-zeta-modified T cells was able to induce a host antitumor immunity in syngeneic mice. Complete tumor elimination by scFv-CD28-zeta-modified T cells required T cell-derived interferon-gamma and perforin, indicating that cytotoxicity and cytokine secretion play a role in the in vivo response.
Collapse
Affiliation(s)
- Hao Wang
- International Joint Cancer Institute and Changhai Hospital Cancer Center, The Second Military Medical University, Shanghai, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Raphael TJ, Kuttan G. Effect of naturally occurring triterpenoids ursolic acid and glycyrrhizic acid on the cell-mediated immune responses of metastatic tumor-bearing animals. Immunopharmacol Immunotoxicol 2008; 30:243-55. [PMID: 18569082 DOI: 10.1080/08923970701675044] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Effect of terpenoids, ursolic acid and glycyrrhizic acid on the cell-mediated immune response was studied in metastatic tumor-bearing C57BL/6 mice. Intraperitoneal administration of ursolic acid and glycyrrhizic acid (50 mumoles/Kg body wt for 5 consecutive days) was found to produce increased natural killer cell activity (NK-activity) in metastatic tumor-bearing animals. In the glycyrrhizic acid- and ursolic acid-treated groups the peak activity was observed on the 4(th) day (60.2% and 43% cell lysis respectively). Whereas in control animals the maximum cell lysis was obtained only on 16(th) day (25% cell lysis). Administration of terpenoids clearly enhanced the antibody dependent cell mediated cytotoxicity (ADCC). In tumor-bearing control animals maximum cell lysis was obtained only on the 16(th) day (20% cell lysis). But in the case of glycyrrhizic acid- and ursolic acid-treated groups, the maximum lysis was obtained on the 12(th) day. and it was 47% and 32.5% cell lysis, respectively. Intraperitoneal administration of these 2 terpenoids was also found to enhance antibody-dependent complement-mediated cytotoxicity (ACC) in metastatic tumor-bearing animals. The elevated level of GM-CSF in tumor-alone treated control animals (37.9 +/- 1.1 pg/ml) was reduced by the treatment with glycyrrhizic acid (20.3 pg/ml) and ursolic acid (22.5 pg/ml). The highly elevated level of IL-6 (370.1 pg/ml) in control animals was also reduced by the treatment of glycyrrhizic acid (313 pg/ml) and ursolic acid (299 pg/ml). The level of IL-2 was enhanced by the treatment with glycyrrhizic acid (37.9 pg/ml) and ursolic acid (35.9) compared to untreated tumor-bearing control animals (24.9 pg/ml).
Collapse
Affiliation(s)
- T J Raphael
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala, India
| | | |
Collapse
|
7
|
Numasaki M, Tagawa M, Iwata F, Suzuki T, Nakamura A, Okada M, Iwakura Y, Aiba S, Yamaya M. IL-28 elicits antitumor responses against murine fibrosarcoma. THE JOURNAL OF IMMUNOLOGY 2007; 178:5086-98. [PMID: 17404291 DOI: 10.4049/jimmunol.178.8.5086] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-28 is a recently described antiviral cytokine. In this study, we investigated the biological effects of IL-28 on tumor growth to evaluate its antitumor activity. IL-28 or retroviral transduction of the IL-28 gene into MCA205 cells did not affect in vitro growth, whereas in vivo growth of MCA205IL-28 was markedly suppressed along with survival advantages when compared with that of controls. When the metastatic ability of IL-28-secreting MCA205 cells was compared with that of controls, the expression of IL-28 resulted in a potent inhibition of metastases formation in the lungs. IL-28-mediated suppression of tumor growth was mostly abolished in irradiated mice, indicating that irradiation-sensitive cells, presumably immune cells, are primarily involved in the IL-28-induced suppression of tumor growth. In vivo cell depletion experiments displayed that polymorphonuclear neutrophils, NK cells, and CD8 T cells, but not CD4 T cells, play an equal role in the IL-28-mediated inhibition of in vivo tumor growth. Consistent with these findings, inoculation of MCA205IL-28 into mice evoked enhanced IFN-gamma production and cytotoxic T cell activity in spleen cells. Antitumor action of IL-28 is partially dependent on IFN-gamma and is independent of IL-12, IL-17, and IL-23. IL-28 increased the total number of splenic NK cells in SCID mice and enhanced IL-12-induced IFN-gamma production in vivo and expanded spleen cells in C57BL/6 mice. Moreover, IL-12 augmented IL-28-mediated antitumor activity in the presence or absence of IFN-gamma. These findings indicate that IL-28 has bioactivities that induce innate and adaptive immune responses against tumors.
