US20180256694A1 - Composition for treating lung cancer, particularly of non-small lung cancers (nsclc) - Google Patents

Composition for treating lung cancer, particularly of non-small lung cancers (nsclc) Download PDF

Info

Publication number
US20180256694A1
US20180256694A1 US15/985,443 US201815985443A US2018256694A1 US 20180256694 A1 US20180256694 A1 US 20180256694A1 US 201815985443 A US201815985443 A US 201815985443A US 2018256694 A1 US2018256694 A1 US 2018256694A1
Authority
US
United States
Prior art keywords
mage
muc1
neu
eso
cea
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/985,443
Inventor
Marijke Barner
Jochen Probst
Thomas Lander
Ingmar Hoerr
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Curevac SE
Original Assignee
Curevac AG
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Curevac AG filed Critical Curevac AG
Priority to US15/985,443 priority Critical patent/US20180256694A1/en
Publication of US20180256694A1 publication Critical patent/US20180256694A1/en
Priority to US17/378,954 priority patent/US20220096616A1/en
Assigned to CureVac SE reassignment CureVac SE CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CUREVAC AG
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/572Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 cytotoxic response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/57Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2
    • A61K2039/575Medicinal preparations containing antigens or antibodies characterised by the type of response, e.g. Th1, Th2 humoral response
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/86Lung

Definitions

  • the present invention relates to an active (immunostimulatory) composition
  • an active (immunostimulatory) composition comprising at least one RNA, preferably a mRNA, encoding at least two (preferably different) antigens capable of eliciting an (adaptive) immune response in a mammal.
  • the invention furthermore relates to a vaccine comprising said active (immunostimulatory) composition, and to the use of said active (immunostimulatory) composition (for the preparation of a vaccine) and/or of the vaccine for eliciting an (adaptive) immune response for the treatment of lung cancer, particularly of non-small cell lung cancers (NSCLC), preferably selected from the three main sub-types squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, or of disorders related thereto.
  • NSCLC non-small cell lung cancers
  • the invention relates to kits, particularly to kits of parts, containing the active (immunostimulatory) composition and/or the vaccine.
  • bronchial carcinoma carcinoma of the lung.
  • TNM classification disease stage
  • subtype of carcinoma lung cancer
  • the main sub-types of lung cancer categorized by the size and appearance of the malignant cells identified under microscope are small cell lung cancer (20%) and non-small cell lung cancer (NSCLC) (80%).
  • This classification although based on simple histological criteria, has very important implications for clinical management and prognosis of the disease, with small cell lung cancer usually being treated by chemotherapy, while non-small cell lung cancer is mostly subject to surgery as a first-line treatment.
  • NSCLC non-small cell lung cancers
  • squamous cell lung carcinoma adenocarcinoma
  • large cell lung carcinoma adenocarcinoma
  • Therapeutic approaches in advanced disease involve—following surgery—both adjuvant chemotherapy and/or adjuvant radiotherapy, whereas chemotherapy as monotherapy (first-line therapy) seems to be an approach associated with relatively poor results. In a comparison of four commonly used combination chemotherapy regimens, none was superior.
  • Response rates varied from 15% to 22%, with 1-year survival rates of 31% to 36% (see e.g.
  • chemotherapeutic approaches used today are combinations of platin-based substances with e.g. Gemcitabin even as first-line-therapy, wheras e.g. Pemetrexed is used as second-line therapy.
  • NSCLC epidermal growth factor receptor
  • Tumorted Therapy Another option used for the treatment of NSCLC is the so-called “Targeted Therapy” trying to enhance success of classical cytotoxic chemotherapy by influencing tumor specific target structures on a molecular level.
  • Substances used include Bevacizumab (an angiogenesis inhibitor) or Erlotinib, which is aimed at the tyrosine kinases of the epidermal growth factor receptor (EGFR).
  • EGFR epidermal growth factor receptor
  • the immune system plays an important role in the treatment and prevention of numerous diseases. According to the present stage of knowledge, various mechanisms are provided by mammalians to protect the organism by identifying and killing e.g. tumor cells. These tumor cells have to be detected and distinguished from the organism's normal cells and tissues.
  • the immune system of vertebrates such as humans consists of many types of proteins, cells, organs, and tissues, which interact in an elaborate and dynamic network. As part of this more complex immune response, the vertebrate system adapts over time to recognize particular pathogens or tumor cells more efficiently.
  • the adaptation process creates immunological memories and allows even more effective protection during future encounters. This process of adaptive or acquired immunity forms the basis for vaccination strategies.
  • the adaptive immune system is antigen-specific and requires the recognition of specific “self” or “non-self” antigens during a process called antigen presentation. Antigen specificity allows for the generation of responses that are tailored to specific pathogens or pathogen-infected cells or tumor cells. The ability to mount these tailored responses is maintained in the body by so called “memory cells”. Should a pathogen infect the body more than once, these specific memory cells are used to quickly eliminate it.
  • the adaptive immune system thus allows for a stronger immune response as well as for an immunological memory, where each pathogen or tumor cell is “remembered” by one or more signature antigens.
  • lymphocytes as cellular components of the adaptive immune system include B cells and T cells which are derived from hematopoietic stem cells in the bone marrow. B cells are involved in the humoral response, whereas T cells are involved in cell mediated immune response. Both B cells and T cells carry receptor molecules that recognize specific targets. T cells recognize a “non-self” target, such as a pathogenic target structure, only after antigens (e.g. small fragments of a pathogen) have been processed and presented in combination with a “self” receptor called a major histocompatibility complex (MHC) molecule.
  • MHC major histocompatibility complex
  • the B cell antigen-specific receptor is an antibody molecule on the B cell surface, and recognizes pathogens as such when antibodies on its surface bind to a specific foreign antigen.
  • This antigen/antibody complex is taken up by the B cell and processed by proteolysis into peptides.
  • the B cell displays these antigenic peptides on its surface MHC class II molecules.
  • This combination of MHC and antigen attracts a matching helper T cell, which releases lymphokines and activates the B cell.
  • the activated B cell then begins to divide, its offspring secretes millions of copies of the antibody that recognizes this antigen.
  • These antibodies circulate in blood plasma and lymph, bind to pathogens or tumor cells expressing the antigen and mark them for destruction by complement activation or for uptake and destruction by phagocytes.
  • Cytotoxic T cells As a cellular component of the adaptive immune system cytotoxic T cells (CDS+) may form a CTL-response. Cytotoxic T cells (CDS+) can recognize peptides from endogenous pathogens and self-antigens bound by MHC type I molecules. CD8+-T cells carry out their killing function by releasing cytotoxic proteins in the cell.
  • Mechanisms of the immune system form targets for curative treatments.
  • Appropriate methods are typically based on the administration of adjuvants to elicit an innate immune response or on the administration of antigens or immunogens in order to evoke an adaptive immune response.
  • antigens are typically based on specific components of pathogens (e.g. surface proteins) or fragments thereof, administration of nucleic acids to the patient which is followed by the expression of desired polypeptides, proteins or antigens is envisaged as well.
  • DNA viruses may likewise be used as a DNA vehicle. Because of their infectious properties, such viruses achieve a very high transfection rate. The viruses used are genetically modified in such a manner that no functional infectious particles are formed in the transfected cell.
  • the DNA introduced into the cell is to be expressed, it is necessary for the corresponding DNA vehicle to contain a strong promoter, such as the viral CMV promoter.
  • a strong promoter such as the viral CMV promoter.
  • the integration of such promoters into the genome of the treated cell may result in unwanted alterations of the regulation of gene expression in the cell.
  • Another risk of using DNA as an agent to induce an immune response is the induction of pathogenic anti-DNA antibodies in the patient into whom the foreign DNA has been introduced, so bringing about a (possibly fatal) immune response.
  • an active (immunostimulatory) composition comprising at least one RNA, encoding at least two (preferably different) antigens selected from the group comprising the antigens:
  • an active (immunostimulatory) composition according to the present invention is capable to effectively stimulate the (adaptive) immune system to allow treatment of lung cancer, especially of non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • antigens, antigenic proteins or antigenic peptides may be used synomously.
  • an active (immunostimulatory) composition shall be further understood as a composition, which is able to elicit an immune response, preferably an adaptive immune response as defined herein, due to one of the component(s) contained or encoded by the components of the active (immunostimulatory) composition, preferably by the at least one RNA, preferably (m)RNA, encoding the at least two (preferably different) antigens.
  • the at least one RNA of the active (immunostimulatory) composition may encode hTERT.
  • hTERT is human telomerase reverse transcriptase and the preferred sequence of the RNA, preferably of the mRNA, encoding “hTERT”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 7 (SEQ ID NO: 7), and—even more preferably, in FIG. 8 (SEQ ID NO: 8).
  • FIG. 7 SEQ ID NO: 7
  • FIG. 8 SEQ ID NO: 8
  • telomere activity is increased in the vast majority of human tumors, making its gene product the first molecule common to all human tumors.
  • CTL cytotoxic T lymphocytes
  • telomerase reverse transcriptase telomerase reverse transcriptase
  • Carpenter and Vonderheide (2006) Carpenter, E. L. and R. H. Vonderheide (2006); “Telomerase-based immunotherapy of cancer.”
  • Expert Opin Biol Ther 6(10): 1031-9) reported that the progression from the cloning of human telomerase reverse transcriptase (hTERT) in 1997 to the first clinical trials of hTERT as an antitumor immunotherapy target has been swift.
  • hTERT is overexpressed in the vast majority of human cancers whereas it has limited expression in normal adult tissue.
  • hTERT is immunogenic both in vitro and in vivo.
  • Several Phase I studies of hTERT immunotherapy have been completed in patients with breast, prostate, lung and other cancers, and clinical and immunological results are encouraging.
  • the opportunity for vaccinating individuals as an immunoprevention strategy can also be envisioned for hTERT-based therapies. Nair, S. K. and Heiser et al.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an hTERT antigen selected from the sequence as shown in FIG. 7 (SEQ ID NO: 7), and more preferably, in FIG. 8 (SEQ ID NO: 8).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an hTERT antigen selected from a fragment, a variant or an epitope of an hTERT sequence as shown in FIG. 7 (SEQ ID NO: 7), and more preferably, as shown in FIG. 8 (SEQ ID NO: 8).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode WT1.
  • WT1 is Wilms tumor 1 and the preferred sequence of the RNA, preferably of the mRNA, encoding “WT1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 9 (SEQ ID NO: 9), more preferably in FIG. 10 (SEQ ID NO: 10), and even more preferably in FIG. 11 (SEQ ID NO: 11).
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an WT1 antigen selected from the sequence as shown in FIG. 9 (SEQ ID NO: 9), and more preferably, as shown in FIG. 10 (SEQ ID NO: 10) and even more preferably as shown in FIG. 11 (SEQ ID NO: 11).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an WT1 antigen selected from a fragment, a variant or an epitope of an WT1 sequence as shown in FIG. 9 (SEQ ID NO: 9), and more preferably, as shown in FIG. 10 (SEQ ID NO: 10) and even more preferably as shown in FIG. 11 (SEQ ID NO: 11).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-A2.
  • MAGE-A2 is the melanoma antigen family A, 2B and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-A2”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 14 (SEQ ID NO: 14), and even more preferably in FIG. 15 (SEQ ID NO: 15). Gillespie and Coleman (1999) (Gillespie, A. M. and R. E. Coleman (1999).
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-A2 antigen selected from the sequence as shown in FIG. 14 (SEQ ID NO: 14), and more preferably, as shown in FIG. 15 (SEQ ID NO: 15).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-A2 antigen selected from a fragment, a variant or an epitope of an MAGE-A2 sequence as shown in FIG. 14 (SEQ ID NO: 14), and—more preferably, as shown in FIG. 15 (SEQ ID NO: 15).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode 5T4.
  • 5T4 is trophoblast glycoprotein and the preferred sequence of the RNA, preferably of the mRNA, encoding “5T4”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 3 (SEQ ID NO: 3), and even more preferably—in FIG. 4 (SEQ ID NO: 4). Harrop, Connolly et al.
  • 5T4 oncotrophoblast glycoprotein janus molecule in life and a novel potential target against tumors.
  • Cell Mol Immunol 4(2): 99-104 reported that 5T4 oncotrophoblast glycoprotein is a transmembrane protein expressed on the embryonic tissue and various malignant tumor cell surfaces. It plays a vital role in the multiple biological and pathological processes including massive cellular migration during the embryogenesis, cell invasion associated with implantation, and neoplastic metastasis in the progression of tumorigenesis.
  • Kopreski, Benko et al. (2001) 5T4 is a trophoblast glycoprotein frequently overexpressed in epithelial malignancies that provides a potential target for cancer therapeutics (see Kopreski, M. S., F. A.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an 5T4 antigen selected from the sequence as shown in FIG. 3 (SEQ ID NO: 3), and more preferably, as shown in FIG. 4 (SEQ ID NO: 4).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an 5T4 antigen selected from a fragment, a variant or an epitope of an 5T4 sequence as shown in FIG. 3 (SEQ ID NO: 3), and—more preferably, as shown in FIG. 4 (SEQ ID NO: 4).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-A3.
  • MAGE-A3 is the melanoma antigen family A, 3 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-A3”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 16 (SEQ ID NO: 16), and even more preferably in FIG. 17 (SEQ ID NO: 17). Gillespie and Coleman (1999) (Gillespie, A. M. and R. E. Coleman (1999).
  • Varwark et al. (2004) described a study performed to determine the rate of MAGE-A3 expression in early-stage non-small cell lung cancer (NSCLC) (see Sienel, W., C. Vartechnik, et al. (2004). “Melanoma associated antigen (MAGE)-A3 expression in Stages I and II non-small cell lung cancer: results of a multi-center study.” Eur J Cardiothorac Surg 25(1): 131-4). Primary tumor samples from 204 patients with operable clinical stages I or II NSCLC were collected and the pathological stage determined. MAGE-A3 expression was analyzed from tissue samples by detection of MAGE-A3 transcripts using reverse-transcriptase polymerase chain reaction.
  • MAGE-A3 expression was observed in 80 out of the 204 (39.2%) examined stages I-II primary tumors. Atanackovic, Altorki et al. (2004) described that MAGE-A3 a tumor-associated antigen originally identified in melanoma, was also found in non-small cell lung tumors (see
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-A3 antigen selected from the sequence as shown in FIG. 16 (SEQ ID NO: 16), and more preferably, as shown in FIG.
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-A3 antigen selected from a fragment, a variant or an epitope of an MAGE-A3 sequence as shown in FIG. 16 (SEQ ID NO: 16), and more preferably, as shown in FIG. 17 (SEQ ID NO: 17).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode MUC1.
  • MUC1 is mucin 1 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MUC1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 1 (SEQ ID NO: 1), and even more preferably in FIG. 2 (SEQ ID NO: 2).
  • Cancer-associated mucins are a potential target for immunotherapy. These molecules are thought to promote metastases by facilitating adhesion of malignant cells to the endothelial cell surface. According to Denda-Nagai and Irimura (2000) (Denda-Nagai, K. and T.
  • MUC1 in carcinoma-host interactions MUC-1 is overexpressed in 90% of all adenocarcinomas, including breast, lung, pancreas, prostate, stomach, colon and ovary.
  • Kontani, Taguchi et al. (2001) found that MUC-1 has been found to be expressed in 60% of lung cancers (see Kontani, K., 0. Taguchi, et al. (2001). “Modulation of MUC1 mucin as an escape mechanism of breast cancer cells from autologous cytotoxic T-lymphocytes.” Br J Cancer 84(9): 1258-64), whereas Kontani, Taguchi et al.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an MUC1 antigen selected from the sequence as shown in FIG. 1 (SEQ ID NO: 1), and more preferably, as shown in FIG. 2 (SEQ ID NO: 2).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MUC1 antigen selected from a fragment, a variant or an epitope of an MUC1 sequence as shown in FIG. 1 (SEQ ID NO: 1), and more preferably, as shown in FIG. 2 (SEQ ID NO: 2).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode Her-2/neu.
  • Her-2/neu is v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 and the preferred sequence of the RNA, preferably of the mRNA, encoding “Her-2/neu”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 5 (SEQ ID NO: 5), and even more preferably in FIG. 6 (SEQ ID NO: 6).
  • FIG. 5 SEQ ID NO: 5
  • FIG. 6 SEQ ID NO: 6
  • HER-2/neu also known as HER2 or c-erb-B2
  • EGF epidermal growth factor
  • HER-2/neu is expressed in many epithelial tumors and known to be overexpressed in approximately 20-25% of all ovarian and breast cancers, 35-45% of all pancreatic adenocarcinomas, and up to 90% of colorectal carcinomas.
  • HER-2/neu overexpression represents a marker of poor prognosis.
  • Overexpression of Her-2 has been observed in malignant tumors of the breast, ovary, pancreas, colon, lung and other tissues.
  • Her-2 is normally expressed at low levels in variety of human tissues (skin, digestive tract epithelium, breast, ovary, hepatocytes).
  • Her-2/neu is a member of the EGFR familiy (see Disis, M. L., T. A. Gooley, et al. (2002). “Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines.” J Clin Oncol 20(11): 2624-32). It is frequently overexpressed in breast, ovary, prostate, colon and lung cancers.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an Her-2/neu antigen selected from the sequence as shown in FIG. 5 (SEQ ID NO: 5), and more preferably, as shown in FIG. 6 (SEQ ID NO: 6).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an Her-2/neu antigen selected from a fragment, a variant or an epitope of an Her-2/neu sequence as shown in FIG. 5 (SEQ ID NO: 5), and more preferably, as shown in FIG. 6 (SEQ ID NO: 6).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode NY-ESO-1.
  • NY-ESO-1 is cancer/testis antigen 1B and the preferred sequence of the RNA, preferably of the mRNA, encoding “NY-ESO-1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 20 (SEQ ID NO: 20), and even more preferably—in FIG. 21 (SEQ ID NO: 21). Chen, Scanlan et al.
  • NY-ESO-1-specific antibody responses and/or specific CD8 and CD4 T cell responses directed against a broad range of NY-ESO-1 epitopes were induced by a course of at least four vaccinations at monthly intervals in a high proportion of patients.
  • CD8 T cell clones derived from five vaccinated patients were shown to lyse NY-ESO-1-expressing melanoma target cells. In several patients with melanoma, there was a strong impression that the natural course of the disease was favorably influenced by vaccination. Davis, Chen et al. (2004) reported that HLA-A2-restricted NY-ESO-1 peptides injected intradermally were shown to be safe and immunogenic (Davis, I.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an NY-ESO-1 antigen selected from the sequence as shown in FIG. 20 (SEQ ID NO: 20), and more preferably, as shown in FIG.
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an NY-ESO-1 antigen selected from a fragment, a variant or an epitope of an NY-ESO-1 sequence as shown in FIG. 20 (SEQ ID NO: 20), and more preferably, as shown in FIG. 21 (SEQ ID NO: 21).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode CEA.
  • CEA is a 180 kDa onco-fetal glycoprotein that acts as an adhesion molecule, and is overexpressed in 70% of NSCLC (Hammarstrom, S. (1999). “The carcinoembryonic antigen (CEA) family: structures, suggested functions and expression in normal and malignant tissues.” Semin Cancer Biol 9(2): 67-81). Berinstein (2002) reported that CEA has many attractive features as a target for active vaccination approaches against cancer (Berinstein, N. L. (2002). “Carcinoembryonic antigen as a target for therapeutic anticancer vaccines: a review.” J Clin Oncol 20(8): 2197-207). It has a favorable expression pattern and is expressed in more than 50% of all human cancers.
  • CEA may play a role in the tumorigenesis process itself, and thus its expression may be selected and conserved throughout cancer progression. It has been well documented that CEA is processed and presented on various MHC class 1 molecules. Moreover, immunologic tolerance to CEA is not absolute. There are extensive data demonstrating that human T cells can recognize, become activated to, and lyse cancer cells that are expressing CEA. Several different therapeutic vaccination approaches using CEA as a target antigen have been assessed. The safety of these approaches has been established. In addition, humoral and/or cellular responses to CEA have been documented.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an CEA antigen selected from the sequence as shown in FIG. 12 (SEQ ID NO: 12), and—more preferably, as shown in FIG. 13 (SEQ ID NO: 13).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an CEA antigen selected from a fragment, a variant or an epitope of an CEA sequence as shown in FIG. 12 (SEQ ID NO: 12), and more preferably, as shown in FIG. 13 (SEQ ID NO: 13).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode Survivin.
  • Survivin is baculoviral IAP repeat-containing 5 (survivin) and the preferred sequence of the RNA, preferably of the mRNA, encoding “survivin”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 18 (SEQ ID NO: 18), and even more preferably in FIG. 19 (SEQ ID NO: 19).
  • FIG. 18 SEQ ID NO: 18
  • FIG. 19 SEQ ID NO: 19
  • CD8+ T cells reactive to survivin antigen in patients with multiple myeloma Survivin is a member of the inhibitors of apoptosis family and is overexpressed in different types of malignancies. Cytotoxic T cells recognizing survivin epitopes can be elicited in vitro and by vaccination in patients with leukemia, breast cancer, and melanoma. It was investigated whether survivin-specific CD8+ T cells occur in patients with multiple myeloma and T cells recognizing HLA-A2.1binding survivin peptide were detected in 9 of 23 patients and in 1 of 21 healthy volunteers.
  • Survivin-reactive T cells were identified as terminally differentiated effector T cells (CD8+, CD45RA+, and CCR7). Positive survivin expression of myeloma cells in bone marrow specimens was shown in 7 of 11 patients. Survivin is highly expressed in most human cancer cells of epithelial and hematopoietic origin, and overexpression is associated with cancer progression, poor prognosis, resistance, and short patient survival. Duffy, O'Donovan (2007) described that Survivin is a 16.5 kDa protein overexpressed in almost all malignancies but rarely detected in normal differentiated adult tissues (see Duffy, M. J., N. O'Donovan, et al. (2007).
  • survivin a promising tumor biomarker. Cancer Lett 249(1): 49-60). Functionally, survivin has been shown to inhibit apoptosis, promote cell proliferation and enhance angiogenesis. Consistent with its role in these processes, survivin was described as playing a key role in cancer progression. Because of the large difference in expression between normal and malignant tissue and its causal role in cancer progression, survivin is currently undergoing intensive investigation as a potential tumor marker. Emerging data suggests that measurement of survivin can aid the early diagnosis of bladder cancer, determine prognosis in multiple cancer types and predict response to diverse anti-cancer therapies. Zeis, Siegel et al.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an Survivin antigen selected from the sequence as shown in FIG. 18 (SEQ ID NO: 18), and more preferably, as shown in FIG. 19 (SEQ ID NO: 19).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an Survivin antigen selected from a fragment, a variant or an epitope of an Survivin sequence as shown in FIG. 18 (SEQ ID NO: 18), and more preferably, as shown in FIG. 19 (SEQ ID NO: 19).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-C1.
  • MAGE-C1 is the melanoma antigen family C, 1 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-C 1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 22 (SEQ ID NO: 22), more preferably in FIG. 23 (SEQ ID NO: 23), and even more preferably—in FIG. 24 (SEQ ID NO: 24).
  • Lucas, De Smet et al. 1998 recently identified MAGE-C1 by performing RDA (see Lucas, S., C. De Smet, et al. (1998). “Identification of a new MAGE gene with tumor-specific expression by representational difference analysis.”
  • MAGE-C1 was not expressed in a panel of normal tissues tested with the exception of testis. Among tumoral samples, MAGE-C1 was frequently expressed in seminomas, melanomas, and bladder carcinomas. It was also expressed in a significant fraction of head and neck carcinomas, breast carcinomas, non-small lung carcinomas, prostate adenocarcinomas and sarcomas.
  • C7 MAGE-C1 antigen expression in normal and neoplastic tissues. Int J Cancer 99(6): 839-45). Gure, Chua et al. (2005) analyzed tumors from 523 non-small-cell lung cancer (NSCLC) patients for the expression of cancer-testis antigens (see Gure, A. O., R. Chua, et al. (2005). “Cancer-testis genes are coordinately expressed and are markers of poor outcome in non-small cell lung cancer.” Clin Cancer Res 11(22): 8055-62). MAGE-C1 was present in 18,8%. Scanlan, Altorki et al.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-C1 antigen selected from the sequence as shown in FIG. 22 (SEQ ID NO: 22), and—more preferably, as shown in FIG. 23 (SEQ ID NO: 23) and even more preferably as shown in FIG.
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-C 1 antigen selected from a fragment, a variant or an epitope of an MAGE-C1 sequence as shown in FIG. 22 (SEQ ID NO: 22), and more preferably, as shown in FIG. 23 (SEQ ID NO: 23) and even more preferably as shown in FIG. 24 (SEQ ID NO: 24).
  • the at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-C2.
  • MAGE-C2 is the melanoma antigen family C2 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-C2”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 25 (SEQ ID NO: 25), and—even more preferably in FIG. 26 (SEQ ID NO: 26).
  • Lucas, De Plaen et al. (2000) recently identified MAGE-C2 by performing RDA on a melanoma cell line (see Lucas, S., E. De Plaen, et al. (2000).
  • MAGE-B5, MAGE-B6, MAGE-C2, and MAGE-C3 four new members of the MAGE family with tumor-specific expression.” Int J Cancer 87(1): 55-60). MAGE-C2 was not expressed in a panel of normal tissues tested with the exception of testis. Among tumoral samples, MAGE-C2 was frequently expressed in seminomas, melanomas, and bladder carcinomas. It was also expressed in a significant fraction of head and neck carcinomas, breast carcinomas, non-small lung carcinomas and sarcomas. Scanlan, Altorki et al. (2000) reported expression of CT antigens in 33 non-small cell lung cancers: MAGE-C2: 30% (see Scanlan, M. J., N. K.
  • the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-C2 antigen selected from the sequence as shown in FIG. 25 (SEQ ID NO: 25), and more preferably, as shown in FIG. 26 (SEQ ID NO: 26).
  • the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-C2 antigen selected from a fragment, a variant or an epitope of an MAGE-C2 sequence as shown in FIG. 25 (SEQ ID NO: 25), and more preferably, as shown in FIG. 26 (SEQ ID NO: 26).
