Genetically modified tumour vaccines: an obstacle race to break host tolerance to cancer

The development of genetically modified tumour vaccines (GMTV) has been prompted by a better understanding of antitumour immune responses and genetic engineering technologies, as well as the identification of numerous tumour antigens (TA) in several malignancies which occasionally induce spontaneous tumour regressions. Cellular vaccines are based on autologous or allogeneic tumour cells genetically engineered to secrete different cytokines, co-stimulatory molecules, or allogeneic HLA molecules in order to provide a strong stimulatory signal together with the presented TA. Another promising approach that is targeted towards breaking immune tolerance to TA, exploits dendritic cells (DC) loaded or genetically modified with TA (and sometimes cytokines). Effective nonviral and viral gene delivery systems have been constructed including a third generation of adenoviral, lentiviral and hybrid vectors. Studies in mice demonstrated that therapeutic, curative immune responses might be elicited by GMTV. Promising results from animal studies are rarely seen in human trials. Several reasons, such as numerous escape mechanisms of slowly evolving spontaneous tumours and immune incompetence of advanced patients, are major concerns. Improved monitoring of immune responses to GMTV is essential to distinguish between responders and non-responders in order to tailor immune therapy strategy to the individual patient.

[1]  Mark M. Davis,et al.  Melanocyte Destruction after Antigen-Specific Immunotherapy of Melanoma , 2000, The Journal of experimental medicine.

[2]  C. Balagué,et al.  Replicative adenoviruses for cancer therapy , 2000, Nature Biotechnology.

[3]  P. Schrier,et al.  Vaccination of melanoma patients with an allogeneic, genetically modified interleukin 2-producing melanoma cell line. , 2000, Human gene therapy.

[4]  J. Leonard,et al.  Vaccines with interleukin-12-transduced acute myeloid leukemia cells elicit very potent therapeutic and long-lasting protective immunity. , 1999, Blood.

[5]  A. Ohta,et al.  Distinct Role of Antigen-Specific T Helper Type 1 (Th1) and Th2 Cells in Tumor Eradication in Vivo , 1999, The Journal of experimental medicine.

[6]  D. Pardoll,et al.  Inducing autoimmune disease to treat cancer. , 1999, Proceedings of the National Academy of Sciences of the United States of America.

[7]  P. Ricciardi-Castagnoli,et al.  Dendritic cells directly trigger NK cell functions: Cross-talk relevant in innate anti-tumor immune responses in vivo , 1999, Nature Medicine.

[8]  C. Lowenstein,et al.  The Central Role of CD4+ T Cells in the Antitumor Immune Response , 1998, The Journal of experimental medicine.

[9]  B. Chain,et al.  In vivo priming of T cells against cryptic determinants by dendritic cells exposed to interleukin 6 and native antigen. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[10]  D. Morton,et al.  Anti-tyrosinase-related protein-2 immune response in vitiligo patients and melanoma patients receiving active-specific immunotherapy. , 1998, The Journal of investigative dermatology.

[11]  D. Neuberg,et al.  Vaccination with irradiated autologous melanoma cells engineered to secrete human granulocyte-macrophage colony-stimulating factor generates potent antitumor immunity in patients with metastatic melanoma. , 1998, Proceedings of the National Academy of Sciences of the United States of America.

[12]  R. Gupta,et al.  Correlation of specific immune responses with survival in melanoma patients with distant metastases receiving polyvalent melanoma cell vaccine. , 1998, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[13]  C. Figdor,et al.  Killer cell inhibitory receptors for MHC class I molecules regulate lysis of melanoma cells mediated by NK cells, gamma delta T cells, and antigen-specific CTL. , 1998, Journal of immunology.

[14]  R. Steinman,et al.  Dendritic cells and the control of immunity , 1998, Nature.

[15]  Dirk Schadendorf,et al.  Vaccination of melanoma patients with peptide- or tumorlysate-pulsed dendritic cells , 1998, Nature Medicine.

[16]  K. Foon,et al.  Induction of IgG antibodies by an anti-idiotype antibody mimicking disialoganglioside GD2. , 1998, Journal of immunotherapy.

[17]  G. Dranoff,et al.  Gene immunotherapy in murine acute myeloid leukemia: granulocyte-macrophage colony-stimulating factor tumor cell vaccines elicit more potent antitumor immunity compared with B7 family and other cytokine vaccines. , 1998, Blood.