Collapse
Affiliation(s)
- Muneo Numasaki
- Department of Geriatric and Respiratory Medicine, Tohoku University School of Medicine, Sendai, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Pappa C, Miyakis S, Tsirakis G, Sfiridaki A, Alegakis A, Kafousi M, Stathopoulos EN, Alexandrakis MG. Serum levels of interleukin-15 and interleukin-10 and their correlation with proliferating cell nuclear antigen in multiple myeloma. Cytokine 2007; 37:171-5. [PMID: 17446083 DOI: 10.1016/j.cyto.2007.02.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2006] [Revised: 12/04/2006] [Accepted: 02/19/2007] [Indexed: 10/23/2022]
Abstract
In order to determine prognostic factors characterizing multiple myeloma (MM) cell kinetics, bone marrow proliferative activity and serum Interleukin-10 (IL-10), and Interleukin-15 (IL-15) levels were measured in 40 newly diagnosed MM patients, compared with 10-age and sex-matched-healthy controls. Cell proliferation was evaluated by employing a monoclonal antibody directed against the proliferating cell nuclear antigen (PCNA), whereas IL-10 and IL-15 were measured with quantitative sandwich enzyme immunoassay methods. IL-15, IL-10 and PCNA were higher in the patient group than in controls (P<0.001). IL-10 levels, and PCNA increased significantly with increasing Durie-Salmon disease stage (I-III, P<0.002, and P=0.001, respectively). Serum IL-15 levels in MM stage III patients were elevated in comparison with stages I and II, the difference however, did not reach statistical significance. There was a significant positive correlation between serum IL-15 and IL-10 levels (r: 0.372, P<0.01), and between serum IL-10 and PCNA (r: 0.608, P<0.0001), as well as a positive correlation of serum IL-15 with PCNA, which marginally failed to reach statistical significance. Serum IL-15 levels are elevated in MM patients, increase with advancing stage, and correlate with Il-10 and PCNA. These proliferative factors may be useful in assessing disease progression in MM.
Collapse
Affiliation(s)
- C Pappa
- Department of Haematology, School of Medicine, University of Crete, Heraklion, Greece
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Mitra-Kaushik S, Harding J, Hess J, Schreiber R, Ratner L. Enhanced tumorigenesis in HTLV-1 tax-transgenic mice deficient in interferon-gamma. Blood 2004; 104:3305-11. [PMID: 15292059 DOI: 10.1182/blood-2004-01-0266] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The oncoprotein Tax of human T-cell leukemia virus type I (HTLV-1) is the major mediator of viral pathogenesis in infected individuals. Expression of Tax under the regulation of the human granzyme B promoter in mice results in a lymphoproliferative disorder resembling adult T-cell leukemia/lymphoma (ATL). Tax expression is associated with the production of high levels interferon-gamma (IFN-gamma) in HTLV-1-infected CD4(+) cells and Tax-transgenic tumors. We examined the role of IFN-gamma in tumorigenesis, by mating Tax-transgenic mice with a gene-specific knockout for IFN-gamma. IFN-gamma(-/-) Tax(+)-transgenic mice show accelerated tumor onset (median, 4 versus 6 months), dissemination (median, 5 versus 7 months), and death (median, 7 versus 10 months), compared with IFN-gamma(+/-) or IFN-gamma(+/+) Tax(+) mice. Pathologic and immunophenotypic characteristics of tumors from all genotypes are indistinguishable, except for enhanced interleukin 2 receptor-beta (IL-2Rbeta) and suppressed intercellular adhesion molecule-1 (ICAM-1) expression on tumors from IFN-gamma(-/-) Tax(+) transgenic mice. IFN-gamma(-/-) tumors demonstrate enhanced CD31 (platelet-endothelial CAM-1 [PECAM-1]) staining compared with those from IFN-gamma(+/-) or IFN-gamma(+/+) Tax(+) mice. Angiogenesis-specific cDNA microarray analysis identified 4 mediators of angiogenic growth differentially expressed in tumors from Tax(+)IFN-gamma(-/-) mice compared with Tax(+)IFN-gamma(+/+) littermates. As confirmed by reverse transcription-polymerase chain reaction (RT-PCR), loss of IFN-gamma results in down-regulation of tumor necrosis factor-alpha (TNF-alpha) and tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) while up-regulating expression of vascular endothelial growth factor (VEGF) and tenascin C. These results provide insight into a possible mechanism by which IFN-gamma contributes to host resistance against HTLV-induced tumors through an angiostatic effect.