  • Antigens, antigenic proteins or antigenic peptides as defined above which may be encoded by the at least one RNA of the active (immunostimulatory) composition according to the present invention may comprise fragments or variants of those sequences.
  • Such fragments or variants may typically comprise a sequence having a sequence homology with one of the above mentioned antigens, antigenic proteins or antigenic peptides or sequences or their encoding nucleic acid sequences of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, preferably at least 70%, more preferably at least 80%, equally more preferably at least 85%, even more preferably at least 90% and most preferably at least 95% or even 97%, to the entire wild-type sequence, either on nucleic acid level or on amino acid level.
  • “Fragments” of antigens, antigenic proteins or antigenic peptides in the context of the present invention may comprise a sequence of an antigen, antigenic protein or antigenic peptide as defined above, which is, with regard to its amino acid sequence (or its encoded nucleic acid sequence), N-terminally, C-terminally and/or intrasequentially truncated compared to the amino acid sequence of the original (native) protein (or its encoded nucleic acid sequence). Such truncation may thus occur either on the amino acid level or correspondingly on the nucleic acid level.
  • a sequence homology with respect to such a fragment as defined above may therefore preferably refer to the entire antigen, antigenic protein or antigenic peptide as defined above or to the entire (coding) nucleic acid sequence of such an antigen, antigenic protein or antigenic peptide.
  • Fragments of antigens, antigenic proteins or antigenic peptides in the context of the present invention may furthermore comprise a sequence of an antigen, antigenic protein or antigenic peptide as defined above, which has a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 11, or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g. 13, 14, 15, 16, 17, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence.
  • These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognized in their native form.
  • Fragments of antigens, antigenic proteins or antigenic peptides as defined herein may also comprise epitopes of those antigens, antigenic proteins or antigenic peptides.
  • Epitopes also called “antigen determinants” in the context of the present invention are typically fragments located on the outer surface of (native) antigens, antigenic proteins or antigenic peptides as defined herein, preferably having 5 to 15 amino acids, more preferably having 5 to 12 amino acids, even more preferably having 6 to 9 amino acids, which may be recognized by antibodies or B-cell receptors, i.e. in their native form.
  • antigenic determinants can be conformational or discontinous epitopes which are composed of segments of the antigens, antigenic proteins or antigenic peptides as defined herein that are discontinuous in the amino acid sequence of the antigens, antigenic proteins or antigenic peptides as defined herein but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain.
  • “Variants” of antigens, antigenic proteins or antigenic peptides as defined above may be encoded by the at least one RNA of the active (immunostimulatory) composition according to the present invention, wherein nucleic acids of the at least one (m)RNA, encoding the antigen, antigenic protein or antigenic peptide as defined above, are exchanged.
  • an antigen, antigenic protein or antigenic peptide may be generated, having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s).
  • these fragments and/or variants have the same biological function or specific activity compared to the full-length native antigen or antigenic potein, e.g. its specific antigenic property.
  • the at least one RNA of the active (immunostimulatory) composition according to the present invention may also encode an antigen or an antigenic protein as defined above, wherein the encoded amino acid sequence comprises conservative amino acid substitution(s) compared to its physiological sequence.
  • the encoded amino acid sequences as well as their encoding nucleotide sequences in particular fall under the term variants as defined above.
  • Substitutions in which amino acids which originate from the same class are exchanged for one another are called conservative substitutions.
  • these are amino acids having aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function. This means that e.g.
  • an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g. serine (threonine) by threonine (serine) or leucine (isoleucine) by isoleucine (leucine)).
  • Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g.
  • CD spectra circular dichroism spectra
  • variants of antigens, antigenic proteins or antigenic peptides as defined above which may be encoded by the at least one RNA of the active (immunostimulatory) composition according to the present invention, may also comprise those sequences, wherein nucleic acids of the at least one (m)RNA are exchanged according to the degeneration of the genetic code, without leading to an alteration of respective amino acid sequence of the antigen, antigenic protein or antigenic peptide, i.e. the amino acid sequence or at least part thereof may not differ from the original sequence in one or more mutation(s) within the above meaning.
  • nucleic acid sequences e.g. RNA or mRNA sequences as defined herein, or amino acid sequences, preferably their encoded amino acid sequences, e.g. the amino acid sequences of the antigens, antigenic proteins or antigenic peptides as defined above
  • the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. gaps can be inserted into the sequence of the first sequence and the component at the corresponding position of the second sequence can be compared. If a position in the first sequence is occupied by the same component as is the case at a position in the second sequence, the two sequences are identical at this position.
  • the percentage to which two sequences are identical is a function of the number of identical positions divided by the total number of positions.
  • the percentage to which two sequences are identical can be determined using a mathematical algorithm.
  • a preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et al. (1993), PNAS USA, 90:5873-5877 or Altschul et al. (1997), Nucleic Acids Res., 25:3389-3402. Such an algorithm is integrated in the BLAST program. Sequences which are identical to the sequences of the present invention to a certain extent can be identified by this program.
  • the active (immunostimulatory) composition according to the present invention comprises, as defined above, at least one RNA, encoding least two (preferably different) antigens selected from any of the antigens of the above group, since according to the invention a specific combination of at least two (preferably different) antigens of the afore mentioned group is capable to effectively stimulate the (adaptive) immune system to allow treatment of lung cancer, especially of non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • the present invention may also provide such active (immunostimulatory) compositions, comprising at least one RNA, encoding three, four, five, six, seven, eight, nine, ten, eleven or even even twelve (preferably different) antigens selected from any of the antigens of the above group, wherein any combination of these antigens is possible and envisaged.
  • the at least one RNA of the active (immunostimulatory) composition according to the present invention may encode at least two (preferably different) antigens selected from any of the antigens of a subgroup comprising the following antigens:
  • the present invention may also provide an active (immunostimulatory) composition, comprising at least one RNA, encoding at least three, four, five or six (preferably different) antigens selected from any of the antigens of the above group or subgroup, wherein any combination of these antigens is possible.
  • an active (immunostimulatory) composition comprising at least one RNA, encoding at least three, four, five or six (preferably different) antigens selected from any of the antigens of the above group or subgroup, wherein any combination of these antigens is possible.
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may encode at least two (preferably different) antigens selected from any of the antigens of the above mentioned group(s) or subgroup(s) comprising (at least) any one of the following combinations of antigens:
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may encode at least two (preferably different) antigens exclusively selected from any of the antigens of the above mentioned group(s) or subgroup(s) comprising (at least) any one of the following combinations of antigens:
  • the present invention provides an active (immunostimulatory) composition
  • an active (immunostimulatory) composition comprising at least one RNA, encoding at least two (preferably different) antigens,
  • the at least one antigen(s) according to a) is (are) selected from:
  • the at least one antigen(s) according to a) is (are) selected from:
  • the at least one antigen(s) according to b) is (are) selected from an antigen (antigens) as defined in one of the following combinations:
  • the at least one antigen(s) according to b) is (are) selected from the following specific combination of antigens as defined above:
  • the at least one RNA of the active (immunostimulatory) composition according to the present invention is typically any RNA, preferably, without being limited thereto, a coding RNA, a circular or linear RNA, a single- or a double-stranded RNA (which may also be regarded as a
  • RNA due to non-covalent association of two single-stranded RNA) or a partially double-stranded or partially single stranded RNA, which are at least partially self complementary (both of these partially double-stranded or partially single stranded RNA molecules are typically formed by a longer and a shorter single-stranded RNA molecule or by two single stranded RNA-molecules, which are about equal in length, wherein one single-stranded RNA molecule is in part complementary to the other single-stranded RNA molecule and both thus form a double-stranded RNA in this region, i.e. a partially double-stranded or partially single stranded RNA with respect to the entire RNA sequence).
  • the at least one RNA of the active (immunostimulatory) composition according to the present invention is a single-stranded RNA, even more preferably a linear RNA.
  • the at least RNA of the active (immunostimulatory) composition according to the present invention is a messenger RNA (mRNA).
  • mRNA messenger RNA
  • a messenger RNA (mRNA) is typically a RNA, which is composed of (at least) several structural elements, e.g. an optional 5′-UTR region, an upstream positioned ribosomal binding site followed by a coding region, an optional 3′-UTR region, which may be followed by a poly-A tail (and/or a poly-C-tail).
  • each of the at least two (preferably different) antigens of the active (immunostimulatory) composition of the present invention may be encoded by one (monocistronic) RNA, preferably one (monocistronic) mRNA.
  • the active (immunostimulatory) composition of the present invention may contain at least two (monocistronic) RNAs, preferably mRNAs, wherein each of these at least two (monocistronic) RNAs, preferably mRNAs, may encode just one (preferably different) antigen, selected from one of the above mentioned groups or subgroups, preferably in one of the above mentioned combinations.
  • the active (immunostimulatory) composition of the present invention may comprise (at least) one bi- or even multicistronic RNA, preferably mRNA, i.e. (at least) one RNA which carries two or even more of the coding sequences of at the least two (preferably different) antigens, selected from one of the above mentioned groups or subgroups, preferably in one of the above mentioned combinations.
  • Such coding sequences of the at least two (preferably different) antigens of the (at least) one bi- or even multicistronic RNA may be separated by at least one IRES (internal ribosomal entry site) sequence, as defined below.
  • the term “encoding at least two (preferably different) antigens” may mean, without being limited thereto, that the (at least) one (bi- or even multicistronic) RNA, preferably a mRNA, may encode e.g. at least two, three, four, five, six, seven, eight, nine, ten, eleven or twelve (preferably different) antigens of the above mentioned group(s) of antigens or their fragments or variants within the above definitions. More preferably, without being limited thereto, the (at least) one (bi- or even multicistronic) RNA, preferably mRNA, may encode e.g.
  • IRES internal ribosomal entry site
  • IRES sequences can function as a sole ribosome binding site, but it can also serve to provide a bi- or even multicistronic RNA as defined above which codes for several proteins, which are to be translated by the ribosomes independently of one another.
  • IRES sequences which can be used according to the invention are those from picornaviruses (e.g.
  • FMDV pestiviruses
  • CFFV pestiviruses
  • PV polioviruses
  • ECMV encephalomyocarditis viruses
  • FMDV foot and mouth disease viruses
  • HCV hepatitis C viruses
  • CSFV classical swine fever viruses
  • MLV mouse leukoma virus
  • SIV simian immunodeficiency viruses
  • CrPV cricket paralysis viruses
  • the active (immunostimulatory) composition of the present invention may comprise a mixture of at least one monocistronic RNA, preferably mRNA, as defined above, and at least one bi- or even multicistronic RNA, preferably mRNA, as defined above.
  • the at least one monocistronic RNA and/or the at least one bi- or even multicistronic RNA preferably encode different antigens or their fragments or variants within the above definitions, the antigens preferably being selected from one of the above mentioned groups or subgroups of antigens, more preferably in one of the above mentioned combinations.
  • the at least one monocistronic RNA and the at least one bi- or even multicistronic RNA may preferably also encode (in part) identical antigens selected from one of the above mentioned groups or subgroups of antigens, preferably in one of the above mentioned combinations, provided that the active (immunostimulatory) composition of the present invention as a whole provides at least two (preferably different) antigens as defined above.
  • Such an embodiment may be advantageous e.g. for a staggered, e.g. time dependent, administration of the active (immunostimulatory) composition of the present invention to a patient in need thereof.
  • Such an active (immunostimulatory) composition of the present invention particularly the different RNAs encoding the at least two (preferably different) antigens, may be e.g. contained in (different parts of) a kit of parts composition or may be e.g. administered separately as components of different active (immunostimulatory) compositions according to the present invention.
  • the at least one RNA of the active (immunostimulatory) composition typically comprises a length of about 50 to about 20000, or 100 to about 20000 nucleotides, preferably of about 250 to about 20000 nucleotides, more preferably of about 500 to about 10000, even more preferably of about 500 to about 5000.
  • the at least one RNA of the active (immunostimulatory) composition encoding at least two (preferably different) antigens selected from the above defined group(s) or subgroup(s) of antigens, more preferably in the above combinations, may be in the form of a modified RNA, wherein any modification, as defined herein, may be introduced into the at least one RNA of the active (immunostimulatory) composition. Modifications as defined herein preferably lead to a stabilized at least one RNA of the active (immunostimulatory) composition of the present invention.
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may thus be provided as a “stabilized RNA”, preferably a stabilized mRNA, that is to say as an (m)RNA that is essentially resistant to in vivo degradation (e.g. by an exo- or endo-nuclease).
  • a stabilized RNA preferably a stabilized mRNA, that is to say as an (m)RNA that is essentially resistant to in vivo degradation (e.g. by an exo- or endo-nuclease).
  • stabilization can be effected, for example, by a modified phosphate backbone of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention.
  • a backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides contained in the RNA are chemically modified.
  • Nucleotides that may be preferably used in this connection contain e.g. a phosphorothioate-modified phosphate backbone, preferably at least one of the phosphate oxygens contained in the phosphate backbone being replaced by a sulfur atom.
  • Stabilized (m)RNAs may further include, for example: non-ionic phosphate analogues, such as, for example, alkyl and aryl phosphonates, in which the charged phosphonate oxygen is replaced by an alkyl or aryl group, or phosphodiesters and alkylphosphotriesters, in which the charged oxygen residue is present in alkylated form.
  • Such backbone modifications typically include, without implying any limitation, modifications from the group consisting of methylphosphonates, phosphoramidates and phosphorothioates (e.g. cytidine-5′-O-(1-thiophosphate)).
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may additionally or alternatively also contain sugar modifications.
  • a sugar modification in connection with the present invention is a chemical modification of the sugar of the nucleotides of the at least one RNA and typically includes, without implying any limitation, sugar modifications selected from the group consisting of 2′-deoxy-2′-fluoro-oligoribonucleotide (2′-fluoro-2′-deoxycytidine-5′-triphosphate, 2′-fluoro-2′-deoxyuridine-5′-triphosphate), 2′-deoxy-2′-deamine oligoribonucleotide (2′-amino-2′-deoxycytidine-5′-triphosphate, 2′-amino-2′-deoxyuridine-5′-triphosphate), 2′-O-alkyl oligoribonucleotide, 2′-deoxy-2′-C-alkyl oligoribonucleotide (2′-O-methyl
  • Significant in this case means an increase in the expression of the protein compared with the expression of the native RNA sequence by at least 20%, preferably at least 30%, 40%, 50% or 60%, more preferably by at least 70%, 80%, 90% or even 100% and most preferably by at least 150%, 200% or even 300% or more.
  • a nucleotide having such a base modification is preferably selected from the group of the base-modified nucleotides consisting of 2-amino-6-chloropurineriboside-5′-triphosphate, 2-aminoadenosine-5′-triphosphate, 2-thiocytidine-5′-triphosphate, 2-thiouridine-5′-triphosphate, 4-thiouridine-5′-triphosphate, 5-aminoallylcytidine-5′-triphosphate, 5-aminoallyluridine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, 5-bromouridine-5′-triphosphate, 5-iodocytidine-5′-triphosphate, 5-iodouridine-5′-triphosphate, 5-methylcytidine-5′-triphosphate, 5-methyluridine-5′-triphosphate, 6-azacytidine-5′-triphosphate, 6-azauridine-5′-triphosphate, 6-chloropurinerib
  • nucleotides for base modifications selected from the group of base-modified nucleotides consisting of 5-methylcytidine-5′-triphosphate, 7-deazaguanosine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, and pseudouridine-5′-triphosphate.
  • the at least one RNA of the active (immunostimulatory) composition of the present invention can likewise be modified (and preferably stabilized) by introducing further modified nucleotides containing modifications of their ribose or base moieties.
  • nucleotide analogues are defined as non-natively occurring variants of naturally occurring nucleotides.
  • analogues are chemically derivatized nucleotides with non-natively occurring functional groups, which are preferably added to or deleted from the naturally occurring nucleotide or which substitute the naturally occurring functional groups of a nucleotide. Accordingly, each component of the naturally occurring nucleotide may be modified, namely the base component, the sugar (ribose) component and/or the phosphate component forming the backbone (see above) of the RNA sequence.
  • Analogues of guanosine, uracil, adenosine, and cytosine include, without implying any limitation, any naturally occurring or non-naturally occurring guanosine, uracil, adenosine, thymidine or cytosine that has been altered chemically, for example by acetylation, methylation, hydroxylation, etc., including 1-methyl-adenosine, 1-methyl-guanosine, 1-methyl-inosine, 2,2-dimethyl-guanosine, 2,6-diaminopurine, 2′-Amino-2′-deoxyadenosine, 2′-Amino-2′-deoxycytidine, 2′-Amino-2′-deoxyguanosine, 2′-Amino-2′-deoxyuridine, 2-Amino-6-chloropurineriboside, 2-Aminopurine-riboside, 2′-Araadenosine, 2′-Aracy
  • analogue as described above, particular preference may be given according to the invention to those analogues that increase the immunogenity of the RNA of the inventive active (immunostimulatory) composition and/or do not interfere with a further modification of the RNA that has been introduced.
  • the at least one RNA of the active (immunostimulatory) composition of the present invention can contain a lipid modification.
  • a lipid-modified RNA typically comprises a RNA as defined herein, encoding at least two antigens selected from the group or subgroup of antigens as defined above, preferably in the above combinations.
  • Such a lipid-modified RNA typically further comprises at least one linker covalently linked with that RNA, and at least one lipid covalently linked with the respective linker.
  • the lipid-modified RNA comprises an at least one RNA as defined herein and at least one (bifunctional) lipid covalently linked (without a linker) with that RNA.
  • the lipid-modified RNA comprises a RNA as defined herein, at least one linker covalently linked with that RNA, and at least one lipid covalently linked with the respective linker, and also at least one (bifunctional) lipid covalently linked (without a linker) with that RNA.
  • the lipid contained in the at least one RNA of the inventive active (immunostimulatory) composition is typically a lipid or a lipophilic residue that preferably is itself biologically active.
  • Such lipids preferably include natural substances or compounds such as, for example, vitamins, e.g. alpha-tocopherol (vitamin E), including RRR-alpha-tocopherol (formerly D-alpha-tocopherol), L-alpha-tocopherol, the racemate D,L-alpha-tocopherol, vitamin E succinate (VES), or vitamin A and its derivatives, e.g. retinoic acid, retinol, vitamin D and its derivatives, e.g.
  • vitamins e.g. alpha-tocopherol (vitamin E), including RRR-alpha-tocopherol (formerly D-alpha-tocopherol), L-alpha-tocopherol, the racemate D,L-alpha-tocopherol, vitamin E succinate (VES), or
  • bile acids for example cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxygenin, testosterone, cholesterol or thiocholesterol.
  • Further lipids or lipophilic residues within the scope of the present invention include, without implying any limitation, polyalkylene glycols (Oberhauser et al., Nucl.
  • aliphatic groups such as, for example, C1-C20-alkanes, C1-C20-alkenes or C1-C20-alkanol compounds, etc., such as, for example, dodecanediol, hexadecanol or undecyl residues (Saison-Behmoaras et al., EMBO J, 1991, 10, 111; Kabanov et al., FEBS Lett., 1990, 259, 327; Svinarchuk et al., Biochimie, 1993, 75, 49), phospholipids such as, for example, phosphatidylglycerol, diacylphosphatidylglycerol, phosphatidylcholine, dipalmitoylphosphatidylcholine, distearoylphosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, di-hexadec
  • polyamines or polyalkylene glycols such as, for example, polyethylene glycol (PEG) (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969), hexaethylene glycol (HEG), palmitin or palmityl residues (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229), octadecylamines or hexylamino-carbonyl-oxycholesterol residues (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923), and also waxes, terpenes, alicyclic hydrocarbons, saturated and mono- or poly-unsaturated fatty acid residues, etc.
  • PEG polyethylene glycol
  • HEG hexaethylene glycol
  • HOG hexaethylene glycol
  • palmitin or palmityl residues Mishra et al
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may likewise be stabilized in order to prevent degradation of the RNA in vivo by various approaches. It is known in the art that instability and (fast) degradation of mRNA or of RNA in vivo in general may represent a serious problem in the application of RNA based compositions.
  • RNAases RNA-degrading enzymes
  • RNase contaminations may be generally removed by special treatment prior to use of said compositions, in particular with diethyl pyrocarbonate (DEPC).
  • DEPC diethyl pyrocarbonate
  • a number of mechanisms of natural degradation are known in this connection in the prior art, which may be utilized as well.
  • the terminal structure is typically of critical importance for a mRNA in vivo.
  • cap structure a modified guanosine nucleotide
  • poly-A tail a sequence of up to 200 adenosine nucleotides
  • the at least one RNA of the active (immunostimulatory) composition of the present invention can therefore be stabilized against degradation by RNases by the addition of a so-called “5′ cap” structure.
  • a so-called “5′ cap” structure Particular preference is given in this connection to an m7G(5′)ppp (5′(A,G(5′)ppp(5′)A or G(5′)ppp(5′)G as the 5′ cap” structure.
  • such a modification is introduced only if a modification, for example a lipid modification, has not already been introduced at the 5′ end of the (m)RNA of the inventive immunostimulatory composition or if the modification does not interfere with the immunogenic properties of the (unmodified or chemically modified) (m)RNA.
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may contain, especially if the RNA is in the form of a mRNA, a poly-A tail on the 3′ terminus of typically about 10 to 200 adenosine nucleotides (SEQ ID NO: 30), preferably about 10 to 100 adenosine nucleotides (SEQ ID NO: 31), more preferably about 20 to 100 adenosine nucleotides (SEQ ID NO: 32) or even more preferably about 40 to 80 adenosine nucleotides (SEQ ID NO: 33).
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may contain, especially if the RNA is in the form of a mRNA, a poly-C tail on the 3′ terminus of typically about 10 to 200 cytosine nucleotides (SEQ ID NO: 34), preferably about 10 to 100 cytosine nucleotides (SEQ ID NO: 35), more preferably about 20 to 70 cytosine nucleotides (SEQ ID NO: 36) or even more preferably about 20 to 60 (SEQ ID NO: 37) or even 10 to 40 cytosine nucleotides (SEQ ID NO: 38).
  • the at least one RNA of the active (immunostimulatory) composition of the present invention may be modified, and thus stabilized, especially if the RNA is in the form of a mRNA, by modifying the G/C content of the RNA, preferably of the coding region of the at least one RNA.
  • the G/C content of the coding region of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is modified, particularly increased, compared to the G/C content of the coding region of its particular wild-type (m)RNA, i.e. the unmodified (m)RNA.
  • the encoded amino acid sequence of the at least one (m)RNA is preferably not modified compared to the coded amino acid sequence of the particular wild-type (m)RNA.
  • This modification of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is based on the fact that the sequence of any (m)RNA region to be translated is important for efficient translation of that (m)RNA.
  • the composition and the sequence of various nucleotides is important.
  • sequences having an increased G (guanosine)/C (cytosine) content are more stable than sequences having an increased A (adenosine)/U (uracil) content.
  • the codons of the (m)RNA are therefore varied compared to its wild-type (m)RNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides.
  • the most favorable codons for the stability can be determined (so-called alternative codon usage).
  • the amino acid to be encoded by the at least one (m)RNA there are various possibilities for modification of the at least one (m)RNA sequence, compared to its wild-type sequence.
  • amino acids which are encoded by codons which contain exclusively G or C nucleotides no modification of the codon is necessary.
  • the codons for Pro (CCC or CCG), Arg (CGC or CGG), Ala (GCC or GCG) and Gly (GGC or GGG) require no modification, since no A or U is present.
  • codons which contain A and/or U nucleotides can be modified by substitution of other codons which code for the same amino acids but contain no A and/or U. Examples of these are:
  • the codons for Pro can be modified from CCU or CCA to CCC or CCG;
  • codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG;
  • the codons for Ala can be modified from GCU or GCA to GCC or GCG;
  • the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • the codons for Phe can be modified from UUU to UUC;
  • the codons for Leu can be modified from UUA, UUG, CUU or CUA to CUC or CUG;
  • the codons for Ser can be modified from UCU or UCA or AGU to UCC, UCG or AGC;
  • the codon for Tyr can be modified from UAU to UAC;
  • the codon for Cys can be modified from UGU to UGC;
  • the codon for His can be modified from CAU to CAC;
  • the codon for Gln can be modified from CAA to CAG;
  • the codons for Ile can be modified from AUU or AUA to AUC;
  • codons for Thr can be modified from ACU or ACA to ACC or ACG;
  • the codon for Asn can be modified from AAU to AAC;
  • the codon for Lys can be modified from AAA to AAG;
  • the codons for Val can be modified from GUU or GUA to GUC or GUG;
  • the codon for Asp can be modified from GAU to GAC;
  • the codon for Glu can be modified from GAA to GAG;
  • the stop codon UAA can be modified to UAG or UGA.
  • substitutions listed above can be used either individually or in all possible combinations to increase the G/C content of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention compared to its particular wild-type (m)RNA (i.e. the original sequence).
  • all codons for Thr occurring in the wild-type sequence can be modified to ACC (or ACG).
  • combinations of the above substitution possibilities are used:
  • the G/C content of the coding region of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the coded region of the wild-type (m)RNA which codes for an antigen, antigenic protein or antigenic peptide as deinined herein or its fragment or variant thereof.
  • the G/C content of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention to the maximum (i.e. 100% of the substitutable codons), in particular in the region coding for a protein, compared to the wild-type sequence.
  • a further preferred modification of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells.
  • the corresponding modified at least one (m)RNA sequence is translated to a significantly poorer degree than in the case where codons coding for relatively “frequent” tRNAs are present.
  • the region which codes for the adjuvant protein is modified compared to the corresponding region of the wild-type (m)RNA such that at least one codon of the wild-type sequence which codes for a tRNA which is relatively rare in the cell is exchanged for a codon which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA.
  • the sequences of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is modified such that codons for which frequently occurring tRNAs are available are inserted.
  • the sequential G/C content which is increased, in particular maximized, in the modified at least one (m)RNA of the active (immunostimulatory) composition of the present invention with the “frequent” codons without modifying the amino acid sequence of the protein encoded by the coding region of the (m)RNA.
  • This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) at least one (m)RNA of the active (immunostimulatory) composition of the present invention.