[18]  D. Curiel,et al.  Stable in vivo gene transduction via a novel adenoviral/retroviral chimeric vector , 1997, Nature Biotechnology.

[19]  Antonio Lanzavecchia,et al.  Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells , 1997, Nature.

[20]  P. Walker,et al.  Role of Fas ligand (CD95L) in immune escape: the tumor cell strikes back. , 1997, Journal of immunology.

[21]  D. Kufe,et al.  Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells , 1997, Nature Medicine.

[22]  H. Seigler,et al.  Generation of primary tumor-specific cytotoxic T lymphocytes from autologous and human lymphocyte antigen class I-matched allogeneic peripheral blood lymphocytes by B7 gene-modified melanoma cells. , 1997, Cancer research.

[23]  F. Sallusto,et al.  Origin, maturation and antigen presenting function of dendritic cells. , 1997, Current opinion in immunology.

[24]  T. Grogan,et al.  Phase I study of direct gene transfer of an allogeneic histocompatibility antigen, HLA-B7, in patients with metastatic melanoma. , 1997, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[25]  F. Levine,et al.  Development of a VSV-G protein pseudotyped retroviral vector system expressing dominant oncogenes from a lacO-modified inducible LTR promoter. , 1996, Gene.

[26]  P. Galle,et al.  Lymphocyte apoptosis induced by CD95 (APO–1/Fas) ligand–expressing tumor cells — A mechanism of immune evasion? , 1996, Nature Medicine.

[27]  M. Rudnicki,et al.  A helper-dependent adenovirus vector system: removal of helper virus by Cre-mediated excision of the viral packaging signal. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[28]  J. Tschopp,et al.  Melanoma Cell Expression of Fas(Apo-1/CD95) Ligand: Implications for Tumor Immune Escape , 1996, Science.

[29]  Edgar G. Engleman,et al.  Vaccination of patients with B–cell lymphoma using autologous antigen–pulsed dendritic cells , 1996, Nature Medicine.

[30]  D. Longo,et al.  Alterations in T cell receptor and signal transduction molecules in melanoma patients. , 1995, Clinical cancer research : an official journal of the American Association for Cancer Research.

[31]  D. Longo,et al.  Gradual loss of T-helper 1 populations in spleen of mice during progressive tumor growth. , 1995, Journal of the National Cancer Institute.

[32]  J. Allison,et al.  CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation , 1995, The Journal of experimental medicine.

[33]  J. Allison,et al.  T-cell activation: integration of signals from the antigen receptor and costimulatory molecules. , 1995, Immunology today.

[34]  P. Murawa,et al.  Gene therapy of human melanoma. Immunization of patients with autologous tumor cells admixed with allogeneic melanoma cells secreting interleukin 6 and soluble interleukin 6 receptor. , 1995, Human gene therapy.

[35]  S. Swain CD4 T cell development and cytokine polarization: an overview , 1995, Journal of leukocyte biology.

[36]  R. Brasseur,et al.  BAGE: a new gene encoding an antigen recognized on human melanomas by cytolytic T lymphocytes. , 1995, Immunity.

[37]  M. Wiznerowicz,et al.  Soluble interleukin 6 receptor is biologically active in vivo. , 1995, Cytokine.

[38]  L. Lanier,et al.  The role of CD28 costimulation in the generation of cytotoxic T lymphocytes. , 1995, Current topics in microbiology and immunology.

[39]  S. Nagata,et al.  Fas and Fas ligand: lpr and gld mutations. , 1995, Immunology today.

[40]  D. Longo,et al.  Alteration of signal transduction in T cells from cancer patients. , 1995, Important advances in oncology.

[41]  J. Allison,et al.  Specificity and longevity of antitumor immune responses induced by B7-transfected tumors. , 1994, Cancer research.

[42]  P. Bruggen,et al.  Autologous cytolytic T lymphocytes recognize a MAGE‐1 nonapeptide on melanomas expressing HLA‐Cw* 1601 , 1994, European journal of immunology.

[43]  P. Linsley,et al.  Comparative analysis of B7-1 and B7-2 costimulatory ligands: expression and function , 1994, The Journal of experimental medicine.

[44]  J. Bluestone,et al.  The B7 and CD28 receptor families. , 1994, Immunology today.

[45]  Lieping Chen,et al.  Costimulation of tumor-reactive CD4+ and CD8+ T lymphocytes by B7, a natural ligand for CD28, can be used to treat established mouse melanoma. , 1994, Journal of immunology.