Collapse
Affiliation(s)
- Shibani Mitra-Kaushik
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Ave, Box 8069, St Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
10
|
Ikezoe T, Yang Y, Heber D, Taguchi H, Koeffler HP. PC-SPES: a potent inhibitor of nuclear factor-kappa B rescues mice from lipopolysaccharide-induced septic shock. Mol Pharmacol 2003; 64:1521-9. [PMID: 14645683 DOI: 10.1124/mol.64.6.1521] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Septic shock is the most common cause of death in intensive care units, and no effective treatment is available at present. Lipopolysaccharide (LPS) is the primary mediator of Gram-negative sepsis by inducing the production of macrophage-derived proinflammatory cytokines, in which activation of nuclear factor-kappaB (NF-kappaB) plays an important role. PC-SPES is an eight-herb mixture active against a variety of malignancies, including prostate cancer and leukemia. In this study, we demonstrated that PC-SPES inhibited the LPS-induced NF-kappaB reporter activity in RAW264.7 macrophages. Electrophoretic mobility shift assay showed that PC-SPES inhibited the binding of NF-kappaB to specific DNA sequences; however, it did not affect either degradation of inhibitory kappaBalpha or nuclear translocation of NF-kappaB. Also, we explored the effect of PCSPES on LPS-induced mitogen-activated protein (MAP) kinase signaling; PC-SPES did not affect LPS-induced phosphorylation of MAP kinases, including c-Jun NH2-terminal kinase, p38, and extracellular signal-regulated kinase 1/2. Moreover, PC-SPES decreased the production of proinflammatory cytokines and inducible enzymes, such as tumor necrosis factor (TNF) alpha, interleukin (IL)-1beta, IL-6, cyclooxygenase-2, as well as inducible nitric-oxide synthase in RAW264.7 macrophages and peritoneal macrophages from C57BL/6 mice after the cells were stimulated by either LPS or LPS and interferon-gamma. Furthermore, PC-SPES rescued C57BL/6 mice from death caused by LPS-induced septic shock in conjunction with decreased serum levels of TNFalpha and IL-1beta. Together, PC-SPES is a potent inhibitor of NF-kappaB and might be useful for the treatment of sepsis and inflammatory diseases.
Collapse
Affiliation(s)
- Takayuki Ikezoe
- Department of Internal Medicine, Kochi Medical School, Nankoku, Kochi 783-8505, Japan.
| | | | | | | | | |
Collapse
|
11
|
Ma HL, Whitters MJ, Konz RF, Senices M, Young DA, Grusby MJ, Collins M, Dunussi-Joannopoulos K. IL-21 activates both innate and adaptive immunity to generate potent antitumor responses that require perforin but are independent of IFN-gamma. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:608-15. [PMID: 12847225 DOI: 10.4049/jimmunol.171.2.608] [Citation(s) in RCA: 144] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
IL-21 is a key factor in the transition between innate and adaptive immune responses. We have used the cytokine gene therapy approach to study the antitumor responses mediated by IL-21 in the B16F1 melanoma and MethA fibrosarcoma tumor models in mice. Retrovirally transduced tumor cells secreting biologically functional IL-21 have growth patterns in vitro similar to that of control green fluorescent protein-transduced cells, but are completely rejected in vivo. We show that IL-21 activates NK and CD8(+) T cells in vivo, thus mediating complete rejection of poorly immunogenic tumors. Rejection of IL-21-secreting tumors requires the presence of cognate IL-21R and does not depend on CD4(+) T cell help. Interestingly, perforin, but not IFN-gamma or other major Th1 and Th2 cytokines (IL-12, IL-4, or IL-10), is required for the IL-21-mediated antitumor response. Moreover, IL-21 results in 50% protection and 70% cure of nonimmunogenic tumors when given before and after tumor challenge, respectively, in C57BL/6 mice. We conclude that IL-21 immunotherapy warrants clinical evaluation as a potential treatment for cancer.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antigens, Neoplasm/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/metabolism
- Cell Division/genetics
- Cell Division/immunology
- Cytotoxicity, Immunologic/genetics
- Female
- Growth Inhibitors/genetics
- Growth Inhibitors/metabolism
- Growth Inhibitors/physiology
- Immunity, Active/genetics
- Immunity, Innate/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Interleukin-10/physiology
- Interleukin-12/physiology
- Interleukin-21 Receptor alpha Subunit
- Interleukin-4/physiology
- Interleukins/administration & dosage
- Interleukins/genetics
- Interleukins/metabolism
- Interleukins/physiology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/therapy
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Nude
- Mice, SCID
- Perforin
- Pore Forming Cytotoxic Proteins
- Receptors, Interleukin/deficiency
- Receptors, Interleukin/genetics
- Receptors, Interleukin/physiology
- Receptors, Interleukin-21
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/metabolism
- Sarcoma, Experimental/therapy
- T-Lymphocytes, Cytotoxic/immunology