  • a modified at least one (m)RNA of the active (immunostimulatory) composition of the present invention as described above can be carried out using the computer program explained in WO 02/098443—the disclosure content of which is included in its full scope in the present invention.
  • the nucleotide sequence of any desired (m)RNA can be modified with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence coded by the modified at least one (m)RNA preferably not being modified compared to the non-modified sequence.
  • the A/U content in the environment of the ribosome binding site of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is increased compared to the A/U content in the environment of the ribosome binding site of its particular wild-type (m)RNA.
  • This modification increases the efficiency of ribosome binding to the at least one (m)RNA.
  • An effective binding of the ribosomes to the ribosome binding site (Kozak sequence: GCCGCCACCAUGG (SEQ ID NO: 27), the AUG forms the start codon) in turn has the effect of an efficient translation of the at least one (m)RNA.
  • the at least one (m)RNA of the active (immunostimulatory) composition of the present invention may be modified with respect to potentially destabilizing sequence elements.
  • the coding region and/or the 5′ and/or 3′ untranslated region of this at least one (m)RNA may be modified compared to the particular wild-type (m)RNA such that is contains no destabilizing sequence elements, the coded amino acid sequence of the modified at least one (m)RNA preferably not being modified compared to its particular wild-type (m)RNA.
  • DSE destabilizing sequence elements
  • one or more such modifications compared to the corresponding region of the wild-type (m)RNA can therefore be carried out, so that no or substantially no destabilizing sequence elements are contained there.
  • DSE present in the untranslated regions (3′- and/or 5′-UTR) can also be eliminated from the at least one (m)RNA of the active (immunostimulatory) composition of the present invention by such modifications.
  • Such destabilizing sequences are e.g. AU-rich sequences (AURES), which occur in 3′-UTR sections of numerous unstable RNAs (Caput et al., Proc. Natl. Acad. Sci. USA 1986, 83: 1670 to 1674).
  • AURES AU-rich sequences
  • the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is therefore preferably modified compared to the wild-type (m)RNA such that the at least one (m)RNA contains no such destabilizing sequences.
  • sequence motifs which are recognized by possible endonucleases, e.g.
  • sequence GAACAAG which is contained in the 3′-UTR segment of the gene which codes for the transferrin receptor (Binder et al., EMBO J. 1994, 13: 1969 to 1980).
  • sequence motifs are also preferably removed in the at least one (m)RNA of the active (immunostimulatory) composition of the present invention.
  • the at least one (m)RNA of the active (immunostimulatory) composition of the present invention has, in a modified form, at least one IRES as defined above and/or at least one 5′ and/or 3′ stabilizing sequence, in a modified form, e.g. to enhance ribosome binding or to allow expression of different encoded antigens located on an at least one (bi- or even multicistronic) RNA of the active (immunostimulatory) composition of the present invention.
  • the at least one (m)RNA of the active (immunostimulatory) composition of the present invention furthermore preferably has at least one 5′ and/or 3′ stabilizing sequence.
  • These stabilizing sequences in the 5′ and/or 3′ untranslated regions have the effect of increasing the half-life of the at least one (m)RNA in the cytosol.
  • These stabilizing sequences can have 100% sequence homology to naturally occurring sequences which occur in viruses, bacteria and eukaryotes, but can also be partly or completely synthetic.
  • the untranslated sequences (UTR) of the ⁇ globin gene e.g.
  • stabilizing sequences which can be used in the present invention for a stabilized RNA.
  • Another example of a stabilizing sequence has the general formula (C/U)CCANxCCC(U/A)PyxUC(C/U)CC (SEQ ID NO: 28), which is contained in the 3′UTR of the very stable RNA which codes for aglobin, ⁇ (I)-collagen, 15-lipoxygenase or for tyrosine hydroxylase (cf. Holcik et al., Proc. Natl. Acad. Sci. USA 1997, 94: 2410 to 2414).
  • the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is therefore preferably present as globin UTR (untranslated regions)-stabilized RNA, in particular as ⁇ globin UTR-stabilized RNA.
  • substitutions, additions or eliminations of bases are preferably carried out with the at least one RNA of the active (immunostimulatory) composition of the present invention, using a DNA matrix for preparation of the at least one RNA of the active (immunostimulatory) composition of the present invention by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation strategy (see e.g. Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 3rd ed., Cold Spring Harbor, N.Y., 2001). In such a process, for preparation of the at least one (m)RNA, a corresponding DNA molecule may be transcribed in vitro.
  • This DNA matrix preferably comprises a suitable promoter, e.g. a T7 or SP6 promoter, for in vitro transcription, which is followed by the desired nucleotide sequence for the at least one RNA to be prepared and a termination signal for in vitro transcription.
  • the DNA molecule which forms the matrix of an at least one RNA of interest, may be prepared by fermentative proliferation and subsequent isolation as part of a plasmid which can be replicated in bacteria. Plasmids which may be mentioned as suitable for the present invention are e.g. the plasmids pT7Ts (GENBANK ® accession number U26404; Lai et al., Development 1995, 121: 2349 to 2360), pGEM® series, e.g.
  • pGEM®-1 GenBANK accession number X65300; from Promega
  • pSP64 GenBANK® accession number X65327
  • Mezei and Storts Purification of PCR Products, in: Griffin and Griffin (ed.), PCR Technology: Current Innovation, CRC Press, Boca Raton, Fla., 2001.
  • the stabilization of the at least one RNA of the active (immunostimulatory) composition of the present invention can likewise by carried out by associating or complexing the at least one RNA with, or binding it to, a cationic compound, in particular a polycationic compound, for example a (poly)cationic peptide or protein.
  • a cationic compound in particular a polycationic compound, for example a (poly)cationic peptide or protein.
  • a cationic compound in particular a polycationic compound, for example a (poly)cationic peptide or protein.
  • protamine, nucleoline, spermin or spermidine as the polycationic, nucleic-acid-binding protein to the RNA is particularly effective.
  • other cationic peptides or proteins such as poly-L-lysine or histones, is likewise possible.
  • cationic substances which can be used for stabilizing the RNA of the active (immunostimulatory) composition of the present invention include cationic polysaccharides, for example chitosan, polybrene, polyethyleneimine (PEI) or poly-L-lysine (PLL), etc.
  • cationic polysaccharides for example chitosan, polybrene, polyethyleneimine (PEI) or poly-L-lysine (PLL), etc.
  • oligofectamine as a lipid based complexation reagent preferably increases the transfer of the at least one RNA present as a pharmaceutically active component into the cells to be treated or into the organism to be treated. It is also referred to the disclosure herein with regard to the stabilizing effect for the at least one RNA of the active (immunostimulatory) composition of the present invention by complexation, which holds for the stabilization of RNA as well.
  • the at least RNA of the active (immunostimulatory) composition may additionally or alternatively encode a secretory signal peptide.
  • signal peptides are sequences, which typically exhibit a length of about 15 to 30 amino acids and are preferably located at the N-terminus of the encoded peptide, without being limited thereto.
  • Signal peptides as defined herein preferably allow the transport of the antigen, antigenic protein or antigenic peptide as encoded by the at least one RNA of the active (immunostimulatory) composition into a defined cellular association, preferably the cell surface, the endoplasmic reticulum (ER) or the endosomal-lysosomal association.
  • secretory signal peptide sequences as defined herein include, without being limited thereto, signal sequences of classical or non-classical MHC-molecules (e.g. signal sequences of MHC I and II molecules, e.g. of the MHC class I molecule HLA-A*0201), signal sequences of cytokines or immunoglobulines as defined herein, signal sequences of the invariant chain of immunoglobulines or antibodies as defined herein, signal sequences of Lamp1, Tapasin, Erp57, Calretikulin, Calnexin, and further membrane associated proteins or of proteins associated with the endoplasmic reticulum (ER) or the endosomal-lysosomal juxtapos.
  • signal sequences of MHC class I molecule HLA-A*0201 may be used according to the present invention.
  • any of the above modifications may be applied to the at least one RNA of the active (immunostimulatory) composition of the present invention, and further to any (m)RNA as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the respective at least one RNA.
  • a person skilled in the art will be able to take his choice accordingly.
  • the active (immunostimulatory) composition according to the invention may comprise an adjuvant.
  • an adjuvant may be understood as any compound, which is suitable to support administration and delivery of the active (immunostimulatory) composition according to the invention.
  • an adjuvant may, without being bound thereto, initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response.
  • the active (immunostimulatory) composition according to the invention typically initiates an adaptive immune response due to the at least two antigens encoded by the at least one RNA contained in the inventive active (immunostimulatory) composition.
  • the active (immunostimulatory) composition according to the invention may generate an (supportive) innate immune response due to addition of an adjuvant as defined herein to the active (immunostimulatory) composition according to the invention.
  • an adjuvant may be selected from any adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an immune response in a mammal.
  • the adjuvant may be selected from the group consisting of, without being limited thereto, TDM, MDP, muramyl dipeptide, pluronics, alum solution, aluminium hydroxide, ADJUMERTM (polyphosphazene); aluminium phosphate gel; glucans from algae; algammulin; aluminium hydroxide gel (alum); highly protein-adsorbing aluminium hydroxide gel; low viscosity aluminium hydroxide gel; AF or SPT (emulsion of squalane (5%), Tween 80 (0.2%), Pluronic L121 (1.25%), phosphate-buffered saline, pH 7.4); AVRIDINETM (propanediamine); BAY R1005TM ((N-(2-deoxy-2-L-leucylamino-b-D-glucopyranosyl)-N-octadecyl-dodecanoyl-amide hydroacetate); CALCITRIOLTM (1-
  • Suitable adjuvants may also be selected from cationic or polycationic compounds wherein the adjuvant is preferably prepared upon complexing the at least one RNA of the inventive active (immmunostimulatory composition) with the cationic or polycationic compound. Association or complexing the RNA of the active (immunostimulatory) composition with cationic or polycationic compounds as defined herein preferably provides adjuvant properties and confers a stabilizing effect to the at least one RNA of the active (immunostimulatory) composition.
  • cationic or polycationic compounds are selected from cationic or polycationic peptides or proteins, including protamine, nucleoline, spermin or spermidine, or other cationic peptides or proteins, such as poly-L-lysine (PLL), poly-arginine, basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, Tat, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs, PpT620, prolin-rich peptides, arginine-rich peptides, lysine-rich peptides, MPG-peptide(s), Pep-1, L-oligomers, Calcitonin peptide(s), Antennapedia-derived peptides (particularly from Dros
  • cationic or polycationic compounds may include cationic polysaccharides, for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g.
  • cationic polysaccharides for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g.
  • PEI polyethyleneimine
  • DOTMA [1-(2,3-sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride
  • DMRIE di-C14-amidine
  • DOTIM DOTIM
  • SAINT DC-Chol
  • BGTC CTAP
  • DOPC DODAP
  • DOPE Dioleyl phosphatidylethanol-amine
  • DOSPA DODAB
  • DOIC DOIC
  • DMEPC DOGS: Dioctadecylamidoglicylspermin
  • DIMRI Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide
  • DOTAP dioleoyloxy-3-(trimethylammonio)propane
  • DC-6-14 0,0-ditetradecanoyl-N-( ⁇ -trimethylammonioacetyl)diethanolamine chloride
  • CLIP1 rac-[(2,3-dioctadecyloxypropyl)(2-hydroxyethyl)]-d
  • modified polyaminoacids such as 13-aminoacid-polymers or reversed polyamides, etc.
  • modified polyethylenes such as PVP (poly(N-ethyl-4-vinylpyridinium bromide)), etc.
  • modified acrylates such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), etc.
  • modified Amidoamines such as pAMAM (poly(amidoamine)), etc.
  • modified polybetaaminoester (PBAE) such as diamine end modified 1,4 butanediol diacrylate-co-5-amino-1-pentanol polymers, etc.
  • dendrimers such as polypropylamine dendrimers or pAMAM based dendrimers, etc.
  • polyimine(s) such as PEI: poly(ethyleneimine), poly(propyleneimine), etc.
  • polyallylamine sugar backbone based polymers,
  • oligoarginines in this context are e.g. Arg 7 (SEQ ID NO: 42), Arg 8 (SEQ ID NO: 43), Arg 9 (SEQ ID NO: 44), Arg 7 (SEQ ID NO: 42), H 3 R 9 (SEQ ID NO: 45), R 9 H 3 (SEQ ID NO: 46), H 3 R 9 H 3 (SEQ ID NO: 47), YSSR 9 SSY (SEQ ID NO: 48), (RKH) 4 (SEQ ID NO: 49), Y(RKH) 2 R (SEQ ID NO: 50), etc.
  • Suitable adjuvants may furthermore be selected from nucleic acids having the formula (II): G l X m G n , (SEQ ID NO: 51) wherein: G is guanosine, uracil or an analogue of guanosine or uracil;
  • the present invention may furthermore provide a vaccine containing the active (immunostimulatory) composition according to the invention.
  • the inventive vaccine may additionally contain a pharmaceutically acceptable carrier and/or further auxiliary substances and additives and/or adjuvants.
  • the antigens encoded by the at least one RNA of the active (immunostimulatory) composition, contained in the inventive vaccine are selected from the above mentioned groups or subgroups.
  • the protein antigens are selected from any of the antigens of the following subgroup comprising NY-ESO1 [accession number NM_001327], hTERT [accession number NM_198253], survivin [accession number AF077350], 5T4 [accession number NM_006670] and WT1 [accession number NM_000378], and/or from any of the antigens of the following subgroup comprising MAGE-C1 and MAGE-C2, as defined herein, and/or from any of the antigens of the following subgroup comprising MAGE-A2 and MAGE-A3, as defined herein.
  • the inventive vaccine typically comprises a safe and effective amount of the at least one RNA of the active (immunostimulatory) composition as defined above encoding at least two antigens as defined above, more preferably encoding at least two antigens selected from any of the above goups or subgroups, most preferably in any of the indicated combinations.
  • safety and effective amount means an amount of the at least one RNA of the active (immunostimulatory) composition in the vaccine as defined above, that is sufficient to significantly induce a positive modification of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition to be treated, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma.
  • NSCLC non-small-cell lung cancer
  • a “safe and effective amount” is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment.
  • the expression “safe and effective amount” preferably means an amount of the RNA (and thus of the encoded at least two antigens) that is suitable for stimulating the adaptive immune system in such a manner that no excessive or damaging immune reactions are achieved but, preferably, also no such immune reactions below a measurable level.
  • a “safe and effective amount” of the at least one RNA of the active (immunostimulatory) composition in the vaccine as defined above may furthermore be selected in dependence of the type of RNA, e.g.
  • a “safe and effective amount” of the at least one RNA of the active (immunostimulatory) composition as defined above, which is contained in the inventive vaccine, will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying doctor.
  • the vaccine according to the invention can be used according to the invention for human and also for veterinary medical purposes, as a pharmaceutical composition or as a vaccine.
  • the vaccine according to the invention typically contains a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier as used herein preferably includes the liquid or non-liquid basis of the inventive vaccine. If the inventive vaccine is provided in liquid form, the carrier will typically be pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate-, citrate-buffered solutions, etc..
  • water or preferably a buffer preferably an aqueous buffer
  • a sodium salt preferably at least 50 mM of a sodium salt
  • a calcium salt preferably at least 0,01 mM of a calcium salt
  • optionally a potassium salt preferably at least 3 mM of a potassium salt.
  • the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc.
  • examples of sodium salts include e.g.
  • examples of the optional potassium salts include e.g. KCl, KI, KBr, K 2 CO 3 , KHCO 3 , K 2 SO 4
  • examples of calcium salts include e.g. CaCl 2 , CaI 2 , CaBr 2 , CaCO 3 , CaSO 4 , Ca(OH) 2 .
  • organic anions of the aforementioned cations may be contained in the buffer.
  • the buffer suitable for injection purposes as defined above may contain salts selected from sodium chloride (NaCl), calcium chloride (CaCl 2 ) and optionally potassium chloride (KCl), wherein further anions may be present additional to the chlorides.
  • CaCl 2 can also be replaced by another salt like KCl.
  • the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCl), at least 3 mM potassium chloride (KCl) and at least 0,01 mM calcium chloride (CaCl 2 ).
  • the injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e.
  • the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects.
  • Reference media are e.g. in “in vivo” methods occurring liquids such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in “in vitro” methods, such as common buffers or liquids. Such common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis.
  • compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person.
  • the term “compatible” as used herein means that the constituents of the inventive vaccine are capable of being mixed with the at least one RNA of the active (immunostimulatory) composition, encoding at least two antigens as defined above, in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the inventive vaccine under typical use conditions.
  • Pharmaceutically acceptable carriers, fillers and diluents must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated.
  • Some examples of compounds which can be used as pharmaceutically acceptable carriers, fillers or constituents thereof are sugars, such as, for example, lactose, glucose and sucrose; starches, such as, for example, corn starch or potato starch; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulfate; vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid.
  • sugars such as, for example, lactose, glucose and sucrose
  • starches such as, for example, corn star
  • the choice of a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered.
  • the inventive vaccine can be administered, for example, systemically or locally.
  • Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes.
  • Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial, intrapulmonal, intracardial, and sublingual injections.
  • vaccines may be administered by an intradermal, subcutaneous, or intramuscular route.
  • Compositions/vaccines are therefore preferably formulated in liquid or solid form.
  • the suitable amount of the inventive vaccine to be administered can be determined by routine experiments with animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non-human primate models.
  • Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4.
  • Suitable carriers for injection include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices.
  • Suitable pharmaceutically acceptable carriers for topical application include those which are suitable for use in lotions, creams, gels and the like.
  • tablettes and the like are the preferred unit dose form.
  • pharmaceutically acceptable carriers for the preparation of unit dose forms which can be used for oral administration are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art.
  • the inventive vaccine can additionally contain one or more auxiliary substances in order to further increase the immunogenicity.
  • auxiliary substances various mechanisms can come into consideration in this respect. For example, compounds that permit the maturation of dendritic cells (DCs), for example lipopolysaccharides, TNF-alpha or CD40 ligand, form a first class of suitable auxiliary substances.
  • DCs dendritic cells
  • TNF-alpha or CD40 ligand form a first class of suitable auxiliary substances.
  • auxiliary substance any agent that influences the immune system in the manner of a “danger signal” (LPS, GP96, etc.) or cytokines, such as GM-CFS, which allow an immune response produced by the immune-stimulating adjuvant according to the invention to be enhanced and/or influenced in a targeted manner.
  • a “danger signal” LPS, GP96, etc.
  • cytokines such as GM-CFS
  • auxiliary substances are cytokines, such as monokines, lymphokines, interleukins or chemokines, that—additional to induction of the adaptive immune response by the encoded at least two antigens—promote the innate immune response, such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, INF-alpha, IFN-beta, INF-gamma, GM-CSF, G-CSF, M-CSF, LT-beta or TNF-alpha, growth factors
  • emulsifiers such as, for example, Tween®
  • wetting agents such as, for example, sodium lauryl sulfate
  • colouring agents such as, for example, sodium lauryl sulfate
  • taste-imparting agents pharmaceutical carriers
  • tablet-forming agents such as, for example, stabilizers; antioxidants; preservatives.
  • the inventive vaccine can also additionally contain any further compound, which is known to be immune-stimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLRS, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLRS, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 or TLR13.
  • any further compound which is known to be immune-stimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLRS, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 or TLR13.
  • CpG nucleic acids in particular CpG-RNA or CpG-DNA.
  • a CpG-RNA or CpG-DNA can be a single-stranded CpG-DNA (ss CpG-DNA), a double-stranded CpG-DNA (dsDNA), a single-stranded CpG-RNA (ss CpG-RNA) or a double-stranded CpG-RNA (ds CpG-RNA).
  • the CpG nucleic acid is preferably in the form of CpG-RNA, more preferably in the form of single-stranded CpG-RNA (ss CpG-RNA).
  • the CpG nucleic acid preferably contains at least one or more (mitogenic) cytosine/guanine dinucleotide sequence(s) (CpG motif(s)).
  • CpG motif(s) cytosine/guanine dinucleotide sequence(s)
  • at least one CpG motif contained in these sequences that is to say the C (cytosine) and the G (guanine) of the CpG motif, is unmethylated. All further cytosines or guanines optionally contained in these sequences can be either methylated or unmethylated.
  • the C (cytosine) and the G (guanine) of the CpG motif can also be present in methylated form.
  • the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein may be used (for the preparation of a vaccine according to the present invention) for the treatment of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma.
  • NSCLC non-small-cell lung cancer
  • the inventive vaccine or the at least one RNA encoding at least two (preferably) different antigens as defined herein may be used for the treatment of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma.
  • NSCLC non-small-cell lung cancer
  • lung cancer preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, by administering to a patient in need thereof a pharmaceutically effective amount of an inventive vaccine, or a pharmaceutically effective amount of an inventive active (immunostimulatory) composition.
  • NSCLC non-small-cell lung cancer
  • Such a method typically comprises an optional first step of preparing the inventive active (immunostimulatory) composition, or the inventive vaccine, and a second step, comprising administering (a pharmaceutically effective amount of) said inventive active (immunostimulatory) composition or said inventive vaccine to a patient in need thereof.
  • a patient in need thereof will be typically selected from any mammal.
  • a mammal is preferably selected from the group comprising, without being limited thereto, e.g.
  • lung cancer preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma or a condition related thereto.
  • NSCLC non-small-cell lung cancer
  • the invention relates also to the use of the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein (for the preparation of an inventive vaccine), preferably for eliciting an immune response in a mammal, preferably for the treatment of lung cancer, more preferably for the treatment of a non-small-cell lung cancer (NSCLC) related condition as defined herein.
  • inventive active immunological active
  • the at least one RNA encoding at least two (preferably) different antigens as defined herein for the preparation of an inventive vaccine
  • the invention also relates also to the use of the inventive vaccine per se or the at least one RNA encoding at least two (preferably) different antigens as defined herein for eliciting an adaptive immune response in a mammal, preferably for the treatment of lung cancer, more preferably of a non-small-cell lung cancer (NSCLC) related condition as defined herein.
  • NSCLC non-small-cell lung cancer
  • Prevention or treatment of lung cancer in a patient in need thereof, preferably of a non-small-cell lung cancer (NSCLC) related condition as defined herein, may be carried out by administering the inventive active (immunostimulatory) composition and/or the inventive vaccine at once or in a time staggered manner, e.g. as a kit of parts, each part containing at least one preferably different antigen.
  • the inventive active (immunostimulatory) composition and/or the inventive vaccine at once or in a time staggered manner, e.g. as a kit of parts, each part containing at least one preferably different antigen.
  • any of the administration routes may be used as defined above.
  • NSCLC non-small-cell lung cancer
  • Administering of the inventive active (immunostimulatory) composition and/or the inventive vaccine may then occur prior, concurrent and/or subsequent to administering another inventive inventive active (immunostimulatory) composition and/or inventive vaccine as defined herein which may contain another combination of RNAs encoding different antigens, wherein each antigen encoded by the at least one RNA of the inventive active (immunostimulatory) composition may preferably be suitable for the therapy of lung cancer, more preferably for the treatment of a non-small-cell lung cancer (NSCLC) related condition as defined herein.
  • a therapy as defined herein may also comprise the modulation of a disease associated to lung cancer, preferably a disease associated to non-small-cell lung cancer (NSCLC) as defined herein.
  • the present invention furthermore comprises the use of the active (immunostimulatory) composition (for the preparation of an (inventive) vaccine) for modulating, preferably to induce or enhance, an immune response in a mammal as defined above, more preferably to support the treatment of lung cancer, especially NSCLC as defined herein.
  • support of the treatment of lung cancer may be any combination of a conventional cancer therapy for lung cancer, especially for NSCLC as defined herein, such as radiation therapy, chemotherapy, proton therapy, hormonal therapy, antibody therapy, adjuvant therapies, therapies including other vaccines than an inventive vaccine, therapies including kinase inhibitors or small nucleotides, etc., or some combination of these, and a therapy using the inventive active (immunostimulatory) composition or the inventive vaccine as defined herein.
  • Support of the treatment of lung cancer, especially NSCLC as defined herein may be also envisaged in any of the other embodiments defined herein.
  • a time staggered treatment may be e.g. administration of the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein or the inventive vaccine prior, concurrent and/or subsequent to a therapy of lung cancer, especially NSCLC, e.g. by administration of the inventive active (immunostimulatory) composition or vaccine prior, concurrent and/or subsequent to a therapy or an administration of a therapeutic suitable for the treatment of lung cancer, especially of NSCLC as defined herein.
  • Such time staggered treatment may be carried out using e.g. a kit, preferably a kit of parts as defined below.
  • Time staggered treatment may additionally or alternatively also comprise an administration of the inventive active (immunostimulatory) composition or vaccine, preferably of the at least one RNA encoding at least two (preferably different) antigens as defined above, in a form, wherein the at least one RNA encoding at least two (preferably different) antigens as defined above, preferably forming part of the inventive active (immunostimulatory) composition or vaccine, is administered parallel, prior or subsequent to another at least one RNA encoding at least two (preferably different) antigens as defined above, preferably forming part of the same inventive active (immunostimulatory) composition or vaccine.
  • the administration occurs within an hour, more preferably within 30 minutes, even more preferably within 15, 10, 5, 4, 3, or 2 minutes or even within 1 minute.
  • time staggered treatment may be carried out using e.g. a kit, preferably a kit of parts as defined below.
  • kits particularly kits of parts, comprising the active inventive (immunostimulatory) composition, and/or the inventive vaccine, and optionally technical instructions with information on the administration and dosage of the inventive active (immunostimulatory) composition and/or the inventive vaccine.
  • the technical instructions may contain information about administration and dosage of the inventive active (immunostimulatory) composition, and/or the inventive vaccine.
  • kits preferably kits of parts, may applied e.g. for any of the above mentioned applications or uses, preferably for the use of at least one inventive active (immunostimulatory) composition (for the preparation of an inventive vaccine) for the treatment of lung cancer, especially of NSCLC as defined herein.
  • kits may also be applied for the use of at least one inventive active (immunostimulatory) composition (for the preparation of an inventive vaccine) for the treatment of lung cancer, preferably NSCLC as defined herein, wherein the inventive active (immunostimulatory) composition) and/or the vaccine due to the encoded at least two antigens may be capable to induce or enhance an immune response in a mammal as defined above.