[46]  K. Sakaguchi,et al.  Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes , 1994, The Journal of experimental medicine.

[47]  E. Jaffee,et al.  Role of bone marrow-derived cells in presenting MHC class I-restricted tumor antigens. , 1994, Science.

[48]  D. Pardoll,et al.  In vivo priming of two distinct antitumor effector populations: the role of MHC class I expression , 1994, The Journal of experimental medicine.

[49]  C. Figdor,et al.  Melanocyte lineage-specific antigen gp100 is recognized by melanoma- derived tumor-infiltrating lymphocytes , 1994, The Journal of experimental medicine.

[50]  C. Janeway,et al.  Signals and signs for lymphocyte responses , 1994, Cell.

[51]  P. Matzinger Tolerance, danger, and the extended family. , 1994, Annual review of immunology.

[52]  M Cohn,et al.  The wisdom of hindsight. , 1994, Annual review of immunology.

[53]  P. Stern,et al.  Natural history of HLA expression during tumour development. , 1993, Immunology today.

[54]  F. Marincola,et al.  Molecular mechanisms used by tumors to escape immune recognition: immunogenetherapy and the cell biology of major histocompatibility complex class I. , 1993, Journal of immunotherapy with emphasis on tumor immunology : official journal of the Society for Biological Therapy.

[55]  G. Starling,et al.  B7/BB-1 is a leucocyte differentiation antigen on human dendritic cells induced by activation. , 1993, Immunology.

[56]  E. Jaffee,et al.  Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. , 1993, Proceedings of the National Academy of Sciences of the United States of America.

[57]  J. Yewdell,et al.  Identification of human cancers deficient in antigen processing , 1993, The Journal of experimental medicine.

[58]  J. Allison,et al.  Tumor rejection after direct costimulation of CD8+ T cells by B7-transfected melanoma cells. , 1993, Science.

[59]  D. Lancki,et al.  Differential regulation of murine T lymphocyte subsets. , 1993, Annual review of immunology.

[60]  P. Linsley,et al.  Costimulation of antitumor immunity by the B7 counterreceptor for the T lymphocyte molecules CD28 and CTLA-4 , 1992, Cell.

[61]  Catia,et al.  A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E , 1992, The Journal of experimental medicine.

[62]  A. Porgador,et al.  Interleukin 6 gene transfection into Lewis lung carcinoma tumor cells suppresses the malignant phenotype and confers immunotherapeutic competence against parental metastatic cells. , 1992, Cancer research.

[63]  S. Romagnani Human TH1 and TH2 subsets: regulation of differentiation and role in protection and immunopathology. , 1992, International archives of allergy and immunology.

[64]  P. Chomez,et al.  A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. , 1991, Science.

[65]  T. Mosmann Cytokine secretion patterns and cross-regulation of T cell subsets , 1991, Immunologic research.

[66]  P. Greenberg Adoptive T cell therapy of tumors: mechanisms operative in the recognition and elimination of tumor cells. , 1991, Advances in immunology.

[67]  M. Herlyn,et al.  Structure, function, and clinical significance of human tumor antigens. , 1990, Journal of the National Cancer Institute.

[68]  E. Gilboa,et al.  Interleukin 2 gene transfer into tumor cells abrogates tumorigenicity and induces protective immunity , 1990, The Journal of experimental medicine.

[69]  B. Vogelstein,et al.  Interleukin-2 production by tumor cells bypasses T helper function in the generation of an antitumor response , 1990, Cell.

[70]  H. Schreiber,et al.  Unique tumor-specific antigens. , 1988, Annual review of immunology.

[71]  S. Rosenberg,et al.  A new approach to the adoptive immunotherapy of cancer with tumor-infiltrating lymphocytes. , 1986, Science.

[72]  P. Greenberg,et al.  H-2 restriction of adoptive immunotherapy of advanced tumors. , 1981, Journal of immunology.

[73]  F. Shen,et al.  Role of different T cells sets in the rejection of syngeneic chemically induced tumors. , 1979, Journal of immunology.

[74]  M. Herlyn,et al.  Study of antibodies against human melanoma produced by somatic cell hybrids. , 1978, Proceedings of the National Academy of Sciences of the United States of America.

[75]  H. Oettgen,et al.  Cell surface antigens of human malignant melanoma. II. Serological typing with immune adherence assays and definition of two new surface antigens , 1976, The Journal of experimental medicine.