- Transduction, Genetic
- Tumor Cells, Cultured
Collapse
|
12
|
Haynes NM, Trapani JA, Teng MWL, Jackson JT, Cerruti L, Jane SM, Kershaw MH, Smyth MJ, Darcy PK. Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5780-6. [PMID: 12421958 DOI: 10.4049/jimmunol.169.10.5780] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A new strategy to improve the therapeutic utility of redirected T cells for cancer involves the development of novel Ag-specific chimeric receptors capable of stimulating optimal and sustained T cell antitumor activity in vivo. Given that T cells require both primary and costimulatory signals for optimal activation and that many tumors do not express critical costimulatory ligands, modified single-chain Ab receptors have been engineered to codeliver CD28 costimulation. In this study, we have compared the antitumor potency of primary T lymphocytes expressing carcinoembryonic Ag (CEA)-reactive chimeric receptors that incorporate either TCR-zeta or CD28/TCR-zeta signaling. Although both receptor-transduced T cell effector populations demonstrated cytolysis of CEA(+) tumors in vitro, T cells expressing the single-chain variable fragment of Ig (scFv)-CD28-zeta chimera had a far greater capacity to control the growth of CEA(+) xenogeneic and syngeneic colon carcinomas in vivo. The observed enhanced antitumor activity of T cells expressing the scFv-CD28-zeta receptor was critically dependent on perforin and the production of IFN-gamma. Overall, this study has illustrated the ability of a chimeric scFv receptor capable of harnessing the signaling machinery of both TCR-zeta and CD28 to augment T cell immunity against tumors that have lost expression of both MHC/peptide and costimulatory ligands in vivo.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Adenocarcinoma/prevention & control
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/genetics
- Animals
- Binding Sites, Antibody/genetics
- CD28 Antigens/administration & dosage
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Carcinoembryonic Antigen/administration & dosage
- Carcinoembryonic Antigen/biosynthesis
- Carcinoembryonic Antigen/genetics
- Carcinoembryonic Antigen/immunology
- Cells, Cultured
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/metabolism
- Graft Rejection/genetics
- Graft Rejection/immunology
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/genetics
- Humans
- Immunoglobulin Variable Region/administration & dosage
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Immunotherapy, Adoptive/methods
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Perforin
- Pore Forming Cytotoxic Proteins
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Immunologic/administration & dosage
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
- Transplantation, Isogeneic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Nicole M Haynes
- Cancer Immunology Program, Sir Donald and Lady Trescowthick Laboratories, Peter MacCallum Cancer Institute, St. Andrew's Place, East Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Song L, Li Y, Shen B. Protein kinase ERK contributes to differential responsiveness of human myeloma cell lines to IFNalpha. Cancer Cell Int 2002; 2:9. [PMID: 12234375 PMCID: PMC140137 DOI: 10.1186/1475-2867-2-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2002] [Accepted: 07/08/2002] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND: Despite IFNalpha has been used extensively in the treatment of multiple myeloma (MM), there are also several reports suggesting that IFNalpha may aggravate isease in some MM patients. That means the effect of IFNalpha on the growth of myeloma cells in vivo may be different. In this study, we selected two human myeloma cell lines that vary remarkably in response to IFNalpha and focused on elucidating the mechanism of differential IFNalpha responsiveness. RESULTS: Sko-007 is a myeloma cell line whose growth is arrested by IFNalpha; however, IFNalpha promoted the proliferation of the other myeloma cell line U266. We observed that the growth-stimulation effect of IFNalpha on U266 cells did not result from up-regulation of the IL-6 receptors on cell surface; while IFNalpha treatment on Sko-007 cells significantly reduced gp130 expression. Moreover, the transcription factors STAT3 and STAT1, which are involved in the JAK/STAT signal transduction pathway, can be activated in both IFNalpha-stimulated and -inhibited myeloma cell lines; while the activation of the protein kinase ERK, which is involved in the Ras/MAPK signal transduction pathway, can be down-regulated in IFNalpha-arrested Sko-007 cells and up-regulated in IFNalpha-stimulated U266 cells. In addition, both IFNalpha-induced growth-stimulation effect and the up-regulated activation of ERK in U266 cells were efficiently inhibited by PD98059, the specific inhibitor of MAPK/ERK kinase (MEK). CONCLUSION: Myeloma cells responsiveness to IFNalpha is heterogeneous and the activation state of ERK in the Ras/MAPK signalling pathway mainly contributed to this difference.