  • inventive kits may further be applied for the use of at least one inventive active (immunostimulatory) composition, (for the preparation of an inventive vaccine) for modulating, preferably for eliciting, e.g. to induce or enhance, an immune response in a mammal as defined above, and preferably to support treatment of lung cancer, especially of NSCLC.
  • Kits of parts may contain one or more identical or different active inventive (immunostimulatory) compositions and/or one or more identical or different inventive vaccines in different parts of the kit.
  • Kits of parts may also contain an (e.g. one) active inventive (immunostimulatory) composition, an (e.g. one) inventive vaccine and/or the at least one RNA encoding at least one antigen as defined above in different parts of the kit, e.g. each part of the kit containing at least one RNA encoding a preferably different antigen.
  • Kits of parts may be used, e.g. when a time staggered treatment is envisaged, e.g.
  • kits of parts may also be used when a separated formulation or administration of the different antigens of the inventive active (immunostimulatory) composition (i.e. in parts) is envisaged or necessary (e.g. for technical reasons), but e.g. a combined presence of the different antigens in vivo is still to be achieved.
  • kits of parts as a special form of kits are envisaged, wherein each part of the kit contains at least one preferably different antigen as defined above, all parts of the kit of parts preferably forming the active inventive (immunostimulatory) composition or the inventive vaccine as defined herein.
  • Such specific kits of parts may particularly be suitable, e.g. if different antigens are formulated separately as different parts of the kits, but are then administered at once together or in a time staggered manner to the mammal in need thereof. In the latter case administration of all of the different parts of such a kit typically occurs within a short time limit, such that all antigens are present in the mammal at about the same time subsequent to administration of the last part of the kit. Any of the above kits may be used in a treatment as defined above.
  • the present invention provides an active (immunostimulatory) composition for the treatment of lung cancer, particularly of non-small lung cancer (NSCLC), wherein the composition comprises at least one RNA, preferably a mRNA, encoding at least two (preferably different) antigens capable of eliciting an (adaptive) immune response in a mammal wherein the antigens are selected from the group consisting of hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1, or MAGE-C2.
  • an active (immunostimulatory) composition allows efficient treatment of lung cancer, particularly of non-small lung cancer (NSCLC), or supplementary treatment when using conventional therapies.
  • RNA as used in the inventive active (immunostimulatory) composition has additional considerable advantages over DNA expression systems e.g. in immune response, immunization or vaccination. These advantages include, inter alfa, that RNA introduced into a cell is not integrated into the genome. This avoids the risk of mutation of this gene, which otherwise may be completely or partially inactivated or give rise to misinformation. It further avoids other risks of using DNA as an agent to induce an immune response (e.g. as a vaccine) such as the induction of pathogenic anti-DNA antibodies in the patient into whom the foreign DNA has been introduced, so bringing about a (possibly fatal) immune response. In contrast, no anti-RNA antibodies have yet been detected.
  • an immune response e.g. as a vaccine
  • FIG. 1 depicts a RNA sequence (SEQ ID NO: 1) (starting sequence based on the wildtype) encoding MUC1 (HsMUC1-5xVNTR (The wildtype sequence normally shows 40 tandem repeats. These were—for cloning reasons—reduced to 5 tandem repeats). GC content: 61.27%; length: 1668 bp).
  • FIG. 3 depicts a RNA sequence (SEQ ID NO: 3) (starting sequence based on the wildtype) encoding 5T4 (Hs5T4 (trophoblast glycoprotein TPBG); GC content: 61.60%; length: 1263 bp.
  • FIG. 5 depicts a RNA sequence (SEQ ID NO: 5) (starting sequence based on the wildtype) encoding Her-2/neu (HsHer2/neu (v-erb-b2 erythroblastic leukemia viral oncogene homolog 2)); GC content: 60.78% ; length: 3768 bp.
  • FIG. 7 depicts a RNA sequence (SEQ ID NO: 7) (starting sequence based on the wildtype) encoding hTERT (HsTERT (telomerase reverse transcriptase); GC Content: 66.08% ; Length: 3399 bp.
  • FIG. 9 depicts a RNA sequence (SEQ ID NO: 9) (starting sequence based on the wildtype) encoding WT1 (HsWT1 (Wilms tumor 1)); GC Content: 61.78% ; Length: 1554 bp.
  • FIGS. 10A-D depicts a RNA sequence (SEQ ID NO: 10) encoding WT1 (HsWT1 (Wilms tumor 1)) showing a sequence with a reduced GC content in region 325-408 of said sequence compared to the corresponding region of the wildtype sequence.
  • FIG. 12 depicts a RNA sequence (SEQ ID NO: 12) (starting sequence based on the wildtype) encoding CEA (CEA (carcinoembryonic antigen) HsCEACAMS); GC Content: 52.20% ; Length: 2109 bp.
  • CEA CEACAMS GC
  • FIG. 14 depicts a RNA sequence (SEQ ID NO: 14) (starting sequence based on the wildtype) encoding MAGE-A2 (HsMAGE-A2 (melanoma antigen family A, 2) HsMAGE-A2B).
  • GC Content 55.87% ; Length: 945 bp.
  • FIG. 16 depicts a RNA sequence (SEQ ID NO: 16) (starting sequence based on the wildtype) encoding MAGE-A3 (MAGE-A3 (melanoma antigen family A, 3) MAGE-A3) GC Content: 56.30% ; Length: 945 bp.
  • FIG. 18 depicts a RNA sequence (SEQ ID NO: 18) (starting sequence based on the wildtype) encoding Survivin (Survivin (baculoviral IAP repeat-containing 5, BIRC5) HsSurvivin(wt)); GC Content: 52.68% ; Length: 429 bp.
  • FIG. 20 depicts a RNA sequence (SEQ ID NO: 20) (starting sequence based on the wildtype) encoding NY-ESO-1 (Homo sapiens NY-ESO-1 (NY-ESO-1(wt)); GC-Content 67.4%.
  • FIG. 21 depicts a (GC) stabilized RNA sequence (SEQ ID NO: 21) encoding NY-ESO-1 (NY-E SO-1(GC), GC-Content 79.56%, (already known GC-Enrichment); Difference to wt ( FIG. 20 (SEQ ID NO: 20)): 112/543 Bases, 20.63%.
  • FIG. 22 depicts a RNA sequence (SEQ ID NO: 22) (starting sequence based on the wildtype) encoding MAGE-C1 (HsMAGEC1 (melanoma antigen family C, 1) HsMAGEC1(wt)) GC Content: 51.86%; Length: 3429 bp.
  • FIG. 24 depicts a (GC) stabilized RNA sequence (SEQ ID NO: 24) encoding a truncated MAGE-C1 (HsMAGEC1(GC), 1. GC-maximized, 2. Codon usage).
  • a (GC) stabilized RNA sequence SEQ ID NO: 24
  • HsMAGEC1(GC) truncated MAGE-C1
  • FIG. 22 SEQ ID NO: 22
  • AGT initial start codon
  • FIG. 23 SEQ ID NO: 23
  • FIG. 25 depicts a RNA sequence (SEQ ID NO: 25) (starting sequence based on the wildtype) encoding MAGE-C2 (HsMAGE-C2 (melanoma antigen family C, 2)HsMAGE-C2); GC Content: 50.81% ; Length: 1122 bp.
  • FIG. 27 shows the presence of IgG1 antibodies specific for the tumour antigen NY-ESO-1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 28 shows the presence of IgG2a antibodies specific for the tumour antigen NY-ESO-1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 29 shows the presence of IgG1 antibodies specific for the tumour antigen MAGE-C1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • NSCLC related antigen NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4
  • FIG. 30 shows the presence of IgG2a antibodies specific for the tumour antigen MAGE-C1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 31 shows the presence of IgG1 antibodies specific for the tumour antigen MAGE-C2 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • NSCLC related antigen NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4
  • FIG. 32 shows the presence of IgG2a antibodies specific for the tumour antigen MAGE-C2 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • NSCLC related antigen NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4
  • FIG. 33 shows the induction of antigen-specific T-lymphocytes directed against the tumour antigen 5T4 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1,MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • NSCLC related antigen NY-ESO-1,MAGE-C1, MAGE-C2, Survivin and 5T4
  • FIG. 34 shows the induction of antigen-specific T-lymphocytes directed against the tumour antigen NY-ESO-1 iin mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • RNA sequences were prepared by in vitro transcription. Therefore, the recombinant plasmid DNA was linearized and subsequently in vitro transcribed using the T7 RNA polymerase. The DNA template was then degraded by DNase I digestion, and the RNA was recovered by LiCl precipitation and further cleaned by HPLC extraction (PUREMessenger®, CureVac GmbH, Tubingen, Germany).
  • the RNA obtained by in vitro transcription was preferably complexed, more preferably with protamine upon mixing the RNA with protamine.
  • mice For vaccination the RNA obtained by the in vitro transcription experiment as shown above (see Experiment 2) was transfected into mice (Mice: C57 BL/6), preferably when complexed with protamine (see Experiment 3). Transfection occurred in different groups, wherein 5 mice (C57 BL/6) per group were immunized intradermally 8 times within 3 weeks with the inventive mRNA cocktail, i.e. a mixture of mRNA complexed with protamine, wherein the RNA codes for at least two of the antigens hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1, or MAGE-C2.
  • inventive mRNA cocktail i.e. a mixture of mRNA complexed with protamine
  • B-cell immune response Detection of an antigen-specific immune response (B-cell immune response) was carried out by detecting antigen-specific antibodies. Therefore, blood samples were taken from the vaccinated mice one week after the last vaccination and sera were prepared. MaxiSorb plates (Nalgene Nunc International) were coated with the antigenic protein as encoded by the mRNA-Cocktail (0.5 ⁇ g/well). After blocking with 1 ⁇ PBS containing 0.05% Tween-20 and 1% BSA the plates were incubated with diluted mouse serum (1:30, 1:90, 1:270, 1:810). Subsequently a biotin-coupled secondary antibody (Anti-mouse-IgG2a Pharmingen) was added.
  • Anti-mouse-IgG2a Pharmingen Anti-mouse-IgG2a Pharmingen
  • the plate was incubated with Horseradish peroxidase-streptavidin and subsequently the conversion of the ABTS substrate (2,2′-azino-bis(3-ethyl-benzthiazoline-6-sulfonic acid) was measured.
  • spleens were removed and the splenocytes were isolated.
  • the splenocytes were restimulated for 7 days in the presence of peptides from the above antigens (peptide library) or coincubated with dendritic cells generated from bone marrow cells of native syngeneic mice, which are electroporated with RNA coding for the antigen.
  • peptide library peptide library
  • dendritic cells generated from bone marrow cells of native syngeneic mice, which are electroporated with RNA coding for the antigen.
  • a coat multiscreen plate (Millipore) was incubated overnight with coating buffer 0.1 M carbonate-bicarbonate buffer pH 9.6, 10.59 g/l Na 2 CO 3 , 8.4 g/l NaHCO 3 ) comprising antibody against INF ⁇ (BD Pharmingen, Heidelberg, Germany). Stimulators and effector cells were incubated together in the plate in the ratio of 1:20 for 24 h. The plate was washed with 1 ⁇ PBS and incubated with a biotin-coupled secondary antibody.
  • the substrate (5-Bromo-4-Cloro-3-Indolyl Phosphate/Nitro Blue Tetrazolium Liquid Substrate System from Sigma Aldrich, Taufmün, Germany) was added to the plate and the conversion of the substrate could be detected visually.
  • inventive active (immunostimulatory) composition comprising a combination of several antigens for the use as a vaccine for the treatment of non-small cell lung cancer (NSCLC) was prepared in the following according to the above disclosure.
  • the exemplary inventive active (immunostimulatory) composition consisted of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4, according to SEQ ID NOs: 4, 19, 21, 24 and 26 (GC-enriched sequences)) formulated with protamine at a mass ratio of 4:1.
  • Vaccination C57BL/6 mice were vaccinated intradermally with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4, according to SEQ ID NOs: 4, 19, 21, 24 and 26 (GC-enriched sequences)) formulated with protamine (64 ⁇ g per antigen per cycle, divided into 4 injections/cycle).
  • Control vaccination was performed using the corresponding total doses of RNA coding for LacZ (control mRNA lacZ).
  • the vaccination comprised three immunization cycles (week 1, 3, and 5).
  • the groups, number of mice and mouse strains are indicated in the following table:
  • mice mRNA vaccine C57BL/6 10 5 for Elispot and 5 for antibody detection in serum by ELISA
  • TMB-substrate was added. The colorimetric reaction was measured at 450 nm using an ELISA reader (Tecan GmbH, Crailsheim, Germany).
  • cytotoxic T-lymphocyte For the detection of cytotoxic T-lymphocyte (CTL) responses the analysis of the secretion of the effector cytokine IFN- ⁇ in response to a specific stimulus can be visualized at a single cell level using the ELISPOT technique.
  • Splenocytes from antigen-vaccinated and control mice were isolated 6 days after last vaccination and then transferred into 96-well ELISPOT plates coated with an anti-IFN- ⁇ capture antibody (10 ⁇ g/ml). The cells were then stimulated for 24 hours at 37° C. either with relevant antigen-derived peptide library or with the HIV-derived library or the solvent of the peptides, DMSO, or incubated in pure medium as a control. All libraries were used at a concentration of 1 ⁇ g/peptide/ml.
  • the cells were washed out of the plate and the IFN- ⁇ secreted by the cells was detected using a biotinylated secondary antibody against murine IFN- ⁇ (1 ⁇ g/ml), followed by streptavidin-AKP. Spots were visualized using BCIP/NBT substrate and counted using an automated ELISPOT reader (Immunospot Analyzer, CTL Analyzers LLC).
  • mice were vaccinated with the mRNA vaccine containing five components as defined above, particularly GC-enriched mRNAs coding for the NSCLC-associated antigens NY-ESO-1, MAGE-C2, MAGE-C1, Survivin and 5T4, (according to SEQ ID NOs: 4, 19, 21, 24 and 26 (GC-enriched sequences)) each formulated separately with the cationic peptide protamine at a mass ratio of 4:1.
  • Control mice were treated with irrelevant RNA coding for LacZ formulated with protamine at the same ratio as the mRNA vaccine.
  • IFN- ⁇ is the main mediator of Th1 responses and secreted by activated CTLs. Therefore the presence of antigen-specific cytotoxic T-cells in splenocytes from vaccinated mice was investigated using the ELISPOT technique. As an antigenic stimulus for splenocytes restricted peptide libraries were used. Because distinct epitopes of the used human antigens for mouse MHC (H-2K b and H-2D b in C57BL/6 mice) are not known, we had to use a hypothetical selection of peptides selected due to potential binding affinity by search of the SYFPEITHI database.
  • the number of IFN- ⁇ spots by splenocytes incubated in medium alone represents the basal activation of the freshly isolated cells. Due to the fact that the basal activation is strongly individual dependent, the background correction was performed individually by subtraction of the number of spots in medium wells from all other values.

Abstract

The present invention relates to an active (immunostimulatory) composition comprising at least one RNA, preferably a mRNA, encoding at least two (preferably different) antigens capable of eliciting an (adaptive) immune response in a mammal. The invention furthermore relates to a vaccine comprising said active (immunostimulatory) composition, and to the use of said active (immunostimulatory) composition (for the preparation of a vaccine) and/or of the vaccine for eliciting an (adaptive) immune response for the treatment of lung cancer, particularly of non-small cell lung cancers (NSCLC), preferably selected from the three main sub-types squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, or of disorders related thereto. Finally, the invention relates to kits, particularly to kits of parts, containing the active (immunostimulatory) composition and/or the vaccine.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. application Ser. No. 15/145,746, filed May 3, 2016, which is a divisional of U.S. application Ser. No. 13/750,664, filed Jan. 25, 2013, now U.S. Pat. No. 9,352,028, which is a continuation of U.S. application Ser. No. 12/682,213, filed Jul. 9, 2010, now abandoned, which is a national phase application under 35 U.S.C. § 371 of International Application No. PCT/EP2008/008503, filed Oct. 8, 2008, which claims priority to International Application No. PCT/EP2007/008770, filed Oct. 9, 2007, the disclosures of which are incorporated herein by reference in their entirety.
  • SEQUENCE LISTING
  • The sequence listing that is contained in the file named “CRVCP0068USD1.txt”, which is 76 KB (as measured in Microsoft Windows®) and was created on May 3, 2016, is filed herewith via EFS-Web and is hereby incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • The present invention relates to an active (immunostimulatory) composition comprising at least one RNA, preferably a mRNA, encoding at least two (preferably different) antigens capable of eliciting an (adaptive) immune response in a mammal. The invention furthermore relates to a vaccine comprising said active (immunostimulatory) composition, and to the use of said active (immunostimulatory) composition (for the preparation of a vaccine) and/or of the vaccine for eliciting an (adaptive) immune response for the treatment of lung cancer, particularly of non-small cell lung cancers (NSCLC), preferably selected from the three main sub-types squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, or of disorders related thereto. Finally, the invention relates to kits, particularly to kits of parts, containing the active (immunostimulatory) composition and/or the vaccine.
  • Of all malignant tumors 25% are bronchial carcinoma (carcinoma of the lung). Worldwide, it is the most common cause of cancer-related death in men and the second most common in women. In Germany it is the third most abundant sort of carcinoma following carcinoma of the prostata and the colorectal carcinoma. It is responsible for 1.3 million deaths worldwide annually. In Central Europe the incidence is approximately 60 per 100.000 inhabitants and the number of newly people diagnosed with lung cancer is steadily on the rise (in Germany currently being at approximately 50.000 per year). When diagosed with lung cancer the avarage overall fife-year survival rate is a mere 5 percent. Nevertheless, the life expectancy of each single patient is wholly dependent on the disease stage (TMN classification) and the subtype of carcinoma (lung cancer) encountered (see below).
  • The main sub-types of lung cancer categorized by the size and appearance of the malignant cells identified under microscope are small cell lung cancer (20%) and non-small cell lung cancer (NSCLC) (80%). This classification, although based on simple histological criteria, has very important implications for clinical management and prognosis of the disease, with small cell lung cancer usually being treated by chemotherapy, while non-small cell lung cancer is mostly subject to surgery as a first-line treatment.
  • The non-small cell lung cancers (NSCLC) are grouped together because their prognosis and management are roughly identical. There are three main sub-types: squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma. Surgery is the mainstay of treatment; however, only a quarter of the patients undergo successful resection, with a recurrence rate of 50%. Therapeutic approaches in advanced disease involve—following surgery—both adjuvant chemotherapy and/or adjuvant radiotherapy, whereas chemotherapy as monotherapy (first-line therapy) seems to be an approach associated with relatively poor results. In a comparison of four commonly used combination chemotherapy regimens, none was superior. Response rates varied from 15% to 22%, with 1-year survival rates of 31% to 36% (see e.g. O'Mahony, D., S. Kummar, et al. (2005). “Non-small-cell lung cancer vaccine therapy: a concise review.” J Clin Oncol 23(35): 9022-8). Thus, even though preoperative chemotherapy seems to have not resulted in a prolongation of life expectancy, adjuvant chemotherapy—also if combined with radiotherapy—did show a significant increase in life expectancy.
  • One of the chemotherapeutic approaches used today are combinations of platin-based substances with e.g. Gemcitabin even as first-line-therapy, wheras e.g. Pemetrexed is used as second-line therapy.
  • Another option used for the treatment of NSCLC is the so-called “Targeted Therapy” trying to enhance success of classical cytotoxic chemotherapy by influencing tumor specific target structures on a molecular level. Substances used include Bevacizumab (an angiogenesis inhibitor) or Erlotinib, which is aimed at the tyrosine kinases of the epidermal growth factor receptor (EGFR).
  • Even though doubtless there is some improvement in the current therapeutic approaches treatment of lung cancer, especially of NSCLC, is still an uphill-struggle with—given the high mortality rates—a strong need for further, alternative or improved ways of treatment.
  • Thus, it is suggested here to use the immune system in an approach for the treatment of the NSCLC. The immune system plays an important role in the treatment and prevention of numerous diseases. According to the present stage of knowledge, various mechanisms are provided by mammalians to protect the organism by identifying and killing e.g. tumor cells. These tumor cells have to be detected and distinguished from the organism's normal cells and tissues.
  • The immune system of vertebrates such as humans consists of many types of proteins, cells, organs, and tissues, which interact in an elaborate and dynamic network. As part of this more complex immune response, the vertebrate system adapts over time to recognize particular pathogens or tumor cells more efficiently. The adaptation process creates immunological memories and allows even more effective protection during future encounters. This process of adaptive or acquired immunity forms the basis for vaccination strategies.
  • The adaptive immune system is antigen-specific and requires the recognition of specific “self” or “non-self” antigens during a process called antigen presentation. Antigen specificity allows for the generation of responses that are tailored to specific pathogens or pathogen-infected cells or tumor cells. The ability to mount these tailored responses is maintained in the body by so called “memory cells”. Should a pathogen infect the body more than once, these specific memory cells are used to quickly eliminate it. The adaptive immune system thus allows for a stronger immune response as well as for an immunological memory, where each pathogen or tumor cell is “remembered” by one or more signature antigens.
  • The major components of the adaptive immune system in vertebrates predominantly include lymphocytes on the cellular level and antibodies on the molecular level. Lymphocytes as cellular components of the adaptive immune system include B cells and T cells which are derived from hematopoietic stem cells in the bone marrow. B cells are involved in the humoral response, whereas T cells are involved in cell mediated immune response. Both B cells and T cells carry receptor molecules that recognize specific targets. T cells recognize a “non-self” target, such as a pathogenic target structure, only after antigens (e.g. small fragments of a pathogen) have been processed and presented in combination with a “self” receptor called a major histocompatibility complex (MHC) molecule. In contrast, the B cell antigen-specific receptor is an antibody molecule on the B cell surface, and recognizes pathogens as such when antibodies on its surface bind to a specific foreign antigen. This antigen/antibody complex is taken up by the B cell and processed by proteolysis into peptides. The B cell then displays these antigenic peptides on its surface MHC class II molecules. This combination of MHC and antigen attracts a matching helper T cell, which releases lymphokines and activates the B cell. As the activated B cell then begins to divide, its offspring secretes millions of copies of the antibody that recognizes this antigen. These antibodies circulate in blood plasma and lymph, bind to pathogens or tumor cells expressing the antigen and mark them for destruction by complement activation or for uptake and destruction by phagocytes.
  • As a cellular component of the adaptive immune system cytotoxic T cells (CDS+) may form a CTL-response. Cytotoxic T cells (CDS+) can recognize peptides from endogenous pathogens and self-antigens bound by MHC type I molecules. CD8+-T cells carry out their killing function by releasing cytotoxic proteins in the cell.
  • Mechanisms of the immune system form targets for curative treatments. Appropriate methods are typically based on the administration of adjuvants to elicit an innate immune response or on the administration of antigens or immunogens in order to evoke an adaptive immune response. As antigens are typically based on specific components of pathogens (e.g. surface proteins) or fragments thereof, administration of nucleic acids to the patient which is followed by the expression of desired polypeptides, proteins or antigens is envisaged as well.
  • Hitherto conventional methods for eliciting the immune response, immunization or vaccination are based on the use of DNA molecules in order to incorporate the required genetic information into the cell. Various methods have been developed for introducing DNA into cells, such as calcium phosphate transfection, polyprene transfection, protoplast fusion, electroporation, microinjection and lipofection, lipofection having in particular proven to be a suitable method. DNA viruses may likewise be used as a DNA vehicle. Because of their infectious properties, such viruses achieve a very high transfection rate. The viruses used are genetically modified in such a manner that no functional infectious particles are formed in the transfected cell. Despite these precautions, however, it is not possible to rule out the risk of uncontrolled propagation of the introduced gene and viral genes, for example due to potential recombination events. This also entails the risk of the DNA being inserted into an intact gene of the host cell's genome by e.g. recombination, with the consequence that this gene may be mutated and thus completely or partially inactivated or may give rise to misinformation. In other words, synthesis of a gene product which is vital to the cell may be completely suppressed or alternatively a modified or incorrect gene product is expressed. One particular risk occurs if the DNA is integrated into a gene which is involved in the regulation of cell growth. In this case, the host cell may become degenerate and lead to cancer or tumor formation. Furthermore, if the DNA introduced into the cell is to be expressed, it is necessary for the corresponding DNA vehicle to contain a strong promoter, such as the viral CMV promoter. The integration of such promoters into the genome of the treated cell may result in unwanted alterations of the regulation of gene expression in the cell. Another risk of using DNA as an agent to induce an immune response (e.g. as a vaccine) is the induction of pathogenic anti-DNA antibodies in the patient into whom the foreign DNA has been introduced, so bringing about a (possibly fatal) immune response.
  • Thus overall, there is room and a need for an efficient system, which may be used to effectively stimulate the immune system to allow treatment of lung cancer, especially of non-small cell lung cancer (NSCLC), while avoiding the problems of uncontrolled propagation of an introduced gene due to DNA based compositions.
  • It is thus an object of the present invention to provide a composition, which a) allows treatment of lung cancer by stimulating the immune system, while b) avoiding the above mentioned disadvantages.
  • SUMMARY OF THE INVENTION
  • This object is solved by the subject matter of the present invention, particularly by an active (immunostimulatory) composition comprising at least one RNA, encoding at least two (preferably different) antigens selected from the group comprising the antigens:
      • hTERT,
      • WT1,
      • MAGE-A2,
      • 5T4,
      • MAGE-A3,
      • MUC1,
      • Her-2/neu,
      • NY-ESO-1,
      • CEA,
      • Survivin,
      • MAGE-C1, and/or
      • MAGE-C2.
  • Surprisingly, it has been found that a specific combination of at least two antigens, antigenic proteins or antigenic peptides of the afore mentioned group, as contained in an active (immunostimulatory) composition according to the present invention, is capable to effectively stimulate the (adaptive) immune system to allow treatment of lung cancer, especially of non-small cell lung cancer (NSCLC). Herein, the terms antigens, antigenic proteins or antigenic peptides may be used synomously. In the context of the present invention, an active (immunostimulatory) composition according to the present invention shall be further understood as a composition, which is able to elicit an immune response, preferably an adaptive immune response as defined herein, due to one of the component(s) contained or encoded by the components of the active (immunostimulatory) composition, preferably by the at least one RNA, preferably (m)RNA, encoding the at least two (preferably different) antigens.