Collapse
Affiliation(s)
- Lun Song
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Taiping Road, 27, Beijing, 100850, P. R. China
| | - Yan Li
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Taiping Road, 27, Beijing, 100850, P. R. China
| | - Beifen Shen
- Department of Molecular Immunology, Institute of Basic Medical Sciences, Taiping Road, 27, Beijing, 100850, P. R. China
| |
Collapse
|
14
|
Cheung WC, Van Ness B. Distinct IL-6 signal transduction leads to growth arrest and death in B cells or growth promotion and cell survival in myeloma cells. Leukemia 2002; 16:1182-8. [PMID: 12040451 DOI: 10.1038/sj.leu.2402481] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2001] [Accepted: 12/05/2001] [Indexed: 11/09/2022]
Abstract
In B cell development, interleukin-6 (IL-6) induces terminal maturation of B lymphocytes into antibody producing plasma cells. Terminal differentiated B cells cell cycle arrest and death follows. In contrast, IL-6 acts as a growth factor for malignant myeloma plasma cells and in some cases protects them from therapeutic treatment. In this study, we examined two cell lines that show different responses to IL-6. Lymphoblastoid CESS cells respond to IL-6 by terminally differentiating into antibody producing plasma cells, cell cycle arrest, and undergo cell death. Continuous addition of IL-6 to these cells induces transient activation of STAT3, SHP-2 phosphorylation, and does not alter bcl-X(L) and c-myc expression. In contrast, the myeloma line ANBL6 proliferates when stimulated with IL-6 and this correlates with prolonged STAT3 activation and up-regulation of bcl-X(L) and c-myc. Interestingly, gp130-associated SHP-2 phosphorylation was detected in the IL-6-induced CESS cells but not myeloma cell lines. The data show a very distinct IL-6 signal transduction and kinetics in these cell lines and the distinct molecular events correlate closely to the cell fate of the lymphoblast and myeloma cell lines.
Collapse
Affiliation(s)
- W-C Cheung
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | |
Collapse
|
15
|
Dredge K, Marriott JB, Todryk SM, Muller GW, Chen R, Stirling DI, Dalgleish AG. Protective antitumor immunity induced by a costimulatory thalidomide analog in conjunction with whole tumor cell vaccination is mediated by increased Th1-type immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4914-9. [PMID: 11994441 DOI: 10.4049/jimmunol.168.10.4914] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thalidomide and its novel T cell costimulatory analogs (immunomodulatory drugs) are currently being assessed in the treatment of patients with advanced cancer. However, neither tumor-specific T cell costimulation nor effective antitumor activity has been demonstrated in vivo. In this study, we assessed the ability of an immunomodulatory drug (CC-4047/ACTIMID) to prime a tumor-specific immune response following tumor cell vaccination. We found that the presence of CC-4047 during the priming phase strongly enhanced antitumor immunity in the vaccinated group, and this correlated with protection from subsequent live tumor challenge. Protection was associated with tumor-specific production of IFN-gamma and was still observed following a second challenge with live tumor cells 60 days later. Furthermore, CD8(+) and CD4(+) splenocyte fractions from treated groups secreted increased IFN-gamma and IL-2 in response to tumor cells in vitro. Coculture of naive splenocytes with anti-CD3 mAb in the presence of CC-4047 directly costimulated T cells and increased Th1-type cytokines. Our results are the first to demonstrate that a costimulatory thalidomide analog can prime protective, long-lasting, tumor-specific, Th1-type responses in vivo and further support their ongoing clinical development as novel anti-cancer agents.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antigens, CD/biosynthesis
- Antineoplastic Combined Chemotherapy Protocols/immunology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- B7-1 Antigen/biosynthesis
- B7-2 Antigen
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cells, Cultured
- Coculture Techniques
- Cytokines/biosynthesis
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/immunology
- Histocompatibility Antigens Class I/biosynthesis
- Injections, Intraperitoneal
- Injections, Subcutaneous
- Interferon-gamma/biosynthesis
- Melanoma, Experimental
- Membrane Glycoproteins/biosynthesis
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasm Transplantation/immunology
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocyte Subsets/immunology
- Th1 Cells/immunology
- Thalidomide/administration & dosage
- Thalidomide/analogs & derivatives
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- Keith Dredge
- Division of Oncology, Department of Oncology, Gastroenterology, Endocrinology, and Metabolism, St. George's Hospital Medical School, Cranmer Terrace, Tooting, London, United Kingdom SW17 0RE.