  • The at least one RNA of the active (immunostimulatory) composition may encode hTERT. In the context of this invention “hTERT” is human telomerase reverse transcriptase and the preferred sequence of the RNA, preferably of the mRNA, encoding “hTERT”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 7 (SEQ ID NO: 7), and—even more preferably, in FIG. 8 (SEQ ID NO: 8). Minev, Hipp et al. (2000) described that telomerase is a ribonucleoprotein enzyme which has been linked to malignant transformation in human cells (Minev, B., J. Hipp, et al. (2000). “Cytotoxic T cell immunity against telomerase reverse transcriptase in humans.” Proc Natl Acad Sci U S A 97(9): 4796-801). Telomerase activity is increased in the vast majority of human tumors, making its gene product the first molecule common to all human tumors. The generation of endogenously processed telomerase peptides bound to Class I MHC molecules could therefore target cytotoxic T lymphocytes (CTL) to tumors of different origins. Thus, according to them this could advance vaccine therapy against cancer provided that precursor CTL recognizing telomerase peptides in normal adults and cancer patients can be expanded through immunization. They further demonstrated that the majority of normal individuals and patients with prostate cancer immunized in vitro against two HLA-A2.1 restricted peptides from telomerase reverse transcriptase (hTRT) developed hTRT-specific CTL. Carpenter and Vonderheide (2006) (Carpenter, E. L. and R. H. Vonderheide (2006); “Telomerase-based immunotherapy of cancer.” Expert Opin Biol Ther 6(10): 1031-9) reported that the progression from the cloning of human telomerase reverse transcriptase (hTERT) in 1997 to the first clinical trials of hTERT as an antitumor immunotherapy target has been swift. hTERT is overexpressed in the vast majority of human cancers whereas it has limited expression in normal adult tissue. It plays a critical role in oncogenesis and may be expressed by cancer stem cells. However, despite being a self antigen, hTERT is immunogenic both in vitro and in vivo. Several Phase I studies of hTERT immunotherapy have been completed in patients with breast, prostate, lung and other cancers, and clinical and immunological results are encouraging. Immunotherapy induced functional, antitumor T cells in patients in the absence of clinical toxicity. The opportunity for vaccinating individuals as an immunoprevention strategy can also be envisioned for hTERT-based therapies. Nair, S. K. and Heiser et al. (2000) described the induction of anti-murine TERT immunity in mice vaccinated with dendritic cells transduced with murine TERT RNA (see Nair, S. K., A. Heiser, et al. (2000). “Induction of cytotoxic T cell responses and tumor immunity against unrelated tumors using telomerase reverse transcriptase RNA transfected dendritic cells.” Nat Med 6(9): 1011-7.). According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an hTERT antigen selected from the sequence as shown in FIG. 7 (SEQ ID NO: 7), and more preferably, in FIG. 8 (SEQ ID NO: 8). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an hTERT antigen selected from a fragment, a variant or an epitope of an hTERT sequence as shown in FIG. 7 (SEQ ID NO: 7), and more preferably, as shown in FIG. 8 (SEQ ID NO: 8).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode WT1. In the context of this invention “WT1” is Wilms tumor 1 and the preferred sequence of the RNA, preferably of the mRNA, encoding “WT1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 9 (SEQ ID NO: 9), more preferably in FIG. 10 (SEQ ID NO: 10), and even more preferably in FIG. 11 (SEQ ID NO: 11). Oka, Y. A. and Tsuboi et al. (2004) found that Wilm's tumor protein is overexpressed in lung cancer (see Oka, Y., A. Tsuboi, et al. (2004). “Induction of WT1 (Wilms' tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression.” Proc Natl Acad Sci U S A 101(38): 13885-90). Oka et al. (2004, supra) vaccinated 10 patients with lung cancer with a peptide derived from WT1. They could show that clinical response correlated with anti-tumor CD8+T cell activity. The Wilms' tumor gene WT1 is overexpressed in leukemias and various types of solid tumors, and the WT1 protein was demonstrated to be an attractive target antigen for immunotherapy against these malignancies.
  • Oka et al. (2004, supra) reported the outcome of a phase I clinical study of WT1 peptide-based immunotherapy for patients with breast or lung cancer, myelodysplastic syndrome, or acute myeloid leukemia. Twelve of the 20 patients for whom the efficacy of WT1 vaccination could be assessed showed clinical responses such as reduction in leukemic blast cells or tumor sizes and/or tumor markers. A clear correlation was observed between an increase in the frequencies of WT1-specific cytotoxic T lymphocytes after WT1 vaccination and clinical responses. It was therefore demonstrated that WT1 vaccination could induce WT1-specific cytotoxic T lymphocytes and resulted in cancer regression without damage to normal tissues. According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an WT1 antigen selected from the sequence as shown in FIG. 9 (SEQ ID NO: 9), and more preferably, as shown in FIG. 10 (SEQ ID NO: 10) and even more preferably as shown in FIG. 11 (SEQ ID NO: 11). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an WT1 antigen selected from a fragment, a variant or an epitope of an WT1 sequence as shown in FIG. 9 (SEQ ID NO: 9), and more preferably, as shown in FIG. 10 (SEQ ID NO: 10) and even more preferably as shown in FIG. 11 (SEQ ID NO: 11).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-A2. In the context of this invention “MAGE-A2” is the melanoma antigen family A, 2B and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-A2”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 14 (SEQ ID NO: 14), and even more preferably in FIG. 15 (SEQ ID NO: 15). Gillespie and Coleman (1999) (Gillespie, A. M. and R. E. Coleman (1999). “The potential of melanoma antigen expression in cancer therapy.” Cancer Treat Rev 25(4): 219-27) reported expression in bladder cancer, breast cancer, colorectal cancer, gastric cancer, head and neck cancer, lung cancer, maxillary cancer, melanoma, oesophagus cancer, osteosarcoma and ovary cancer. According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-A2 antigen selected from the sequence as shown in FIG. 14 (SEQ ID NO: 14), and more preferably, as shown in FIG. 15 (SEQ ID NO: 15). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-A2 antigen selected from a fragment, a variant or an epitope of an MAGE-A2 sequence as shown in FIG. 14 (SEQ ID NO: 14), and—more preferably, as shown in FIG. 15 (SEQ ID NO: 15).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode 5T4. In the context of this invention “5T4” is trophoblast glycoprotein and the preferred sequence of the RNA, preferably of the mRNA, encoding “5T4”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 3 (SEQ ID NO: 3), and even more preferably—in FIG. 4 (SEQ ID NO: 4). Harrop, Connolly et al. (2006) reported that the human oncofetal antigen 5T4 is a 72-kDa leucine-rich membrane glycoprotein which is expressed at high levels on the placenta and also on a wide range of human carcinomas including colorectal, gastric, renal, and ovarian cancers but rarely on normal tissues (see Harrop, R., N. Connolly, et al. (2006). “Vaccination of colorectal cancer patients with modified Vaccinia Ankara delivering the tumor antigen 5T4 (TroVax) induces immune responses which correlate with disease control: a phase I/II trial.” Clin Cancer Res 12(11 Pt 1): 3416-24). Overexpression of 5T4 is associated with poor prognosis in patients with colorectal, gastric, and ovarian carcinoma. Despite such compounding factors, 5T4-specific cellular and/or humoral immune responses were induced in the majority of patients (16 of 17; 94%) following TroVax immunization, which was considered encouraging compared with many other cancer immunotherapy trials. In summary, they showed safety and immunogenicity of TroVax delivered via i.m. and i.d. routes of administration. Zhao and Wang (2007) (Zhao, Y. and Y. Wang (2007). “5T4 oncotrophoblast glycoprotein: janus molecule in life and a novel potential target against tumors.” Cell Mol Immunol 4(2): 99-104) reported that 5T4 oncotrophoblast glycoprotein is a transmembrane protein expressed on the embryonic tissue and various malignant tumor cell surfaces. It plays a vital role in the multiple biological and pathological processes including massive cellular migration during the embryogenesis, cell invasion associated with implantation, and neoplastic metastasis in the progression of tumorigenesis. According to Kopreski, Benko et al. (2001) 5T4 is a trophoblast glycoprotein frequently overexpressed in epithelial malignancies that provides a potential target for cancer therapeutics (see Kopreski, M. S., F. A. Benko, et al. (2001). “Circulating RNA as a tumor marker: detection of 5T4 mRNA in breast and lung cancer patient serum.” Ann N Y Acad Sci 945: 172-8). Serum was collected from 19 patients with advanced breast cancer (5 patients) or non-small-cell lung cancer (14 patients), and from 25 normal control volunteers having amplifiable RNA. RNA extracted from the serum was RT-PCR amplified using heminested, two-stage reactions, with products detected by gel electrophoresis. 5T4 mRNA was reproducibly detected in 8/19 (42%) cancer patient sera, including 2/5 breast cancer patient sera and 6/14 lung cancer patient sera, but in only 3/25 (12%) normal control sera (p=0.035). According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an 5T4 antigen selected from the sequence as shown in FIG. 3 (SEQ ID NO: 3), and more preferably, as shown in FIG. 4 (SEQ ID NO: 4). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an 5T4 antigen selected from a fragment, a variant or an epitope of an 5T4 sequence as shown in FIG. 3 (SEQ ID NO: 3), and—more preferably, as shown in FIG. 4 (SEQ ID NO: 4).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-A3. In the context of this invention “MAGE-A3” is the melanoma antigen family A, 3 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-A3”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 16 (SEQ ID NO: 16), and even more preferably in FIG. 17 (SEQ ID NO: 17). Gillespie and Coleman (1999) (Gillespie, A. M. and R. E. Coleman (1999). “The potential of melanoma antigen expression in cancer therapy.” Cancer Treat Rev 25(4): 219-27) reported expression in bladder cancer, breast cancer, colorectal cancer, gastric cancer, glioma, head and neck cancer, lung, maxillary cancer, melanoma, neuroblastoma, oesophagus cancer and ovary cancer. Sienel,
  • Varwark et al. (2004) described a study performed to determine the rate of MAGE-A3 expression in early-stage non-small cell lung cancer (NSCLC) (see Sienel, W., C. Varwerk, et al. (2004). “Melanoma associated antigen (MAGE)-A3 expression in Stages I and II non-small cell lung cancer: results of a multi-center study.” Eur J Cardiothorac Surg 25(1): 131-4). Primary tumor samples from 204 patients with operable clinical stages I or II NSCLC were collected and the pathological stage determined. MAGE-A3 expression was analyzed from tissue samples by detection of MAGE-A3 transcripts using reverse-transcriptase polymerase chain reaction. MAGE-A3 expression was observed in 80 out of the 204 (39.2%) examined stages I-II primary tumors. Atanackovic, Altorki et al. (2004) described that MAGE-A3 a tumor-associated antigen originally identified in melanoma, was also found in non-small cell lung tumors (see
  • Atanackovic, D., N. K. Altorki, et al. (2004). “Vaccine-induced CD4+T cell responses to MAGE-3 protein in lung cancer patients.” J Immunol 172(5): 3289-96). In a clinical trial nine NSCLC patients were vaccinated with the protein; 3 developed antibody responses. Seven of 8 patients who received MAGE-A3 combined with adjuvant ASO2B generated antibodies against MAGE-A3. Several of these patients also developed T cell responses to the protein. According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-A3 antigen selected from the sequence as shown in FIG. 16 (SEQ ID NO: 16), and more preferably, as shown in FIG. 17 (SEQ ID NO: 17). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-A3 antigen selected from a fragment, a variant or an epitope of an MAGE-A3 sequence as shown in FIG. 16 (SEQ ID NO: 16), and more preferably, as shown in FIG. 17 (SEQ ID NO: 17).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode MUC1. In the context of this invention “MUC1” is mucin 1 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MUC1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 1 (SEQ ID NO: 1), and even more preferably in FIG. 2 (SEQ ID NO: 2). Cancer-associated mucins are a potential target for immunotherapy. These molecules are thought to promote metastases by facilitating adhesion of malignant cells to the endothelial cell surface. According to Denda-Nagai and Irimura (2000) (Denda-Nagai, K. and T. Irimura (2000). “MUC1 in carcinoma-host interactions.” Glycoconj J 17(7-9): 649-58) MUC-1 is overexpressed in 90% of all adenocarcinomas, including breast, lung, pancreas, prostate, stomach, colon and ovary. Kontani, Taguchi et al. (2001) found that MUC-1 has been found to be expressed in 60% of lung cancers (see Kontani, K., 0. Taguchi, et al. (2001). “Modulation of MUC1 mucin as an escape mechanism of breast cancer cells from autologous cytotoxic T-lymphocytes.” Br J Cancer 84(9): 1258-64), whereas Kontani, Taguchi et al. (2003) found in a study analyzing the use of pulsed DCs with MUC1 antigens to elicit cellular immunity in MUC1 positive cancers, that clinically seven of nine MUC-1 positive patients responded to the treatment with either a reduction in tumor marker levels or disappearance of malignant pleural effusion (see Kontani, K., 0. Taguchi, et al. (2003). “Dendritic cell vaccine immunotherapy of cancer targeting MUC1 mucin.” Int J Mol Med 12(4): 493-502). Three of these responding patients had NSCLC. Palmer, Parker et al. (2001) reported that in a phase I clinical trial using MUC1 peptide in stage III/IV NSCLC, safety and tolerability of this agent was established (see Palmer, M., J. Parker, et al. (2001). “Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer.” Clin Lung Cancer 3(1): 49-57; discussion 58). Five of 12 patients (42%) had immunologic responses, and 4 of 12 patients (33%) achieved stable disease. Wierecky, Mueller et al. (2006) further identified two HLA-A2 binding novel 9-mer peptides of the TAA MUC1, which is overexpressed on various hematological and epithelial malignancies (see Wierecky, J., M. Mueller, et al. (2006). “Dendritic cell-based cancer immunotherapy targeting MUC-1.” Cancer Immunol Immunother 55(1): 63-7). Cytotoxic T cells generated after pulsing DC with these peptides were able to induce lysis of tumor cells expressing MUC1 in an antigen-specific and HLA-restricted fashion. Within two clinical studies, it was demonstrated that vaccination of patients with advanced cancer using DCs pulsed with MUC1 derived peptides was well tolerated without serious side effects and was able to induce immunological responses. Of 20 patients with metastatic renal cell carcinoma, 6 patients showed regression of metastases with 3 objective responses (1 CR, 2 PR). According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an MUC1 antigen selected from the sequence as shown in FIG. 1 (SEQ ID NO: 1), and more preferably, as shown in FIG. 2 (SEQ ID NO: 2). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MUC1 antigen selected from a fragment, a variant or an epitope of an MUC1 sequence as shown in FIG. 1 (SEQ ID NO: 1), and more preferably, as shown in FIG. 2 (SEQ ID NO: 2).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode Her-2/neu. In the context of this invention “Her-2/neu” is v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 and the preferred sequence of the RNA, preferably of the mRNA, encoding “Her-2/neu”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 5 (SEQ ID NO: 5), and even more preferably in FIG. 6 (SEQ ID NO: 6). According to Baxevanis, Sotiropolou et al. (2004) HER-2/neu (also known as HER2 or c-erb-B2) is a 185-kDa protein receptor with tyrosine kinase activity and extensive homology to the epidermal growth factor (EGF) receptor (see Baxevanis, C. N., P. A. Sotiropoulou, et al. (2004). “Immunobiology of HER-2/neu oncoprotein and its potential application in cancer immunotherapy.” Cancer Immunol Immunother 53(3): 166-75). HER-2/neu is expressed in many epithelial tumors and known to be overexpressed in approximately 20-25% of all ovarian and breast cancers, 35-45% of all pancreatic adenocarcinomas, and up to 90% of colorectal carcinomas. HER-2/neu overexpression represents a marker of poor prognosis. Overexpression of Her-2 has been observed in malignant tumors of the breast, ovary, pancreas, colon, lung and other tissues. Her-2 is normally expressed at low levels in variety of human tissues (skin, digestive tract epithelium, breast, ovary, hepatocytes). Bernhard, Salazar (2002) report in their conclusion that early results of clinical trials actively immunizing cancer patients against HER-2/neu demonstrated that immunity could be generated and that immune responses persisted over a period of time (see Bernhard, H., Salazar L., et al. (2002). “Vaccination against the HER-2/neu oncogenic protein.” Endocr Relat Cancer 9(1): 33-44). Current vaccine trials were focused solely on the use of epitope- or peptide-based vaccines, largely due to the observation that peptide vaccine strategies could circumvent neu-specific tolerance in rodent models. The next generation of vaccine approaches according to Bernhard et al. (2002, supra) will likely include protein-based vaccines, HER-2/neu antigen preparations loaded onto DC, and nucleic acid based formulations. Studies in rodent models exploring these strategies at a pre-clinical level were promising. Expansion of HER-2/neu-specific T-cell ex vivo following active immunization or in vitro culture with HER-2/neu-expressing DC was thus considered as being a therapeutic option for treating advanced stage HER-2/neu-overexpressing tumors. Baxevanis, Sotiridou et al. (2006) found that in humans, although immunological responses have been detected against the peptides used for vaccination no clinical responses have been described (see Baxevanis, C. N., N. N. Sotiriadou, et al. (2006). “Immunogenic HER-2/neu peptides as tumor vaccines.” Cancer Immunol Immunother 55(1): 85-95). According to Disis, Gooley et al. (2002) Her-2/neu is a member of the EGFR familiy (see Disis, M. L., T. A. Gooley, et al. (2002). “Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines.” J Clin Oncol 20(11): 2624-32). It is frequently overexpressed in breast, ovary, prostate, colon and lung cancers. In a phase I clinical trial 38 patients (2 with NSCLC) were vaccinated with a Her-2/neu peptide. 92% of the patients developed T-cell immunity to Her-2/neu. According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an Her-2/neu antigen selected from the sequence as shown in FIG. 5 (SEQ ID NO: 5), and more preferably, as shown in FIG. 6 (SEQ ID NO: 6). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an Her-2/neu antigen selected from a fragment, a variant or an epitope of an Her-2/neu sequence as shown in FIG. 5 (SEQ ID NO: 5), and more preferably, as shown in FIG. 6 (SEQ ID NO: 6).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode NY-ESO-1. In the context of this invention “NY-ESO-1” is cancer/testis antigen 1B and the preferred sequence of the RNA, preferably of the mRNA, encoding “NY-ESO-1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 20 (SEQ ID NO: 20), and even more preferably—in FIG. 21 (SEQ ID NO: 21). Chen, Scanlan et al. (1997) reported the mRNA expression of NY-ESO-1 in various human tumors by RT-PCR finding Melanoma 23/67, Ovarian cancer 2/8, Breast cancer 10/33, Thyroid cancer 2/5, Prostate cancer 4/16, Bladder cancer 4/5, Colon cancer 0/16, Burkitt lymphoma 1/2, Glioma 0/15, Basal cell carcinoma 0/2, Gastric cancer 0/12, Leiomyosarcoma 0/2, Lung cancer 2/12, Other sarcomas 0/2, Renal cancer 0/10, Pancreatic cancer 0/2, Lymphoma 0/10, Seminoma 0/1, Hepatoma 2/7, Spinal cord tumor 0/1 (see Chen, Y. T., M. J. Scanlan, et al. (1997). “A testicular antigen aberrantly expressed in human cancers detected by autologous antibody screening.” Proc Natl Acad Sci U S A 94(5): 1914-8). Jager, Karbach et al. (2006) reported that NY-ESO-1 is a cancer/testis antigen expressed in a range of human malignancies, and that a number of vaccine strategies targeting NY-ESO-1 were being developed (see Jager, E., J. Karbach, et al. (2006). “Recombinant vaccinia/fowlpox NY-ESO-1 vaccines induce both humoral and cellular NY-ESO-1-specific immune responses in cancer patients.” Proc Natl Acad Sci U S A 103(39): 14453-8). In the presented study, the safety and immunogenicity of recombinant vaccinia-NY-ESO-1 and recombinant fowlpox-NY-ESO-1 were analyzed in a series of 36 patients with a range of different tumor types. Each construct was first tested individually at two different dose levels and then in a prime-boost setting with recombinant vaccinia-NY-ESO-1 followed by recombinant fowlpox-NY-ESO-1. The vaccines were well tolerated either individually or together. NY-ESO-1-specific antibody responses and/or specific CD8 and CD4 T cell responses directed against a broad range of NY-ESO-1 epitopes were induced by a course of at least four vaccinations at monthly intervals in a high proportion of patients. CD8 T cell clones derived from five vaccinated patients were shown to lyse NY-ESO-1-expressing melanoma target cells. In several patients with melanoma, there was a strong impression that the natural course of the disease was favorably influenced by vaccination. Davis, Chen et al. (2004) reported that HLA-A2-restricted NY-ESO-1 peptides injected intradermally were shown to be safe and immunogenic (Davis, I. D., W. Chen, et al. (2004). “Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4(+) and CD8(+) T cell responses in humans.” Proc Natl Acad Sci U S A 101(29): 10697-702). Although these trials were designed only to determine safety and immunogenicity, some patients showed tumor regression or stabilization of disease. It was further expressed by Jager, Gnjatic et al. (2000) that a broad NY-ESO-1-specific immune response including antibody and CD4 and CD8 T cell responses was seen after immunization with recombinant NY-ESO-1 protein combined with ISCOMATRIX adjuvant (CSL Ltd., Parkville, Victoria, Australia) in patients with resected NY-ESO-1-expressing melanoma (see Jager, E., S. Gnjatic, et al. (2000). “Induction of primary NY-ESO-1 immunity: CD8+ T lymphocyte and antibody responses in peptide-vaccinated patients with NY-ESO-1+cancers.” Proc Natl Acad Sci U S A 97(22): 12198-203). This immune response to the vaccine appeared to be associated with long disease-free survival. Furthermore Odunsi, Qian et al. (2007) reported that vaccination with an NY-ESO-1 peptide induces integrated humoral and T cell responses in ovarian cancer (see Odunsi, K., F. Qian, et al. (2007). “Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer.” Proc Natl Acad Sci U S A 104(31): 12837-42). According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an NY-ESO-1 antigen selected from the sequence as shown in FIG. 20 (SEQ ID NO: 20), and more preferably, as shown in FIG. 21 (SEQ ID NO: 21). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an NY-ESO-1 antigen selected from a fragment, a variant or an epitope of an NY-ESO-1 sequence as shown in FIG. 20 (SEQ ID NO: 20), and more preferably, as shown in FIG. 21 (SEQ ID NO: 21).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode CEA. In the context of this invention “CEA” is carcinoembryonic antigen (CECAM5=carcinoembryonic antigen-related cell adhesion molecule 5) and the preferred sequence of the RNA, preferably of the mRNA, encoding “CEA”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 12 (SEQ ID NO: 12), and even more preferably in FIG. 13 (SEQ ID NO: 13). According to Hammarstrom (1999) CEA is a 180 kDa onco-fetal glycoprotein that acts as an adhesion molecule, and is overexpressed in 70% of NSCLC (Hammarstrom, S. (1999). “The carcinoembryonic antigen (CEA) family: structures, suggested functions and expression in normal and malignant tissues.” Semin Cancer Biol 9(2): 67-81). Berinstein (2002) reported that CEA has many attractive features as a target for active vaccination approaches against cancer (Berinstein, N. L. (2002). “Carcinoembryonic antigen as a target for therapeutic anticancer vaccines: a review.” J Clin Oncol 20(8): 2197-207). It has a favorable expression pattern and is expressed in more than 50% of all human cancers. It may play a role in the tumorigenesis process itself, and thus its expression may be selected and conserved throughout cancer progression. It has been well documented that CEA is processed and presented on various MHC class 1 molecules. Moreover, immunologic tolerance to CEA is not absolute. There are extensive data demonstrating that human T cells can recognize, become activated to, and lyse cancer cells that are expressing CEA. Several different therapeutic vaccination approaches using CEA as a target antigen have been assessed. The safety of these approaches has been established. In addition, humoral and/or cellular responses to CEA have been documented. Although for the most part the patients chosen for these studies presented by Berinstein (2002, supra) had very advanced and refractory metastatic colon cancer, some evidence of clinical activity has been documented, with disease stabilization and even objective responses occurring in some patients. Dendritic cells pulsed with an agonist CEA MHC class I binding peptide (CAP1-6D) and poxvirus-based vectors incorporating CEA, with or without costimulatory molecules, seemed most active in activating CD8 T-cell responses. Unfortunately, dendritic cell approaches may be limited by the logistical difficulty of obtaining patient-specific preparations of dendritic cells. Four phase I studies using the canarypox vector system to target CEA were reported. These trials showed that such approaches were safe, with mild grade 1 and grade 2 toxicities limited primarily to the site of injection. Moreover, the trials showed that specific cellular T-cell responses can be activated to CEA in the majority of patients. These responses may be enhanced by the inclusion of the B7.1 costimulatory molecule in the vector or by the addition of recombinant GM-CSF at the injection site. Although no objective clinical responses were reported, a significant proportion of patients in these phase I studies have experienced disease stabilization. Vaccination strategies to further enhance the frequency of T cells recognizing CEA where considered to further augment the clinical activity of these vaccines. There are data that suggest that at least some vaccines may be more effective in minimal disease states. Ueda, Itoh et al (2004) described one study, in which 18 patients with metastatic gastrointestinal or lung cancer were treated with autologous dendritic cells pulsed with CEA-derived peptide (see Ueda, Y., T. Itoh, et al. (2004). “Dendritic cell-based immunotherapy of cancer with carcinoembryonic antigen-derived, HLA-A24-restricted CTL epitope: Clinical outcomes of 18 patients with metastatic gastrointestinal or lung adenocarcinomas.” Int .1 Oncol 24(4): 909-17). Immune reactions measured by skin testing and in vitro T cell assays were observed in most of the patients. Although no objective clinical responses were reported, some patients had stable disease while receiving this immunotherapy. According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an CEA antigen selected from the sequence as shown in FIG. 12 (SEQ ID NO: 12), and—more preferably, as shown in FIG. 13 (SEQ ID NO: 13). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an CEA antigen selected from a fragment, a variant or an epitope of an CEA sequence as shown in FIG. 12 (SEQ ID NO: 12), and more preferably, as shown in FIG. 13 (SEQ ID NO: 13).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode Survivin. In the context of this invention “Survivin” is baculoviral IAP repeat-containing 5 (survivin) and the preferred sequence of the RNA, preferably of the mRNA, encoding “survivin”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 18 (SEQ ID NO: 18), and even more preferably in FIG. 19 (SEQ ID NO: 19). Grube, Moritz et al. (2007) described Survivin (see Grube, M., S. Moritz, et al. (2007). “CD8+ T cells reactive to survivin antigen in patients with multiple myeloma.” Clin Cancer Res 13(3): 1053-60). Survivin is a member of the inhibitors of apoptosis family and is overexpressed in different types of malignancies. Cytotoxic T cells recognizing survivin epitopes can be elicited in vitro and by vaccination in patients with leukemia, breast cancer, and melanoma. It was investigated whether survivin-specific CD8+ T cells occur in patients with multiple myeloma and T cells recognizing HLA-A2.1binding survivin peptide were detected in 9 of 23 patients and in 1 of 21 healthy volunteers. Survivin-reactive T cells were identified as terminally differentiated effector T cells (CD8+, CD45RA+, and CCR7). Positive survivin expression of myeloma cells in bone marrow specimens was shown in 7 of 11 patients. Survivin is highly expressed in most human cancer cells of epithelial and hematopoietic origin, and overexpression is associated with cancer progression, poor prognosis, resistance, and short patient survival. Duffy, O'Donovan (2007) described that Survivin is a 16.5 kDa protein overexpressed in almost all malignancies but rarely detected in normal differentiated adult tissues (see Duffy, M. J., N. O'Donovan, et al. (2007). “Survivin: a promising tumor biomarker.” Cancer Lett 249(1): 49-60). Functionally, survivin has been shown to inhibit apoptosis, promote cell proliferation and enhance angiogenesis. Consistent with its role in these processes, survivin was described as playing a key role in cancer progression. Because of the large difference in expression between normal and malignant tissue and its causal role in cancer progression, survivin is currently undergoing intensive investigation as a potential tumor marker. Emerging data suggests that measurement of survivin can aid the early diagnosis of bladder cancer, determine prognosis in multiple cancer types and predict response to diverse anti-cancer therapies. Zeis, Siegel et al. (2003) demonstrated that human survivin-specific CTLs generated from PBMC by stimulation with autologous dendritic cells transfected with survivin-RNA were cytotoxic for a range of hemopoietic malignant cell lines and primary tumor cells isolated from patients with acute myeloid leukemia (see Zeis, M., S. Siegel, et al. (2003). “Generation of cytotoxic responses in mice and human individuals against hematological malignancies using survivin-RNA-transfected dendritic cells.” J Immunol 170(11): 5391-7). It was also shown that vaccination of mice with survivin-RNA-transfected dendritic cells lead to long term resistance to challenge by a survivin-expressing lymphoma, demonstrating the potential of survivin as a tumor rejection Ag. Evidence for the use of survivin as a target structure for immunotherapeutic strategies against hematological neoplasms was provided. According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an Survivin antigen selected from the sequence as shown in FIG. 18 (SEQ ID NO: 18), and more preferably, as shown in FIG. 19 (SEQ ID NO: 19). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an Survivin antigen selected from a fragment, a variant or an epitope of an Survivin sequence as shown in FIG. 18 (SEQ ID NO: 18), and more preferably, as shown in FIG. 19 (SEQ ID NO: 19).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-C1. In the context of this invention “MAGE-C1” is the melanoma antigen family C, 1 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-C 1”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 22 (SEQ ID NO: 22), more preferably in FIG. 23 (SEQ ID NO: 23), and even more preferably—in FIG. 24 (SEQ ID NO: 24). Lucas, De Smet et al. (1998) recently identified MAGE-C1 by performing RDA (see Lucas, S., C. De Smet, et al. (1998). “Identification of a new MAGE gene with tumor-specific expression by representational difference analysis.”
  • Cancer Res 58(4): 743-52). MAGE-C1 was not expressed in a panel of normal tissues tested with the exception of testis. Among tumoral samples, MAGE-C1 was frequently expressed in seminomas, melanomas, and bladder carcinomas. It was also expressed in a significant fraction of head and neck carcinomas, breast carcinomas, non-small lung carcinomas, prostate adenocarcinomas and sarcomas. Jungbluth, Chen et al. (2002) described expression in breast cancer, ovary cancer, liver cancer, testis cancer, bladder cancer, melanoma and non-small cell lung cancer (39%) (see Jungbluth, A. A., Y. T. Chen, et al. (2002). “CT7 (MAGE-C1) antigen expression in normal and neoplastic tissues.” Int J Cancer 99(6): 839-45). Gure, Chua et al. (2005) analyzed tumors from 523 non-small-cell lung cancer (NSCLC) patients for the expression of cancer-testis antigens (see Gure, A. O., R. Chua, et al. (2005). “Cancer-testis genes are coordinately expressed and are markers of poor outcome in non-small cell lung cancer.” Clin Cancer Res 11(22): 8055-62). MAGE-C1 was present in 18,8%. Scanlan, Altorki et al. (2000) furthermore reported expression of CT antigens in 33 non-small cell lung cancers: MAGE-C 1 : 30% (see Scanlan, M. J., N. K. Altorki, et al. (2000). “Expression of cancer-testis antigens in lung cancer: definition of bromodomain testis-specific gene (BRDT) as a new CT gene, CT9.” Cancer Lett 150(2): 155-64). According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-C1 antigen selected from the sequence as shown in FIG. 22 (SEQ ID NO: 22), and—more preferably, as shown in FIG. 23 (SEQ ID NO: 23) and even more preferably as shown in FIG. 24 (SEQ ID NO: 24). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-C 1 antigen selected from a fragment, a variant or an epitope of an MAGE-C1 sequence as shown in FIG. 22 (SEQ ID NO: 22), and more preferably, as shown in FIG. 23 (SEQ ID NO: 23) and even more preferably as shown in FIG. 24 (SEQ ID NO: 24).
  • The at least one RNA of the active (immunostimulatory) composition may furthermore encode MAGE-C2. In the context of this invention “MAGE-C2” is the melanoma antigen family C2 and the preferred sequence of the RNA, preferably of the mRNA, encoding “MAGE-C2”—if being used in the active (immunostimulatory) composition according to the invention—is shown in FIG. 25 (SEQ ID NO: 25), and—even more preferably in FIG. 26 (SEQ ID NO: 26). Lucas, De Plaen et al. (2000) recently identified MAGE-C2 by performing RDA on a melanoma cell line (see Lucas, S., E. De Plaen, et al. (2000). “MAGE-B5, MAGE-B6, MAGE-C2, and MAGE-C3: four new members of the MAGE family with tumor-specific expression.” Int J Cancer 87(1): 55-60). MAGE-C2 was not expressed in a panel of normal tissues tested with the exception of testis. Among tumoral samples, MAGE-C2 was frequently expressed in seminomas, melanomas, and bladder carcinomas. It was also expressed in a significant fraction of head and neck carcinomas, breast carcinomas, non-small lung carcinomas and sarcomas. Scanlan, Altorki et al. (2000) reported expression of CT antigens in 33 non-small cell lung cancers: MAGE-C2: 30% (see Scanlan, M. J., N. K. Altorki, et al. (2000). “Expression of cancer-testis antigens in lung cancer: definition of bromodomain testis-specific gene (BRDT) as a new CT gene, CT9.” Cancer Lett 150(2): 155-64). According to a preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may thus encode an MAGE-C2 antigen selected from the sequence as shown in FIG. 25 (SEQ ID NO: 25), and more preferably, as shown in FIG. 26 (SEQ ID NO: 26). According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition may alternatively or additionally encode an MAGE-C2 antigen selected from a fragment, a variant or an epitope of an MAGE-C2 sequence as shown in FIG. 25 (SEQ ID NO: 25), and more preferably, as shown in FIG. 26 (SEQ ID NO: 26).
  • Antigens, antigenic proteins or antigenic peptides as defined above which may be encoded by the at least one RNA of the active (immunostimulatory) composition according to the present invention, may comprise fragments or variants of those sequences. Such fragments or variants may typically comprise a sequence having a sequence homology with one of the above mentioned antigens, antigenic proteins or antigenic peptides or sequences or their encoding nucleic acid sequences of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, preferably at least 70%, more preferably at least 80%, equally more preferably at least 85%, even more preferably at least 90% and most preferably at least 95% or even 97%, to the entire wild-type sequence, either on nucleic acid level or on amino acid level.
  • “Fragments” of antigens, antigenic proteins or antigenic peptides in the context of the present invention may comprise a sequence of an antigen, antigenic protein or antigenic peptide as defined above, which is, with regard to its amino acid sequence (or its encoded nucleic acid sequence), N-terminally, C-terminally and/or intrasequentially truncated compared to the amino acid sequence of the original (native) protein (or its encoded nucleic acid sequence). Such truncation may thus occur either on the amino acid level or correspondingly on the nucleic acid level. A sequence homology with respect to such a fragment as defined above may therefore preferably refer to the entire antigen, antigenic protein or antigenic peptide as defined above or to the entire (coding) nucleic acid sequence of such an antigen, antigenic protein or antigenic peptide.
  • Fragments of antigens, antigenic proteins or antigenic peptides in the context of the present invention may furthermore comprise a sequence of an antigen, antigenic protein or antigenic peptide as defined above, which has a length of about 6 to about 20 or even more amino acids, e.g. fragments as processed and presented by MHC class I molecules, preferably having a length of about 8 to about 10 amino acids, e.g. 8, 9, or 10, (or even 6, 7, 11, or 12 amino acids), or fragments as processed and presented by MHC class II molecules, preferably having a length of about 13 or more amino acids, e.g. 13, 14, 15, 16, 17, 18, 19, 20 or even more amino acids, wherein these fragments may be selected from any part of the amino acid sequence. These fragments are typically recognized by T-cells in form of a complex consisting of the peptide fragment and an MHC molecule, i.e. the fragments are typically not recognized in their native form.
  • Fragments of antigens, antigenic proteins or antigenic peptides as defined herein may also comprise epitopes of those antigens, antigenic proteins or antigenic peptides. Epitopes (also called “antigen determinants”) in the context of the present invention are typically fragments located on the outer surface of (native) antigens, antigenic proteins or antigenic peptides as defined herein, preferably having 5 to 15 amino acids, more preferably having 5 to 12 amino acids, even more preferably having 6 to 9 amino acids, which may be recognized by antibodies or B-cell receptors, i.e. in their native form. Such epitopes of antigens, antigenic proteins or antigenic peptides may furthermore be selected from any of the herein mentioned variants of such antigens, antigenic proteins or antigenic peptides. In this context antigenic determinants can be conformational or discontinous epitopes which are composed of segments of the antigens, antigenic proteins or antigenic peptides as defined herein that are discontinuous in the amino acid sequence of the antigens, antigenic proteins or antigenic peptides as defined herein but are brought together in the three-dimensional structure or continuous or linear epitopes which are composed of a single polypeptide chain.
  • “Variants” of antigens, antigenic proteins or antigenic peptides as defined above may be encoded by the at least one RNA of the active (immunostimulatory) composition according to the present invention, wherein nucleic acids of the at least one (m)RNA, encoding the antigen, antigenic protein or antigenic peptide as defined above, are exchanged. Thereby, an antigen, antigenic protein or antigenic peptide may be generated, having an amino acid sequence which differs from the original sequence in one or more mutation(s), such as one or more substituted, inserted and/or deleted amino acid(s). Preferably, these fragments and/or variants have the same biological function or specific activity compared to the full-length native antigen or antigenic potein, e.g. its specific antigenic property.
  • The at least one RNA of the active (immunostimulatory) composition according to the present invention may also encode an antigen or an antigenic protein as defined above, wherein the encoded amino acid sequence comprises conservative amino acid substitution(s) compared to its physiological sequence. Those encoded amino acid sequences as well as their encoding nucleotide sequences in particular fall under the term variants as defined above. Substitutions in which amino acids which originate from the same class are exchanged for one another are called conservative substitutions. In particular, these are amino acids having aliphatic side chains, positively or negatively charged side chains, aromatic groups in the side chains or amino acids, the side chains of which can enter into hydrogen bridges, e.g. side chains which have a hydroxyl function. This means that e.g. an amino acid having a polar side chain is replaced by another amino acid having a likewise polar side chain, or, for example, an amino acid characterized by a hydrophobic side chain is substituted by another amino acid having a likewise hydrophobic side chain (e.g. serine (threonine) by threonine (serine) or leucine (isoleucine) by isoleucine (leucine)). Insertions and substitutions are possible, in particular, at those sequence positions which cause no modification to the three-dimensional structure or do not affect the binding region. Modifications to a three-dimensional structure by insertion(s) or deletion(s) can easily be determined e.g. using CD spectra (circular dichroism spectra) (Urry, 1985, Absorption, Circular Dichroism and ORD of Polypeptides, in: Modern Physical Methods in Biochemistry, Neuberger et al. (ed.), Elsevier, Amsterdam).
  • Furthermore, variants of antigens, antigenic proteins or antigenic peptides as defined above, which may be encoded by the at least one RNA of the active (immunostimulatory) composition according to the present invention, may also comprise those sequences, wherein nucleic acids of the at least one (m)RNA are exchanged according to the degeneration of the genetic code, without leading to an alteration of respective amino acid sequence of the antigen, antigenic protein or antigenic peptide, i.e. the amino acid sequence or at least part thereof may not differ from the original sequence in one or more mutation(s) within the above meaning.
  • In order to determine the percentage to which two sequences (nucleic acid sequences, e.g. RNA or mRNA sequences as defined herein, or amino acid sequences, preferably their encoded amino acid sequences, e.g. the amino acid sequences of the antigens, antigenic proteins or antigenic peptides as defined above) are identical, the sequences can be aligned in order to be subsequently compared to one another. Therefore, e.g. gaps can be inserted into the sequence of the first sequence and the component at the corresponding position of the second sequence can be compared. If a position in the first sequence is occupied by the same component as is the case at a position in the second sequence, the two sequences are identical at this position. The percentage to which two sequences are identical is a function of the number of identical positions divided by the total number of positions. The percentage to which two sequences are identical can be determined using a mathematical algorithm. A preferred, but not limiting, example of a mathematical algorithm which can be used is the algorithm of Karlin et al. (1993), PNAS USA, 90:5873-5877 or Altschul et al. (1997), Nucleic Acids Res., 25:3389-3402. Such an algorithm is integrated in the BLAST program. Sequences which are identical to the sequences of the present invention to a certain extent can be identified by this program.
  • The active (immunostimulatory) composition according to the present invention comprises, as defined above, at least one RNA, encoding least two (preferably different) antigens selected from any of the antigens of the above group, since according to the invention a specific combination of at least two (preferably different) antigens of the afore mentioned group is capable to effectively stimulate the (adaptive) immune system to allow treatment of lung cancer, especially of non-small cell lung cancer (NSCLC). However, the present invention may also provide such active (immunostimulatory) compositions, comprising at least one RNA, encoding three, four, five, six, seven, eight, nine, ten, eleven or even even twelve (preferably different) antigens selected from any of the antigens of the above group, wherein any combination of these antigens is possible and envisaged.
  • According to a particularly preferred embodiment, the at least one RNA of the active (immunostimulatory) composition according to the present invention, may encode at least two (preferably different) antigens selected from any of the antigens of a subgroup comprising the following antigens:
      • hTERT,
      • WT1,
      • 5T4,
      • NY-ESO-1,
      • Survivin, and/or
      • MAGE-C2.
  • More preferably, the present invention may also provide an active (immunostimulatory) composition, comprising at least one RNA, encoding at least three, four, five or six (preferably different) antigens selected from any of the antigens of the above group or subgroup, wherein any combination of these antigens is possible.
  • Accordingly, due to another particularly preferred embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention, may encode at least two (preferably different) antigens selected from any of the antigens of the above mentioned group(s) or subgroup(s) comprising (at least) any one of the following combinations of antigens:
      • hTERT and WT1, or
      • hTERT and 5T4, or
      • hTERT and NY-ESO-1, or
      • hTERT and Survivin, or
      • hTERT and MAGE-C2, or
      • WT1 and 5T4, or
      • WT1 and NY-ESO-1, or
      • WT1 and Survivin, or
      • WT1 and MAGE-C2, or
      • 5T4 and NY-ESO-1, or
      • 5T4 and Survivin, or
      • 5T4 and MAGE-C2, or
      • NY-ESO-1 and Survivin, or
      • NY-ESO-1 and MAGE-C2, or
      • Survivin and MAGE-C2,
      • or
      • hTERT, WT1 and 5T4, or
      • hTERT, WT1 and NY-ESO-1, or
      • hTERT, WT1 and Survivin, or
      • hTERT, WT1 and MAGE-C2, or
      • hTERT, 5T4, and NY-ESO-1, or
      • hTERT, 5T4, and Survivin, or
      • hTERT, 5T4, and MAGE-C2, or
      • hTERT, NY-ESO-1 and Survivin, or
      • hTERT, NY-ESO-1 and MAGE-C2, or
      • hTERT, Survivin and MAGE-C2, or
      • WT1, 5T4 and NY-ESO-1, or
      • WT1, 5T4 and Survivin, or
      • WT1, 5T4 and MAGE-C2, or
      • WT1, NY-ESO-1 and Survivin, or
      • WT1, NY-ESO-1 and MAGE-C2, or
      • WT1, Survivin and MAGE-C2, or
      • 5T4, NY-ESO-1 and Survivin, or
      • 5T4, NY-ESO-1 and MAGE-C2, or
      • 5T4, Survivin and MAGE-C2, or
      • NY-ESO-1, Survivin, and MAGE-C2,
      • or
      • hTERT, WT1, 5T4 and NY-ESO-1, or
      • hTERT, WT1, 5T4 and Survivin, or
      • hTERT, WT1, 5T4 and MAGE-C2, or
      • hTERT, 5T4, NY-ESO-1 and Survivin, or
      • hTERT, 5T4, NY-ESO-1 and MAGE-C2, or
      • hTERT, NY-ESO-1, Survivin and MAGE-C2, or
      • WT1, 5T4, NY-ESO-1, and Survivin, or
      • WT1, 5T4, NY-ESO-1, and MAGE-C2, or
      • WT1, 5T4, Survivin, and MAGE-C2, or
      • 5T4, NY-ESO-1, Survivin, and MAGE-C2,
      • or
      • hTERT, WT1, 5T4, NY-ESO-1 and Survivin, or
      • hTERT, WT1, 5T4, NY-ESO-1 and MAGE-C2, or
      • WT1, 5T4, NY-ESO-1, Survivin and MAGE-C2,
      • or
      • hTERT, WT1, 5T4, NY-ESO-1, Survivin, and MAGE-C2.
  • More preferably, the at least one RNA of the active (immunostimulatory) composition of the present invention, may encode at least two (preferably different) antigens exclusively selected from any of the antigens of the above mentioned group(s) or subgroup(s) comprising (at least) any one of the following combinations of antigens:
      • hTERT and WT1, or
      • hTERT and 5T4, or
      • hTERT and NY-ESO-1, or
      • hTERT and Survivin, or
      • hTERT and MAGE-C2, or
      • WT1 and 5T4, or
      • WT1 and NY-ESO-1, or
      • WT1 and Survivin, or
      • WT1 and MAGE-C2, or
      • 5T4 and NY-ESO-1, or
      • 5T4 and Survivin, or
      • 5T4 and MAGE-C2, or
      • NY-ESO-1 and Survivin, or
      • NY-ESO-1 and MAGE-C2, or
      • Survivin and MAGE-C2,
      • or
      • hTERT, WT1 and 5T4, or
      • hTERT, WT1 and NY-ESO-1, or
      • hTERT, WT1 and Survivin, or
      • hTERT, WT1 and MAGE-C2, or
      • hTERT, 5T4, and NY-ESO-1, or
      • hTERT, 5T4, and Survivin, or
      • hTERT, 5T4, and MAGE-C2, or
      • hTERT, NY-ESO-1 and Survivin, or
      • hTERT, NY-ESO-1 and MAGE-C2, or
      • hTERT, Survivin and MAGE-C2, or
      • WT1, 5T4 and NY-ESO-1, or
      • WT1, 5T4 and Survivin, or
      • WT1, 5T4 and MAGE-C2, or
      • WT1, NY-ESO-1 and Survivin, or
      • WT1, NY-ESO-1 and MAGE-C2, or
      • WT1, Survivin and MAGE-C2, or
      • 5T4, NY-ESO-1 and Survivin, or
      • 5T4, NY-ESO-1 and MAGE-C2, or
      • 5T4, Survivin and MAGE-C2, or
      • NY-ESO-1, Survivin, and MAGE-C2,
      • or
      • hTERT, WT1, 5T4 and NY-ESO-1, or
      • hTERT, WT1, 5T4 and Survivin, or
      • hTERT, WT1, 5T4 and MAGE-C2, or
      • hTERT, 5T4, NY-ESO-1 and Survivin, or
      • hTERT, 5T4, NY-ESO-1 and MAGE-C2, or
      • hTERT, NY-ESO-1, Survivin and MAGE-C2, or
      • WT1, 5T4, NY-ESO-1, and Survivin, or
      • WT1, 5T4, NY-ESO-1, and MAGE-C2, or
      • WT1, 5T4, Survivin, and MAGE-C2, or
      • 5T4, NY-ESO-1, Survivin, and MAGE-C2,
      • or
      • hTERT, WT1, 5T4, NY-ESO-1 and Survivin, or
      • hTERT, WT1, 5T4, NY-ESO-1 and MAGE-C2, or
      • WT1, 5T4, NY-ESO-1, Survivin and MAGE-C2,
      • or
      • hTERT, WT1, 5T4, NY-ESO-1, Survivin, and MAGE-C2.
  • According to a further preferred embodiment, the present invention provides an active (immunostimulatory) composition comprising at least one RNA, encoding at least two (preferably different) antigens,
    • a) wherein at least one, preferably at least two, three, four, five or even six, of these at least two antigens is (are) selected from:
      • 5T4
      • NY-ESO-1,
      • MAGE-A2,
      • MAGE-A3,
      • MAGE-C1, and/or
      • MAGE-C2, and
    • b) wherein the further antigen(s) is (are) selected from at least one antigen as defined herein, preferably in any of the herein mentioned combinations, groups or subgroups of antigens, e.g. the further antigen(s) is (are) selected from, e.g.:
  • hTERT,
      • WT1,
      • MAGE-A2,
      • 5T4,
      • MAGE-A3,
      • MUC1,
      • Her-2/neu,
      • NY-ESO-1,
      • CEA,
      • Survivin,
      • MAGE-C1, and/or
      • MAGE-C2.
  • According to a further preferred embodiment, the at least one antigen(s) according to a) is (are) selected from:
      • NY-ESO-1,
      • MAGE-C1, and/or
      • MAGE-C2.
  • According to another preferred embodiment, the at least one antigen(s) according to a) is (are) selected from:
      • MAGE-C1, and/or
      • MAGE-C2.