| | | | | | | | | | | | | |
Collapse
|
16
|
Takeda K, Smyth MJ, Cretney E, Hayakawa Y, Kayagaki N, Yagita H, Okumura K. Critical role for tumor necrosis factor-related apoptosis-inducing ligand in immune surveillance against tumor development. J Exp Med 2002; 195:161-9. [PMID: 11805143 PMCID: PMC2193611 DOI: 10.1084/jem.20011171] [Citation(s) in RCA: 321] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Natural killer (NK) cells and interferon (IFN)-gamma have been implicated in immune surveillance against tumor development. Here we show that tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plays a critical role in the NK cell-mediated and IFN-gamma-dependent tumor surveillance. Administration of neutralizing monoclonal antibody against TRAIL promoted tumor development in mice subcutaneously inoculated with a chemical carcinogen methylcholanthrene (MCA). This protective effect of TRAIL was at least partly mediated by NK cells and totally dependent on IFN-gamma. In the absence of TRAIL, NK cells, or IFN-gamma, TRAIL-sensitive sarcomas preferentially emerged in MCA-inoculated mice. Moreover, development of spontaneous tumors in p53(+/-) mice was also promoted by neutralization of TRAIL. These results indicated a substantial role of TRAIL as an effector molecule that eliminates developing tumors.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Apoptosis/genetics
- Apoptosis/immunology
- Apoptosis Regulatory Proteins
- Cytotoxicity, Immunologic/genetics
- Cytotoxicity, Immunologic/immunology
- Interferon-gamma/genetics
- Interferon-gamma/immunology
- Killer Cells, Natural/immunology
- Male
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/prevention & control
- TNF-Related Apoptosis-Inducing Ligand
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/immunology
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/immunology
Collapse
Affiliation(s)
- Kazuyoshi Takeda
- Department of Immunology, Juntendo University, School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | | | | | | | | | | | | |
Collapse
|
17
|
Gaemers IC, Vos HL, Volders HH, van der Valk SW, Hilkens J. A stat-responsive element in the promoter of the episialin/MUC1 gene is involved in its overexpression in carcinoma cells. J Biol Chem 2001; 276:6191-9. [PMID: 11084045 DOI: 10.1074/jbc.m009449200] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The mucin-like glycoprotein episialin (MUC1) is highly overproduced by a number of human carcinomas. We have shown previously in a variety of mammalian cell lines that overexpression of this very large transmembrane molecule diminishes cellular adhesion, suggesting that episialin/MUC1 overexpression may play an important role in tumor invasion and metastasis. By using in situ hybridization, we show here that episialin/MUC1 mRNA expression can be increased more than 10-fold in breast carcinoma cells relative to the expression in adjacent normal breast epithelium. In search of the molecular mechanism of this overexpression, we observed that the episialin/MUC1 promoter contains a candidate binding site for transcription factors of the STAT family approximately 500 base pairs upstream of the transcription start site. Cytokines and/or growth factors such as interleukin-6 or interferon-gamma can activate STATs. In the human breast carcinoma cell line T47D, both compounds are able to stimulate transcription of a luciferase reporter gene under the control of a 750-base pair MUC1 promoter fragment proximal to the transcription start site. The observed increase is entirely mediated by the single STAT-binding site, since mutation of this site abolishes stimulation of the reporter by interleukin-6 and interferon-gamma. In addition, mutation of the STAT site also decreased the promoter activity in nonstimulated T47D cells, suggesting that the STAT-binding site is among the elements that are involved in the overexpression of MUC1 in tumor cells.