  • According to another preferred embodiment, the at least one antigen(s) according to b) is (are) selected from an antigen (antigens) as defined in one of the following combinations:
      • hTERT and WT1; or
      • hTERT and MAGE-A2; or
      • hTERT and 5T4; or
      • hTERT and MAGE-A3; or
      • hTERT and MUC1; or
      • hTERT and Her-2/neu; or
      • hTERT and NY-ESO-1; or
      • hTERT and CEA; or
      • hTERT and Survivin; or
      • hTERT and MAGE-C1; or
      • hTERT and MAGE-C2; or
      • WT1 and MAGE-A2; or
      • WT1 and 5T4; or
      • WT1 and MAGE-A3; or
      • WT1 and MUC1; or
      • WT1 and Her-2/neu; or
      • WT1 and NY-ESO-1; or
      • WT1 and CEA; or
      • WT1 and Survivin; or
      • WT1 and MAGE-C1; or
      • WT1 and MAGE-C2; or
      • MAGE-A2 and 5T4; or
      • MAGE-A2 and MAGE-A3; or
      • MAGE-A2 and MUC1; or
      • MAGE-A2 and Her-2/neu; or
      • MAGE-A2 and NY-ESO-1; or
      • MAGE-A2 and CEA; or
      • MAGE-A2 and Survivin; or
      • MAGE-A2 and MAGE-C1; or
      • MAGE-A2 and MAGE-C2; or
      • 5T4 and MAGE-A3; or
      • 5T4 and MUC1; or
      • 5T4 and Her-2/neu; or
      • 5T4 and NY-ESO-1; or
      • 5T4 and CEA; or
      • 5T4 and Survivin; or
      • 5T4 and MAGE-C1; or
      • 5T4 and MAGE-C2; or
      • MAGE-A3 and MUC1; or
      • MAGE-A3 and Her-2/neu; or
      • MAGE-A3 and NY-ESO-1; or
      • MAGE-A3 and CEA; or
      • MAGE-A3 and Survivin; or
      • MAGE-A3 and MAGE-C1
      • MAGE-A3 and MAGE-C2
      • MUC1 and Her-2/neu; or
      • MUC1 and NY-ESO-1; or
      • MUC1 and CEA; or
      • MUC1 and Survivin; or
      • MUC1 and MAGE-C1; or
      • MUC1 and MAGE-C2; or
      • HER-2/NEU and NY-ESO-1; or
      • HER-2/NEU and CEA; or
      • HER-2/NEU and Survivin; or
      • HER-2/NEU and MAGE-C 1; or
      • HER-2/NEU and MAGE-C2; or
      • NY-ESO-1 and CEA; or
      • NY-ESO-1 and Survivin; or
      • NY-ESO-1 and MAGE-C1; or
      • NY-ESO-1 and MAGE-C2; or
      • CEA and Survivin; or
      • CEA and MAGE-C1; or
      • CEA and MAGE-C2; or
      • Survivin and MAGE-C 1; or
      • Survivin and MAGE-C2; or
      • MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1 and MAGE-A2; or
      • hTERT, WT1 and 5T4; or
      • hTERT, WT1 and MAGE-A3; or
      • hTERT, WT1 and MUC1; or
      • hTERT, WT1 and Her-2/neu; or
      • hTERT, WT1 and NY-ESO-1; or
      • hTERT, WT1 and CEA; or
      • hTERT, WT1 and Survivin; or
      • hTERT, WT1 and MAGE-C1; or
      • hTERT, WT1 and MAGE-C2; or
      • WT1, MAGE-A2 and 5T4; or
      • WT1, MAGE-A2 and MAGE-A3; or
      • WT1, MAGE-A2 and MUC1; or
      • WT1, MAGE-A2 and Her-2/neu; or
      • WT1, MAGE-A2 and NY-ESO-1; or
      • WT1, MAGE-A2 and CEA; or
      • WT1, MAGE-A2 and Survivin; or
      • WT1, MAGE-A2 and MAGE-C1; or
      • WT1, MAGE-A2 and MAGE-C2; or
      • MAGE-A2, 5T4 and MAGE-A3; or
      • MAGE-A2, 5T4 and MUC1; or
      • MAGE-A2, 5T4 and Her-2/neu; or
      • MAGE-A2, 5T4 and NY-ESO-1; or
      • MAGE-A2, 5T4 and CEA; or
      • MAGE-A2, 5T4 and Survivin; or
      • MAGE-A2, 5T4 and MAGE-C1; or
      • MAGE-A2, 5T4 and MAGE-C2; or
      • 5T4, MAGE-A3 and MUC1; or
      • 5T4, MAGE-A3 and Her-2/neu; or
      • 5T4, MAGE-A3 and NY-ESO-1; or
      • 5T4, MAGE-A3 and CEA; or
      • 5T4, MAGE-A3 and Survivin; or
      • 5T4, MAGE-A3 and MAGE-C1; or
      • 5T4, MAGE-A3 and MAGE-C2; or
      • MAGE-A3, MUC1 and Her-2/neu; or
      • MAGE-A3, MUC1 and NY-ESO-1; or
      • MAGE-A3, MUC1 and CEA; or
      • MAGE-A3, MUC1 and Survivin; or
      • MAGE-A3, MUC1 and MAGE-C1; or
      • MAGE-A3, MUC1 and MAGE-C2; or
      • MUC1, Her-2/neu and NY-ESO-1; or
      • MUC1, Her-2/neu and CEA; or
      • MUC1, Her-2/neu and Survivin; or
      • MUC1, Her-2/neu and MAGE-C 1; or
      • MUC1, Her-2/neu and MAGE-C2; or
      • HER-2/NEU, NY-ESO-1 and CEA; or
      • HER-2/NEU, NY-ESO-1 and Survivin; or
      • HER-2/NEU, NY-ESO-1 and MAGE-C1; or
      • HER-2/NEU, NY-ESO-1 and MAGE-C2; or
      • NY-ESO-1, CEA and Survivin; or
      • NY-ESO-1, CEA and MAGE-C1; or
      • NY-ESO-1, CEA and MAGE-C2; or
      • CEA, Survivin and MAGE-C 1; or
      • CEA, Survivin and MAGE-C2; or
      • Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2 and 5T4; or
      • hTERT, WT1, MAGE-A2 and MAGE-A3; or
      • hTERT, WT1, MAGE-A2 and MUC1; or
      • hTERT, WT1, MAGE-A2 and Her-2/neu; or
      • hTERT, WT1, MAGE-A2 and NY-ESO-1; or
      • hTERT, WT1, MAGE-A2 and CEA; or
      • hTERT, WT1, MAGE-A2 and Survivin; or
      • hTERT, WT1, MAGE-A2 and MAGE-C1; or
      • hTERT, WT1, MAGE-A2 and MAGE-C2; or
      • WT1, MAGE-A2, 5T4 and MAGE-A3; or
      • WT1, MAGE-A2, 5T4 and MUC1; or
      • WT1, MAGE-A2, 5T4 and Her-2/neu; or
      • WT1, MAGE-A2, 5T4 and NY-ESO-1; or
      • WT1, MAGE-A2, 5T4 and CEA; or
      • WT1, MAGE-A2, 5T4 and Survivin; or
      • WT1, MAGE-A2, 5T4 and MAGE-C1; or
      • WT1, MAGE-A2, 5T4 and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3 and MUC1; or
      • MAGE-A2, 5T4, MAGE-A3 and Her-2/neu; or
      • MAGE-A2, 5T4, MAGE-A3 and NY-ESO-1; or
      • MAGE-A2, 5T4, MAGE-A3 and CEA; or
      • MAGE-A2, 5T4, MAGE-A3 and Survivin; or
      • MAGE-A2, 5T4, MAGE-A3 and MAGE-C1; or
      • MAGE-A2, 5T4, MAGE-A3 and MAGE-C2; or
      • 5T4, MAGE-A3, MUC1, and Her-2/neu; or
      • 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
      • 5T4, MAGE-A3, MUC1 and CEA; or
      • 5T4, MAGE-A3, MUC1 and Survivin; or
      • 5T4, MAGE-A3, MUC1 and MAGE-C1; or
      • 5T4, MAGE-A3, MUC1 and MAGE-C2; or
      • MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
      • MAGE-A3, MUC1, Her-2/neu and CEA; or
      • MAGE-A3, MUC1, Her-2/neu and Survivin; or
      • MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
      • MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
      • MUC1, Her-2/neu, NY-ESO-1 and CEA; or
      • MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
      • MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
      • MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
      • HER-2/NEU, NY-ESO-1, CEA and Survivin; or
      • HER-2/NEU, NY-ESO-1, CEA and MAGE-C1; or
      • HER-2/NEU, NY-ESO-1, CEA and MAGE-C2; or
      • NY-ESO-1, CEA, Survivin and MAGE-C1; or
      • NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4 and MAGE-A3; or
      • hTERT, WT1, MAGE-A2, 5T4 and MUC1; or
      • hTERT, WT1, MAGE-A2, 5T4 and Her-2/neu; or
      • hTERT, WT1, MAGE-A2, 5T4 and NY-ESO-1; or
      • hTERT, WT1, MAGE-A2, 5T4 and CEA; or
      • hTERT, WT1, MAGE-A2, 5T4 and Survivin; or
      • hTERT, WT1, MAGE-A2, 5T4 and MAGE-C1; or
      • hTERT, WT1, MAGE-A2, 5T4 and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and MUC1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and Her-2/neu; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and NY-ESO-1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and CEA; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and Survivin; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1 and Her-2/neu; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1 and CEA; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1 and Survivin; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C2; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
      • MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
      • MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
      • MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
      • HER-2/NEU, NY-ESO-1, CEA, Survivin and MAGE-C1; or
      • HER-2/NEU, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and MUC1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and Her-2/neu; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and NY-ESO-1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and CEA; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and Survivin; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Her-2/neu; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and CEA; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Survivin; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
      • MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
      • MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • HER-2/NEU, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Her-2/neu; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and CEA; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Survivin; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C 1; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C 1; or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
      • WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
      • or
      • hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2.
  • According to another particularly preferred embodiment, the at least one antigen(s) according to b) is (are) selected from the following specific combination of antigens as defined above:
      • Survivin and 5T4
  • The at least one RNA of the active (immunostimulatory) composition according to the present invention is typically any RNA, preferably, without being limited thereto, a coding RNA, a circular or linear RNA, a single- or a double-stranded RNA (which may also be regarded as a
  • RNA due to non-covalent association of two single-stranded RNA) or a partially double-stranded or partially single stranded RNA, which are at least partially self complementary (both of these partially double-stranded or partially single stranded RNA molecules are typically formed by a longer and a shorter single-stranded RNA molecule or by two single stranded RNA-molecules, which are about equal in length, wherein one single-stranded RNA molecule is in part complementary to the other single-stranded RNA molecule and both thus form a double-stranded RNA in this region, i.e. a partially double-stranded or partially single stranded RNA with respect to the entire RNA sequence). More preferably, the at least one RNA of the active (immunostimulatory) composition according to the present invention is a single-stranded RNA, even more preferably a linear RNA. Most preferably, the at least RNA of the active (immunostimulatory) composition according to the present invention is a messenger RNA (mRNA). In this context, a messenger RNA (mRNA) is typically a RNA, which is composed of (at least) several structural elements, e.g. an optional 5′-UTR region, an upstream positioned ribosomal binding site followed by a coding region, an optional 3′-UTR region, which may be followed by a poly-A tail (and/or a poly-C-tail).
  • Due to one particularly preferred embodiment, each of the at least two (preferably different) antigens of the active (immunostimulatory) composition of the present invention, may be encoded by one (monocistronic) RNA, preferably one (monocistronic) mRNA. In other words, the active (immunostimulatory) composition of the present invention may contain at least two (monocistronic) RNAs, preferably mRNAs, wherein each of these at least two (monocistronic) RNAs, preferably mRNAs, may encode just one (preferably different) antigen, selected from one of the above mentioned groups or subgroups, preferably in one of the above mentioned combinations.
  • According to another particularly preferred embodiment, the active (immunostimulatory) composition of the present invention, may comprise (at least) one bi- or even multicistronic RNA, preferably mRNA, i.e. (at least) one RNA which carries two or even more of the coding sequences of at the least two (preferably different) antigens, selected from one of the above mentioned groups or subgroups, preferably in one of the above mentioned combinations. Such coding sequences of the at least two (preferably different) antigens of the (at least) one bi- or even multicistronic RNA may be separated by at least one IRES (internal ribosomal entry site) sequence, as defined below. Thus, the term “encoding at least two (preferably different) antigens” may mean, without being limited thereto, that the (at least) one (bi- or even multicistronic) RNA, preferably a mRNA, may encode e.g. at least two, three, four, five, six, seven, eight, nine, ten, eleven or twelve (preferably different) antigens of the above mentioned group(s) of antigens or their fragments or variants within the above definitions. More preferably, without being limited thereto, the (at least) one (bi- or even multicistronic) RNA, preferably mRNA, may encode e.g. at least two, three, four, five or six (preferably different) antigens of the above mentioned subgroup(s) of antigens or their fragments or variants within the above definitions. In this context, a so-called IRES (internal ribosomal entry site) sequence as defined above can function as a sole ribosome binding site, but it can also serve to provide a bi- or even multicistronic RNA as defined above which codes for several proteins, which are to be translated by the ribosomes independently of one another. Examples of IRES sequences which can be used according to the invention are those from picornaviruses (e.g. FMDV), pestiviruses (CFFV), polioviruses (PV), encephalomyocarditis viruses (ECMV), foot and mouth disease viruses (FMDV), hepatitis C viruses (HCV), classical swine fever viruses (CSFV), mouse leukoma virus (MLV), simian immunodeficiency viruses (SIV) or cricket paralysis viruses (CrPV).
  • According to a further particularly preferred embodiment, the active (immunostimulatory) composition of the present invention, may comprise a mixture of at least one monocistronic RNA, preferably mRNA, as defined above, and at least one bi- or even multicistronic RNA, preferably mRNA, as defined above. The at least one monocistronic RNA and/or the at least one bi- or even multicistronic RNA preferably encode different antigens or their fragments or variants within the above definitions, the antigens preferably being selected from one of the above mentioned groups or subgroups of antigens, more preferably in one of the above mentioned combinations. However, the at least one monocistronic RNA and the at least one bi- or even multicistronic RNA may preferably also encode (in part) identical antigens selected from one of the above mentioned groups or subgroups of antigens, preferably in one of the above mentioned combinations, provided that the active (immunostimulatory) composition of the present invention as a whole provides at least two (preferably different) antigens as defined above. Such an embodiment may be advantageous e.g. for a staggered, e.g. time dependent, administration of the active (immunostimulatory) composition of the present invention to a patient in need thereof. The components of such an active (immunostimulatory) composition of the present invention, particularly the different RNAs encoding the at least two (preferably different) antigens, may be e.g. contained in (different parts of) a kit of parts composition or may be e.g. administered separately as components of different active (immunostimulatory) compositions according to the present invention.
  • Preferably, the at least one RNA of the active (immunostimulatory) composition, encoding at least two (preferably different) antigens selected from the above defined group or subgroup of antigens, more preferably in the above combinations, typically comprises a length of about 50 to about 20000, or 100 to about 20000 nucleotides, preferably of about 250 to about 20000 nucleotides, more preferably of about 500 to about 10000, even more preferably of about 500 to about 5000.
  • According to one embodiment, the at least one RNA of the active (immunostimulatory) composition, encoding at least two (preferably different) antigens selected from the above defined group(s) or subgroup(s) of antigens, more preferably in the above combinations, may be in the form of a modified RNA, wherein any modification, as defined herein, may be introduced into the at least one RNA of the active (immunostimulatory) composition. Modifications as defined herein preferably lead to a stabilized at least one RNA of the active (immunostimulatory) composition of the present invention.
  • According to a first embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention may thus be provided as a “stabilized RNA”, preferably a stabilized mRNA, that is to say as an (m)RNA that is essentially resistant to in vivo degradation (e.g. by an exo- or endo-nuclease). Such stabilization can be effected, for example, by a modified phosphate backbone of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention. A backbone modification in connection with the present invention is a modification in which phosphates of the backbone of the nucleotides contained in the RNA are chemically modified. Nucleotides that may be preferably used in this connection contain e.g. a phosphorothioate-modified phosphate backbone, preferably at least one of the phosphate oxygens contained in the phosphate backbone being replaced by a sulfur atom. Stabilized (m)RNAs may further include, for example: non-ionic phosphate analogues, such as, for example, alkyl and aryl phosphonates, in which the charged phosphonate oxygen is replaced by an alkyl or aryl group, or phosphodiesters and alkylphosphotriesters, in which the charged oxygen residue is present in alkylated form. Such backbone modifications typically include, without implying any limitation, modifications from the group consisting of methylphosphonates, phosphoramidates and phosphorothioates (e.g. cytidine-5′-O-(1-thiophosphate)).
  • The at least one RNA of the active (immunostimulatory) composition of the present invention may additionally or alternatively also contain sugar modifications. A sugar modification in connection with the present invention is a chemical modification of the sugar of the nucleotides of the at least one RNA and typically includes, without implying any limitation, sugar modifications selected from the group consisting of 2′-deoxy-2′-fluoro-oligoribonucleotide (2′-fluoro-2′-deoxycytidine-5′-triphosphate, 2′-fluoro-2′-deoxyuridine-5′-triphosphate), 2′-deoxy-2′-deamine oligoribonucleotide (2′-amino-2′-deoxycytidine-5′-triphosphate, 2′-amino-2′-deoxyuridine-5′-triphosphate), 2′-O-alkyl oligoribonucleotide, 2′-deoxy-2′-C-alkyl oligoribonucleotide (2′-O-methylcytidine-5′-triphosphate, 2′-methyluridine-5′-triphosphate), 2′-C-alkyl oligoribonucleotide, and isomers thereof (2′-aracytidine-5′-triphosphate, 2′-arauridine-5′-triphosphate), or azidotriphosphate (2′-azido-2′-deoxycytidine-5′-triphosphate, 2′-azido-2′-deoxyuridine-5′-triphosphate).
  • The at least one RNA of the active (immunostimulatory) composition of the present invention may additionally or alternatively also contain at least one base modification, which is preferably suitable for increasing the expression of the protein coded for by the at least one RNA sequence significantly as compared with the unaltered, i.e. natural (=native), RNA sequence. Significant in this case means an increase in the expression of the protein compared with the expression of the native RNA sequence by at least 20%, preferably at least 30%, 40%, 50% or 60%, more preferably by at least 70%, 80%, 90% or even 100% and most preferably by at least 150%, 200% or even 300% or more. In connection with the present invention, a nucleotide having such a base modification is preferably selected from the group of the base-modified nucleotides consisting of 2-amino-6-chloropurineriboside-5′-triphosphate, 2-aminoadenosine-5′-triphosphate, 2-thiocytidine-5′-triphosphate, 2-thiouridine-5′-triphosphate, 4-thiouridine-5′-triphosphate, 5-aminoallylcytidine-5′-triphosphate, 5-aminoallyluridine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, 5-bromouridine-5′-triphosphate, 5-iodocytidine-5′-triphosphate, 5-iodouridine-5′-triphosphate, 5-methylcytidine-5′-triphosphate, 5-methyluridine-5′-triphosphate, 6-azacytidine-5′-triphosphate, 6-azauridine-5′-triphosphate, 6-chloropurineriboside-5′-triphosphate, 7-deazaadenosine-5′-triphosphate, 7-deazaguanosine-5′-triphosphate, 8-azaadenosine-5′-triphosphate, 8-azidoadenosine-5′-triphosphate, benzimidazole-riboside-5′-triphosphate, N1-methyladenosine-5′-triphosphate, N1-methylguanosine-5′-triphosphate, N6-methyladenosine-5′-triphosphate, O6-methylguanosine-5′-triphosphate, pseudouridine-5′-triphosphate, or puromycin-5′-triphosphate, xanthosine-5′-triphosphate. Particular preference is given to nucleotides for base modifications selected from the group of base-modified nucleotides consisting of 5-methylcytidine-5′-triphosphate, 7-deazaguanosine-5′-triphosphate, 5-bromocytidine-5′-triphosphate, and pseudouridine-5′-triphosphate.
  • According to another embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention can likewise be modified (and preferably stabilized) by introducing further modified nucleotides containing modifications of their ribose or base moieties. Generally, the at least one (m)RNA of the active (immunostimulatory) composition of the present invention may contain any native (=naturally occurring) nucleotide, e.g. guanosine, uracil, adenosine, and/or cytosine or an analogue thereof. In this connection, nucleotide analogues are defined as non-natively occurring variants of naturally occurring nucleotides. Accordingly, analogues are chemically derivatized nucleotides with non-natively occurring functional groups, which are preferably added to or deleted from the naturally occurring nucleotide or which substitute the naturally occurring functional groups of a nucleotide. Accordingly, each component of the naturally occurring nucleotide may be modified, namely the base component, the sugar (ribose) component and/or the phosphate component forming the backbone (see above) of the RNA sequence. Analogues of guanosine, uracil, adenosine, and cytosine include, without implying any limitation, any naturally occurring or non-naturally occurring guanosine, uracil, adenosine, thymidine or cytosine that has been altered chemically, for example by acetylation, methylation, hydroxylation, etc., including 1-methyl-adenosine, 1-methyl-guanosine, 1-methyl-inosine, 2,2-dimethyl-guanosine, 2,6-diaminopurine, 2′-Amino-2′-deoxyadenosine, 2′-Amino-2′-deoxycytidine, 2′-Amino-2′-deoxyguanosine, 2′-Amino-2′-deoxyuridine, 2-Amino-6-chloropurineriboside, 2-Aminopurine-riboside, 2′-Araadenosine, 2′-Aracytidine, 2′-Arauridine, 2′-Azido-2′-deoxyadenosine, 2′-Azido-2′-deoxycytidine, 2′-Azido-2′-deoxyguanosine, 2′-Azido-2′-deoxyuridine, 2-Chloroadenosine, 2′-Fluoro-2′-deoxyadenosine, 2′-Fluoro-2′-deoxycytidine, 2′-Fluoro-2′-deoxyguanosine, 2′-Fluoro-2′-deoxyuridine, 2′-Fluorothymidine, 2-methyl-adenosine, 2-methyl-guanosine, 2-methyl-thio-N6-isopenenyl-adenosine, 2′-O-Methyl-2-aminoadenosine, 2′-O-Methyl-2′-deoxyadenosine, 2′-O-Methyl-2′-deoxycytidine, 2′-O-Methyl-2′-deoxyguanosine, 2′-O-Methyl-2′-deoxyuridine, 2′-O-Methyl-5-methyluridine, 2′-O-Methylinosine, 2′-O-Methylpseudouridine, 2-Thiocytidine, 2-thio-cytosine, 3-methyl-cytosine, 4-acetyl-cytosine, 4-Thiouridine, 5-(carboxyhydroxymethyl)-uracil, 5,6-Dihydrouridine, 5-Aminoallylcytidine, 5-Aminoallyl-deoxy-uridine, 5 -Bromouridine, 5 -carboxymehtyl aminom ethyl-2-thio-uracil, 5-carboxymethylamonomethyl-uracil, 5-Chloro-Ara-cytosine, 5-Fluoro-uridine, 5-Iodouridine, 5-methoxycarbonylmethyl-uridine, 5 -methoxy-uridine, 5 -methyl-2-thio-uridine, 6-Azacyti dine, 6-Azauridine, 6-Chloro-7-deaza-guanosine, 6-Chloropurineriboside, 6-Mercapto-guanosine, 6-Methyl-mercaptopurine-riboside, 7-Deaza-2′-deoxy-guanosine, 7-Deazaadenosine, 7-methyl-guanosine, 8-Azaadenosine, 8-Bromo-adenosine, 8-Bromo-guanosine, 8-Mercapto-guanosine, 8-Oxoguanosine, Benzimidazole-riboside, Beta-D-mannosyl-queosine, Dihydro-uracil, Inosine, N1-Methyladenosine, N6-([6-Aminohexyl]carbamoylmethyl)-adenosine, N6-isopentenyl-adenosine, N6-methyl-adenosine, N7-Methyl-xanthosine, N-uracil-5-oxyacetic acid methyl ester, Puromycin, Queosine, Uracil-5-oxyacetic acid, Uracil-5-oxyacetic acid methyl ester, Wybutoxosine, Xanthosine, and Xylo-adenosine. The preparation of such analogues is known to a person skilled in the art, for example from U.S. Pat. No. 4,373,071, U.S. Pat. No. 4,401,796, U.S. Pat. No. 4,415,732, U.S. Pat. No. 4,458,066, U.S. Pat. No. 4,500,707, U.S. Pat. No. 4,668,777, U.S. Pat. No. 4,973,679, U.S. Pat. No. 5,047,524, U.S. Pat. No. 5,132,418, U.S. Pat. No. 5,153,319, U.S. Pat. Nos. 5,262,530 and 5,700,642. In the case of an analogue as described above, particular preference may be given according to the invention to those analogues that increase the immunogenity of the RNA of the inventive active (immunostimulatory) composition and/or do not interfere with a further modification of the RNA that has been introduced.
  • According to a particular embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention can contain a lipid modification. Such a lipid-modified RNA typically comprises a RNA as defined herein, encoding at least two antigens selected from the group or subgroup of antigens as defined above, preferably in the above combinations. Such a lipid-modified RNA typically further comprises at least one linker covalently linked with that RNA, and at least one lipid covalently linked with the respective linker. Alternatively, the lipid-modified RNA comprises an at least one RNA as defined herein and at least one (bifunctional) lipid covalently linked (without a linker) with that RNA. According to a third alternative, the lipid-modified RNA comprises a RNA as defined herein, at least one linker covalently linked with that RNA, and at least one lipid covalently linked with the respective linker, and also at least one (bifunctional) lipid covalently linked (without a linker) with that RNA.
  • The lipid contained in the at least one RNA of the inventive active (immunostimulatory) composition (complexed or covalently bound thereto) is typically a lipid or a lipophilic residue that preferably is itself biologically active. Such lipids preferably include natural substances or compounds such as, for example, vitamins, e.g. alpha-tocopherol (vitamin E), including RRR-alpha-tocopherol (formerly D-alpha-tocopherol), L-alpha-tocopherol, the racemate D,L-alpha-tocopherol, vitamin E succinate (VES), or vitamin A and its derivatives, e.g. retinoic acid, retinol, vitamin D and its derivatives, e.g. vitamin D and also the ergosterol precursors thereof, vitamin E and its derivatives, vitamin K and its derivatives, e.g. vitamin K and related quinone or phytol compounds, or steroids, such as bile acids, for example cholic acid, deoxycholic acid, dehydrocholic acid, cortisone, digoxygenin, testosterone, cholesterol or thiocholesterol. Further lipids or lipophilic residues within the scope of the present invention include, without implying any limitation, polyalkylene glycols (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533), aliphatic groups such as, for example, C1-C20-alkanes, C1-C20-alkenes or C1-C20-alkanol compounds, etc., such as, for example, dodecanediol, hexadecanol or undecyl residues (Saison-Behmoaras et al., EMBO J, 1991, 10, 111; Kabanov et al., FEBS Lett., 1990, 259, 327; Svinarchuk et al., Biochimie, 1993, 75, 49), phospholipids such as, for example, phosphatidylglycerol, diacylphosphatidylglycerol, phosphatidylcholine, dipalmitoylphosphatidylcholine, distearoylphosphatidylcholine, phosphatidylserine, phosphatidylethanolamine, di-hexadecyl-rac-glycerol, sphingolipids, cerebrosides, gangliosides, or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651; Shea et al., Nucl. Acids Res., 1990, 18, 3777), polyamines or polyalkylene glycols, such as, for example, polyethylene glycol (PEG) (Manoharan et al., Nucleosides & Nucleotides, 1995, 14, 969), hexaethylene glycol (HEG), palmitin or palmityl residues (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229), octadecylamines or hexylamino-carbonyl-oxycholesterol residues (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923), and also waxes, terpenes, alicyclic hydrocarbons, saturated and mono- or poly-unsaturated fatty acid residues, etc.
  • The at least one RNA of the active (immunostimulatory) composition of the present invention may likewise be stabilized in order to prevent degradation of the RNA in vivo by various approaches. It is known in the art that instability and (fast) degradation of mRNA or of RNA in vivo in general may represent a serious problem in the application of RNA based compositions.
  • This instability of RNA is typically due to RNA-degrading enzymes, “RNAases” (ribonucleases), wherein contamination with such ribonucleases may sometimes completely degrade RNA in solution. Accordingly, the natural degradation of mRNA in the cytoplasm of cells is very finely regulated and RNase contaminations may be generally removed by special treatment prior to use of said compositions, in particular with diethyl pyrocarbonate (DEPC). A number of mechanisms of natural degradation are known in this connection in the prior art, which may be utilized as well. E.g., the terminal structure is typically of critical importance for a mRNA in vivo. As an example, at the 5′ end of naturally occurring mRNAs there is usually a so-called “cap structure” (a modified guanosine nucleotide), and at the 3′ end is typically a sequence of up to 200 adenosine nucleotides (the so-called poly-A tail) (SEQ ID NO: 29).