Collapse
Affiliation(s)
- I C Gaemers
- Division of Tumor Biology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
18
|
Cheung WC, Van Ness B. The bone marrow stromal microenvironment influences myeloma therapeutic response in vitro. Leukemia 2001; 15:264-71. [PMID: 11236942 DOI: 10.1038/sj.leu.2402022] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The bone marrow microenvironment supports growth and differentiation of normal hematopoietic cells and can contribute to malignant growth. Since myeloma cells localize and accumulate in bone marrow, it is important to understand the influence of the bone marrow microenvironment not only on the growth of the malignant cells, but also on the therapeutic response of myeloma cells. Growth factors such as interleukin-6 (IL-6) produced by bone marrow stromal cells can protect myeloma cells from glucocorticoid-induced apoptosis. We examined the effect of myeloma cells-bone marrow stromal cells interaction in vitro on several therapeutic treatments. An interleukin-6-dependent myeloma cell line ANBL6 was used and treated with dexamethasone, doxorubicin, and melphalan in the presence of bone marrow stromal cells. Stromal cells were able to protect ANBL6 from dexamethasone, but significantly enhanced the effect of doxorubicin and melphalan. IL-6-induced bcl-XL and cyclin D2 expression in ANBL6 cells, but dexamethasone was able to suppress both bcl-XL and cyclin D2 expression in ANBL6. Doxorubicin and melphalan were able to suppress bcl-XL expression only in the presence of IL-6. We also looked at the effect of activating mutations of N-ras in myeloma cells interacting with stromal cells on therapeutic responses. Surprisingly, ANBL6 N-ras shows significant resistance to all drugs used. Notably, the presence of stromal cells did not alter ANBL6 Nras cells' drug resistance. These results suggest both the bone marrow microenvironment and genetic alterations of myeloma cells can independently impact on therapeutic responses.
Collapse
Affiliation(s)
- W C Cheung
- Graduate Program in Biochemistry, Molecular Biology, and Biophysics, The University of Minnesota, Minneapolis 55455, USA
| | | |
Collapse
|
19
|
Hjertner O, Hjorth-Hansen H, Börset M, Seidel C, Waage A, Sundan A. Bone morphogenetic protein-4 inhibits proliferation and induces apoptosis of multiple myeloma cells. Blood 2001; 97:516-22. [PMID: 11154231 DOI: 10.1182/blood.v97.2.516] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) can be isolated from organic bone matrix and are able to initiate de novo cartilage and bone formation. Here it is shown that BMP-4 inhibited DNA synthesis in a dose-dependent manner in 3 IL-6-dependent multiple myeloma (MM) cell lines (OH-2, IH-1, and ANBL-6). In contrast, no effect on DNA synthesis was observed in 3 IL-6-independent MM cell lines (JJN-3, U266, and RPMI 8226). BMP-4 induced cell cycle growth arrest in the G(0)/G(1) phase in OH-2 and ANBL-6 cells but not in IH-1 cells. BMP-4 induced apoptosis in OH-2 and IH-1 cells, but not significantly in ANBL-6 cells. Furthermore, BMP-4 induced apoptosis in freshly isolated MM cells from 4 of 13 patients. In the OH-2 and ANBL-6 cell lines and in a patient sample, immunoblotting showed that BMP-4 down-regulated IL-6-induced tyrosine phosphorylation of Stat3, suggesting a mechanism for the apparent antagonism between IL-6 and BMP-4. BMP-4 or analogues may be attractive therapeutic agents in MM because of possible beneficial effects on both tumor burden and bone disease.
Collapse
Affiliation(s)
- O Hjertner
- Institute of Cancer Research and Molecular Biology, and the Section of Hematology, Institute of Environmental Medicine, Norwegian University of Science and Technology, Trondheim, Norway.
| | | | | | | | | | | |
Collapse
|
20
|
Street SE, Cretney E, Smyth MJ. Perforin and interferon-gamma activities independently control tumor initiation, growth, and metastasis. Blood 2001; 97:192-7. [PMID: 11133760 DOI: 10.1182/blood.v97.1.192] [Citation(s) in RCA: 385] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Perforin (pfp) and interferon-gamma (IFN-gamma) together in C57BL/6 (B6) and BALB/c mouse strains provided optimal protection in 3 separate tumor models controlled by innate immunity. Using experimental (B6, RM-1 prostate carcinoma) and spontaneous (BALB/c, DA3 mammary carcinoma) models of metastatic cancer, mice deficient in both pfp and IFN-gamma were significantly less proficient than pfp- or IFN-gamma-deficient mice in preventing metastasis of tumor cells to the lung. Pfp and IFN-gamma-deficient mice were as susceptible as mice depleted of natural killer (NK) cells in both tumor metastasis models, and IFN-gamma appeared to play an early role in protection from metastasis. Previous experiments in a model of fibrosarcoma induced by the chemical carcinogen methylcholanthrene indicated an important role for NK1.1(+) T cells. Herein, both pfp and IFN-gamma played critical and independent roles in providing the host with protection equivalent to that mediated by NK1.1(+) T cells. Further analysis demonstrated that IFN-gamma, but not pfp, controlled the growth rate of sarcomas arising in these mice. Thus, this is the first study to demonstrate that host IFN-gamma and direct cytotoxicity mediated by cytotoxic lymphocytes expressing pfp independently contribute antitumor effector functions that together control the initiation, growth, and spread of tumors in mice.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Cell Division/drug effects
- Cell Transformation, Neoplastic/drug effects
- Cytotoxicity Tests, Immunologic
- Disease Models, Animal
- Fibrosarcoma/chemically induced
- Fibrosarcoma/drug therapy
- Fibrosarcoma/immunology
- Interferon-gamma/genetics
- Interferon-gamma/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Leukocyte Count
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/pharmacology
- Methylcholanthrene/pharmacology
- Mice
- Mice, Inbred Strains
- Mice, Knockout
- Mice, SCID
- Neoplasm Metastasis/drug therapy
- Neoplasm Metastasis/prevention & control
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Perforin
- Pore Forming Cytotoxic Proteins
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- S E Street
- Cancer Immunology, Peter MacCallum Cancer Institute, Victoria, Australia
| | | | | |
Collapse
|
21
|
Otsuki T, Yamada O, Yata K, Sakaguchi H, Kurebayashi J, Yawata Y, Ueki A. Expression and production of interleukin 10 in human myeloma cell lines. Br J Haematol 2000. [DOI: 10.1111/j.1365-2141.2000.02413.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Otsuki T, Yamada O, Yata K, Sakaguchi H, Kurebayashi J, Yawata Y, Ueki A. Expression and production of interleukin 10 in human myeloma cell lines. Br J Haematol 2000. [DOI: 10.1046/j.1365-2141.2000.02413.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Kastrinakis NG, Gorgoulis VG, Foukas PG, Dimopoulos MA, Kittas C. Molecular aspects of multiple myeloma. Ann Oncol 2000; 11:1217-28. [PMID: 11106109 DOI: 10.1023/a:1008331714186] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Multiple myeloma (MM) is a B-cell neoplasm characterized by bone marrow infiltration with malignant plasma cells, which synthesize and secrete monoclonal immunoglobulin (Ig) fragments. Despite the considerable progress in the understanding of MM biology, the molecular basis of the disease remains elusive. The initial transformation is thought to occur in a postgerminal center B-lineage cell, carrying a somatically hypermutated Ig heavy chain (IGH) gene. This plasmablastic precursor cell colonizes the bone marrow, propagates clonally and differentiates into a slowly proliferating myeloma cell population, all under the influence of specific cell adhesion molecules and cytokines. Production of interleukin-6 by stromal cells, osteoblasts and, in some cases, neoplastic cells is an essential element of myeloma cell growth, with the cytokine stimulus being delivered intracellularly via the Jack-STAT and ras signaling pathways. While karyotypic changes have been identified in up to 50% of MM patients, recent molecular cytogenetic techniques have revealed chromosomal abnormalities in the vast majority of examined cases. Translocations mostly involve illegal switch rearrangements of the IGH locus with various partner genes (CCND1, FGFR3, c-maf). Such events have been assigned a critical role in MM development. Mutations in coding and regulatory regions, as well as aberrant expression patterns of several oncogenes (c-myc, ras) and tumor suppressor genes (p16, p15) have been reported. Key regulators of programmed cell death (BCL-2, Fas), tumor expansion (metalloproteinases) and drug responsiveness (topoisomerase II alpha) have also been implicated in the pathogenesis of this hematologic malignancy. A tumorigenic role for human herpesvirus 8 (HHV8) was postulated recently, following the detection of viral sequences in bone marrow dendritic cells of MM patients. However, since several research groups were unable to confirm this observation, the role of HHV8 remains unclear. Translation of the advances in MM molecular biology into novel therapeutic strategies is essential in order to improve disease prognosis.
Collapse
Affiliation(s)
- N G Kastrinakis
- Department of Histology, School of Medicine, University of Athens, Greece
| | | | | | | | | |
Collapse
|