  • The at least one RNA of the active (immunostimulatory) composition of the present invention, particularly if provided as a mRNA, can therefore be stabilized against degradation by RNases by the addition of a so-called “5′ cap” structure. Particular preference is given in this connection to an m7G(5′)ppp (5′(A,G(5′)ppp(5′)A or G(5′)ppp(5′)G as the 5′ cap” structure. However, such a modification is introduced only if a modification, for example a lipid modification, has not already been introduced at the 5′ end of the (m)RNA of the inventive immunostimulatory composition or if the modification does not interfere with the immunogenic properties of the (unmodified or chemically modified) (m)RNA.
  • According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention may contain, especially if the RNA is in the form of a mRNA, a poly-A tail on the 3′ terminus of typically about 10 to 200 adenosine nucleotides (SEQ ID NO: 30), preferably about 10 to 100 adenosine nucleotides (SEQ ID NO: 31), more preferably about 20 to 100 adenosine nucleotides (SEQ ID NO: 32) or even more preferably about 40 to 80 adenosine nucleotides (SEQ ID NO: 33).
  • According to a further preferred embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention may contain, especially if the RNA is in the form of a mRNA, a poly-C tail on the 3′ terminus of typically about 10 to 200 cytosine nucleotides (SEQ ID NO: 34), preferably about 10 to 100 cytosine nucleotides (SEQ ID NO: 35), more preferably about 20 to 70 cytosine nucleotides (SEQ ID NO: 36) or even more preferably about 20 to 60 (SEQ ID NO: 37) or even 10 to 40 cytosine nucleotides (SEQ ID NO: 38).
  • According to another embodiment, the at least one RNA of the active (immunostimulatory) composition of the present invention may be modified, and thus stabilized, especially if the RNA is in the form of a mRNA, by modifying the G/C content of the RNA, preferably of the coding region of the at least one RNA.
  • In a particularly preferred embodiment of the present invention, the G/C content of the coding region of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is modified, particularly increased, compared to the G/C content of the coding region of its particular wild-type (m)RNA, i.e. the unmodified (m)RNA. The encoded amino acid sequence of the at least one (m)RNA is preferably not modified compared to the coded amino acid sequence of the particular wild-type (m)RNA.
  • This modification of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is based on the fact that the sequence of any (m)RNA region to be translated is important for efficient translation of that (m)RNA. Thus, the composition and the sequence of various nucleotides is important. In particular, sequences having an increased G (guanosine)/C (cytosine) content are more stable than sequences having an increased A (adenosine)/U (uracil) content. According to the invention, the codons of the (m)RNA are therefore varied compared to its wild-type (m)RNA, while retaining the translated amino acid sequence, such that they include an increased amount of G/C nucleotides. In respect to the fact that several codons code for one and the same amino acid (so-called degeneration of the genetic code), the most favorable codons for the stability can be determined (so-called alternative codon usage).
  • Depending on the amino acid to be encoded by the at least one (m)RNA, there are various possibilities for modification of the at least one (m)RNA sequence, compared to its wild-type sequence. In the case of amino acids which are encoded by codons which contain exclusively G or C nucleotides, no modification of the codon is necessary. Thus, the codons for Pro (CCC or CCG), Arg (CGC or CGG), Ala (GCC or GCG) and Gly (GGC or GGG) require no modification, since no A or U is present.
  • In contrast, codons which contain A and/or U nucleotides can be modified by substitution of other codons which code for the same amino acids but contain no A and/or U. Examples of these are:
  • the codons for Pro can be modified from CCU or CCA to CCC or CCG;
  • the codons for Arg can be modified from CGU or CGA or AGA or AGG to CGC or CGG;
  • the codons for Ala can be modified from GCU or GCA to GCC or GCG;
  • the codons for Gly can be modified from GGU or GGA to GGC or GGG.
  • In other cases, although A or U nucleotides cannot be eliminated from the codons, it is however possible to decrease the A and U content by using codons which contain a lower content of A and/or U nucleotides. Examples of these are:
  • the codons for Phe can be modified from UUU to UUC;
  • the codons for Leu can be modified from UUA, UUG, CUU or CUA to CUC or CUG;
  • the codons for Ser can be modified from UCU or UCA or AGU to UCC, UCG or AGC;
  • the codon for Tyr can be modified from UAU to UAC;
  • the codon for Cys can be modified from UGU to UGC;
  • the codon for His can be modified from CAU to CAC;
  • the codon for Gln can be modified from CAA to CAG;
  • the codons for Ile can be modified from AUU or AUA to AUC;
  • the codons for Thr can be modified from ACU or ACA to ACC or ACG;
  • the codon for Asn can be modified from AAU to AAC;
  • the codon for Lys can be modified from AAA to AAG;
  • the codons for Val can be modified from GUU or GUA to GUC or GUG;
  • the codon for Asp can be modified from GAU to GAC;
  • the codon for Glu can be modified from GAA to GAG;
  • the stop codon UAA can be modified to UAG or UGA.
  • In the case of the codons for Met (AUG) and Trp (UGG), on the other hand, there is no possibility of sequence modification.
  • The substitutions listed above can be used either individually or in all possible combinations to increase the G/C content of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention compared to its particular wild-type (m)RNA (i.e. the original sequence). Thus, for example, all codons for Thr occurring in the wild-type sequence can be modified to ACC (or ACG). Preferably, however, for example, combinations of the above substitution possibilities are used:
  • substitution of all codons coding for Thr in the original sequence (wild-type (m)RNA) to ACC (or ACG) and
  • substitution of all codons originally coding for Ser to UCC (or UCG or AGC);
  • substitution of all codons coding for Ile in the original sequence to AUC and
  • substitution of all codons originally coding for Lys to AAG and
  • substitution of all codons originally coding for Tyr to UAC;
  • substitution of all codons coding for Val in the original sequence to GUC (or GUG) and
  • substitution of all codons originally coding for Glu to GAG and
  • substitution of all codons originally coding for Ala to GCC (or GCG) and
  • substitution of all codons originally coding for Arg to CGC (or CGG);
  • substitution of all codons coding for Val in the original sequence to GUC (or GUG) and
  • substitution of all codons originally coding for Glu to GAG and
  • substitution of all codons originally coding for Ala to GCC (or GCG) and
  • substitution of all codons originally coding for Gly to GGC (or GGG) and
  • substitution of all codons originally coding for Asn to AAC;
  • substitution of all codons coding for Val in the original sequence to GUC (or GUG) and
  • substitution of all codons originally coding for Phe to UUC and
  • substitution of all codons originally coding for Cys to UGC and
  • substitution of all codons originally coding for Leu to CUG (or CUC) and
  • substitution of all codons originally coding for Gln to CAG and
  • substitution of all codons originally coding for Pro to CCC (or CCG); etc.
  • Preferably, the G/C content of the coding region of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is increased by at least 7%, more preferably by at least 15%, particularly preferably by at least 20%, compared to the G/C content of the coded region of the wild-type (m)RNA which codes for an antigen, antigenic protein or antigenic peptide as deinined herein or its fragment or variant thereof. According to a specific embodiment at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, more preferably at least 70%, even more preferably at least 80% and most preferably at least 90%, 95% or even 100% of the substitutable codons in the region coding for an antigen, antigenic protein or antigenic peptide as deinined herein or its fragment or variant thereof or the whole sequence of the wild type (m)RNA sequence are substituted, thereby increasing the GC/content of said sequence.
  • In this context, it is particularly preferable to increase the G/C content of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention to the maximum (i.e. 100% of the substitutable codons), in particular in the region coding for a protein, compared to the wild-type sequence.
  • According to the invention, a further preferred modification of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is based on the finding that the translation efficiency is also determined by a different frequency in the occurrence of tRNAs in cells. Thus, if so-called “rare codons” are present in the at least one (m)RNA of the active (immunostimulatory) composition of the present invention to an increased extent, the corresponding modified at least one (m)RNA sequence is translated to a significantly poorer degree than in the case where codons coding for relatively “frequent” tRNAs are present.
  • According to the invention, in the modified at least one (m)RNA of the active (immunostimulatory) composition of the present invention, the region which codes for the adjuvant protein is modified compared to the corresponding region of the wild-type (m)RNA such that at least one codon of the wild-type sequence which codes for a tRNA which is relatively rare in the cell is exchanged for a codon which codes for a tRNA which is relatively frequent in the cell and carries the same amino acid as the relatively rare tRNA. By this modification, the sequences of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is modified such that codons for which frequently occurring tRNAs are available are inserted. In other words, according to the invention, by this modification all codons of the wild-type sequence which code for a tRNA which is relatively rare in the cell can in each case be exchanged for a codon which codes for a tRNA which is relatively frequent in the cell and which, in each case, carries the same amino acid as the relatively rare tRNA.
  • Which tRNAs occur relatively frequently in the cell and which, in contrast, occur relatively rarely is known to a person skilled in the art; cf. e.g. Akashi, Curr. Opin. Genet. Dev. 2001, 11(6): 660-666. The codons which use for the particular amino acid the tRNA which occurs the most frequently, e.g. the Gly codon, which uses the tRNA which occurs the most frequently in the (human) cell, are particularly preferred.
  • According to the invention, it is particularly preferable to link the sequential G/C content which is increased, in particular maximized, in the modified at least one (m)RNA of the active (immunostimulatory) composition of the present invention, with the “frequent” codons without modifying the amino acid sequence of the protein encoded by the coding region of the (m)RNA. This preferred embodiment allows provision of a particularly efficiently translated and stabilized (modified) at least one (m)RNA of the active (immunostimulatory) composition of the present invention.
  • The determination of a modified at least one (m)RNA of the active (immunostimulatory) composition of the present invention as described above (increased G/C content; exchange of tRNAs) can be carried out using the computer program explained in WO 02/098443—the disclosure content of which is included in its full scope in the present invention. Using this computer program, the nucleotide sequence of any desired (m)RNA can be modified with the aid of the genetic code or the degenerative nature thereof such that a maximum G/C content results, in combination with the use of codons which code for tRNAs occurring as frequently as possible in the cell, the amino acid sequence coded by the modified at least one (m)RNA preferably not being modified compared to the non-modified sequence. Alternatively, it is also possible to modify only the G/C content or only the codon usage compared to the original sequence. The source code in Visual Basic 6.0 (development environment used: Microsoft Visual Studio Enterprise 6.0 with Servicepack 3) is also described in WO 02/098443.
  • In a further preferred embodiment of the present invention, the A/U content in the environment of the ribosome binding site of the at least one (m)RNA of the active (immunostimulatory) composition of the present invention is increased compared to the A/U content in the environment of the ribosome binding site of its particular wild-type (m)RNA. This modification (an increased A/U content around the ribosome binding site) increases the efficiency of ribosome binding to the at least one (m)RNA. An effective binding of the ribosomes to the ribosome binding site (Kozak sequence: GCCGCCACCAUGG (SEQ ID NO: 27), the AUG forms the start codon) in turn has the effect of an efficient translation of the at least one (m)RNA.
  • According to a further embodiment of the present invention the at least one (m)RNA of the active (immunostimulatory) composition of the present invention may be modified with respect to potentially destabilizing sequence elements. Particularly, the coding region and/or the 5′ and/or 3′ untranslated region of this at least one (m)RNA may be modified compared to the particular wild-type (m)RNA such that is contains no destabilizing sequence elements, the coded amino acid sequence of the modified at least one (m)RNA preferably not being modified compared to its particular wild-type (m)RNA. It is known that, for example, in sequences of eukaryotic RNAs destabilizing sequence elements (DSE) occur, to which signal proteins bind and regulate enzymatic degradation of RNA in vivo. For further stabilization of the modified at least one (m)RNA, optionally in the region which encodes for an antigen, antigenic protein or antigenic peptide as defined herein, one or more such modifications compared to the corresponding region of the wild-type (m)RNA can therefore be carried out, so that no or substantially no destabilizing sequence elements are contained there. According to the invention, DSE present in the untranslated regions (3′- and/or 5′-UTR) can also be eliminated from the at least one (m)RNA of the active (immunostimulatory) composition of the present invention by such modifications.
  • Such destabilizing sequences are e.g. AU-rich sequences (AURES), which occur in 3′-UTR sections of numerous unstable RNAs (Caput et al., Proc. Natl. Acad. Sci. USA 1986, 83: 1670 to 1674). The at least one (m)RNA of the active (immunostimulatory) composition of the present invention is therefore preferably modified compared to the wild-type (m)RNA such that the at least one (m)RNA contains no such destabilizing sequences. This also applies to those sequence motifs which are recognized by possible endonucleases, e.g. the sequence GAACAAG, which is contained in the 3′-UTR segment of the gene which codes for the transferrin receptor (Binder et al., EMBO J. 1994, 13: 1969 to 1980). These sequence motifs are also preferably removed in the at least one (m)RNA of the active (immunostimulatory) composition of the present invention.
  • Also preferably according to the invention, the at least one (m)RNA of the active (immunostimulatory) composition of the present invention has, in a modified form, at least one IRES as defined above and/or at least one 5′ and/or 3′ stabilizing sequence, in a modified form, e.g. to enhance ribosome binding or to allow expression of different encoded antigens located on an at least one (bi- or even multicistronic) RNA of the active (immunostimulatory) composition of the present invention.
  • According to the invention, the at least one (m)RNA of the active (immunostimulatory) composition of the present invention furthermore preferably has at least one 5′ and/or 3′ stabilizing sequence. These stabilizing sequences in the 5′ and/or 3′ untranslated regions have the effect of increasing the half-life of the at least one (m)RNA in the cytosol. These stabilizing sequences can have 100% sequence homology to naturally occurring sequences which occur in viruses, bacteria and eukaryotes, but can also be partly or completely synthetic. The untranslated sequences (UTR) of the βglobin gene, e.g. from Homo sapiens or Xenopus laevis may be mentioned as an example of stabilizing sequences which can be used in the present invention for a stabilized RNA. Another example of a stabilizing sequence has the general formula (C/U)CCANxCCC(U/A)PyxUC(C/U)CC (SEQ ID NO: 28), which is contained in the 3′UTR of the very stable RNA which codes for aglobin, α(I)-collagen, 15-lipoxygenase or for tyrosine hydroxylase (cf. Holcik et al., Proc. Natl. Acad. Sci. USA 1997, 94: 2410 to 2414). Such stabilizing sequences can of course be used individually or in combination with one another and also in combination with other stabilizing sequences known to a person skilled in the art. The at least one (m)RNA of the active (immunostimulatory) composition of the present invention is therefore preferably present as globin UTR (untranslated regions)-stabilized RNA, in particular as βglobin UTR-stabilized RNA.
  • Nevertheless, substitutions, additions or eliminations of bases are preferably carried out with the at least one RNA of the active (immunostimulatory) composition of the present invention, using a DNA matrix for preparation of the at least one RNA of the active (immunostimulatory) composition of the present invention by techniques of the well known site directed mutagenesis or with an oligonucleotide ligation strategy (see e.g. Maniatis et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, 3rd ed., Cold Spring Harbor, N.Y., 2001). In such a process, for preparation of the at least one (m)RNA, a corresponding DNA molecule may be transcribed in vitro. This DNA matrix preferably comprises a suitable promoter, e.g. a T7 or SP6 promoter, for in vitro transcription, which is followed by the desired nucleotide sequence for the at least one RNA to be prepared and a termination signal for in vitro transcription. The DNA molecule, which forms the matrix of an at least one RNA of interest, may be prepared by fermentative proliferation and subsequent isolation as part of a plasmid which can be replicated in bacteria. Plasmids which may be mentioned as suitable for the present invention are e.g. the plasmids pT7Ts (GENBANK ® accession number U26404; Lai et al., Development 1995, 121: 2349 to 2360), pGEM® series, e.g. pGEM®-1 (GENBANK accession number X65300; from Promega) and pSP64 (GENBANK® accession number X65327); cf. also Mezei and Storts, Purification of PCR Products, in: Griffin and Griffin (ed.), PCR Technology: Current Innovation, CRC Press, Boca Raton, Fla., 2001.
  • The stabilization of the at least one RNA of the active (immunostimulatory) composition of the present invention can likewise by carried out by associating or complexing the at least one RNA with, or binding it to, a cationic compound, in particular a polycationic compound, for example a (poly)cationic peptide or protein. In particular the use of protamine, nucleoline, spermin or spermidine as the polycationic, nucleic-acid-binding protein to the RNA is particularly effective. Furthermore, the use of other cationic peptides or proteins, such as poly-L-lysine or histones, is likewise possible. This procedure for stabilizing RNA is described in EP-A-1083232, the disclosure of which is incorporated by reference into the present invention in its entirety. Further preferred cationic substances which can be used for stabilizing the RNA of the active (immunostimulatory) composition of the present invention include cationic polysaccharides, for example chitosan, polybrene, polyethyleneimine (PEI) or poly-L-lysine (PLL), etc. Association or complexing of the at least one RNA of the inventive active (immunostimulatory) composition with cationic compounds, e.g. cationic proteins or cationic lipids, e.g. oligofectamine as a lipid based complexation reagent) preferably increases the transfer of the at least one RNA present as a pharmaceutically active component into the cells to be treated or into the organism to be treated. It is also referred to the disclosure herein with regard to the stabilizing effect for the at least one RNA of the active (immunostimulatory) composition of the present invention by complexation, which holds for the stabilization of RNA as well.
  • According to another particularly preferred embodiment, the at least RNA of the active (immunostimulatory) composition may additionally or alternatively encode a secretory signal peptide. Such signal peptides are sequences, which typically exhibit a length of about 15 to 30 amino acids and are preferably located at the N-terminus of the encoded peptide, without being limited thereto. Signal peptides as defined herein preferably allow the transport of the antigen, antigenic protein or antigenic peptide as encoded by the at least one RNA of the active (immunostimulatory) composition into a defined cellular compartiment, preferably the cell surface, the endoplasmic reticulum (ER) or the endosomal-lysosomal compartiment. Examples of secretory signal peptide sequences as defined herein include, without being limited thereto, signal sequences of classical or non-classical MHC-molecules (e.g. signal sequences of MHC I and II molecules, e.g. of the MHC class I molecule HLA-A*0201), signal sequences of cytokines or immunoglobulines as defined herein, signal sequences of the invariant chain of immunoglobulines or antibodies as defined herein, signal sequences of Lamp1, Tapasin, Erp57, Calretikulin, Calnexin, and further membrane associated proteins or of proteins associated with the endoplasmic reticulum (ER) or the endosomal-lysosomal compartiment. Particularly preferably, signal sequences of MHC class I molecule HLA-A*0201 may be used according to the present invention.
  • Any of the above modifications may be applied to the at least one RNA of the active (immunostimulatory) composition of the present invention, and further to any (m)RNA as used in the context of the present invention and may be, if suitable or necessary, be combined with each other in any combination, provided, these combinations of modifications do not interfere with each other in the respective at least one RNA. A person skilled in the art will be able to take his choice accordingly.
  • According to another embodiment, the active (immunostimulatory) composition according to the invention may comprise an adjuvant. In this context, an adjuvant may be understood as any compound, which is suitable to support administration and delivery of the active (immunostimulatory) composition according to the invention. Furthermore, such an adjuvant may, without being bound thereto, initiate or increase an immune response of the innate immune system, i.e. a non-specific immune response. With other words, when administered, the active (immunostimulatory) composition according to the invention typically initiates an adaptive immune response due to the at least two antigens encoded by the at least one RNA contained in the inventive active (immunostimulatory) composition. Additionally, the active (immunostimulatory) composition according to the invention may generate an (supportive) innate immune response due to addition of an adjuvant as defined herein to the active (immunostimulatory) composition according to the invention. Such an adjuvant may be selected from any adjuvant known to a skilled person and suitable for the present case, i.e. supporting the induction of an immune response in a mammal. Preferably, the adjuvant may be selected from the group consisting of, without being limited thereto, TDM, MDP, muramyl dipeptide, pluronics, alum solution, aluminium hydroxide, ADJUMER™ (polyphosphazene); aluminium phosphate gel; glucans from algae; algammulin; aluminium hydroxide gel (alum); highly protein-adsorbing aluminium hydroxide gel; low viscosity aluminium hydroxide gel; AF or SPT (emulsion of squalane (5%), Tween 80 (0.2%), Pluronic L121 (1.25%), phosphate-buffered saline, pH 7.4); AVRIDINE™ (propanediamine); BAY R1005™ ((N-(2-deoxy-2-L-leucylamino-b-D-glucopyranosyl)-N-octadecyl-dodecanoyl-amide hydroacetate); CALCITRIOL™ (1-alpha,25-dihydroxy-vitamin D3); calcium phosphate gel; CAP™ (calcium phosphate nanoparticles); cholera holotoxin, cholera-toxin-A1-protein-A-D-fragment fusion protein, sub-unit B of the cholera toxin; CRL 1005 (block copolymer P1205); cytokine-containing liposomes; DDA (dimethyldioctadecylammonium bromide); DHEA (dehydroepiandrosterone); DMPC (dimyristoylphosphatidylcholine); DMPG (dimyristoylphosphatidylglycerol); DOC/alum complex (deoxycholic acid sodium salt); Freund's complete adjuvant; Freund's incomplete adjuvant; gamma inulin; Gerbu adjuvant (mixture of: i) N-acetylglucosaminyl-(P1-4)-N-acetylmuramyl-L-alanyl-D-glutamine (GMDP), ii) dimethyldioctadecylammonium chloride (DDA), iii) zinc-L-proline salt complex (ZnPro-8); GM-C SF); GMDP (N-acetylglucosaminyl-(b1-4)-N-acetylmuramyl-L-alanyl-D-isoglutamine); imiquimod (1-(2-methypropyl)-1H-imidazo[4,5-c]quinoline-4-amine); ImmTher™ (N-acetylglucosaminyl-N-acetylmuramyl-L-Ala-D-isoGlu-L-Ala-glycerol dipalmitate); DRVs (immunoliposomes prepared from dehydration-rehydration vesicles); interferon-gamma; interleukin-1beta; interleukin-2; interleukin-7; interleukin-12; ISCOMS™; ISCOPREP 7.0.3.™; liposomes; LOXORIBINE™ (7-allyl-8-oxoguanosine); LT oral adjuvant (E.coli labile enterotoxin-protoxin); microspheres and microparticles of any composition; MF59™; (squalene-water emulsion); MONTANIDEISA 51™(purified incomplete Freund's adjuvant); MONTANIDEISA 720™ (metabolisable oil adjuvant); MPL™ (3-Q-desacyl-4′-monophosphoryl lipid A); MTP-PE and MTP-PE liposomes ((N-acetyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1,2-dipalmitoyl-sn-glycero-3-(hydroxyphosphoryloxy))-ethylamide, monosodium salt); MURAMETIDE™ (Nac-Mur-L-Ala-D-Gln-OCH3); MURAPALMITINE™ and D-MURAPALMITINE™ (Nac-Mur-L-Thr-D-isoGIn-sn-glyceroldipalmitoyl); NAGO (neuraminidase-galactose oxidase); nanospheres or nanoparticles of any composition; NISVs (non-ionic surfactant vesicles); PLEURAN™ ((3-glucan); PLGA, PGA and PLA (homo- and co-polymers of lactic acid and glycolic acid; microspheres/nanospheres); PLURONIC L121™; PMMA (polymethyl methacrylate); PODDS™ (proteinoid microspheres); polyethylene carbamate derivatives; poly-rA: poly-rU (polyadenylic acid-polyuridylic acid complex); polysorbate 80 (Tween 80); protein cochleates (Avanti Polar Lipids, Inc., Alabaster, Ala.); STIMULON™ (QS-21); Quil-A (Quil-A saponin); S-28463 (4-amino-otec-dimethyl-2-ethoxymethyl-1H-imidazo[4,5-c]quinoline-1-ethanol); SAF-1™ (“Syntex adjuvant formulation”); Sendai proteoliposomes and Sendai-containing lipid matrices; Span-85 (sorbitan trioleate); Specol (emulsion of Marcol 52, Span 85 and Tween 85); squalene or Robane® (2,6, 10,15,19,23 -hexamethyltetracosan and 2,6,10,15,19,23 -hexamethyl-2,6, 10,14,18,22-tetracosahexane); stearyltyrosine (octadecyltyrosine hydrochloride); Theramid° (N-acetylglucosaminyl-N-acetylmuramyl-L-Ala-D-isoGlu-L-Ala-dipalmitoxypropylamide); Theronyl-MDP (Termurtide™ or [thr 1]-MDP; N-acetylmuramyl-L-threonyl-D-isoglutamine); Ty particles (Ty-VLPs or virus-like particles); Walter-Reed liposomes (liposomes containing lipid A adsorbed on aluminium hydroxide), and lipopeptides, including Pam3Cys, in particular aluminium salts, such as Adju-phos, Alhydrogel, Rehydragel; emulsions, including CFA, SAF, IFA, MF59, Provax, TiterMax, Montanide, Vaxfectin; copolymers, including Optivax (CRL1005), L121, Poloaxmer4010), etc.; liposomes, including Stealth, cochleates, including BIORAL; plant derived adjuvants, including QS21, Quil A, Iscomatrix, ISCOM; adjuvants suitable for costimulation including Tomatine, biopolymers, including PLG, PMM, Inulin; microbe derived adjuvants, including Romurtide, DETOX, MPL, CWS, Mannose, CpG nucleic acid sequences, CpG7909, ligands of human TLR 1-10, ligands of murine TLR 1-13, ISS-1018, IC31, Imidazoquinolines, Ampligen, Ribi529, IMOxine, IRIVs, VLPs, cholera toxin, heat-labile toxin, Pam3Cys, Flagellin, GPI anchor, LNFPIII/Lewis X, antimicrobial peptides, UC-1V150, RSV fusion protein, cdiGMP; and adjuvants suitable as antagonists including CGRP neuropeptide.
  • Suitable adjuvants may also be selected from cationic or polycationic compounds wherein the adjuvant is preferably prepared upon complexing the at least one RNA of the inventive active (immmunostimulatory composition) with the cationic or polycationic compound. Association or complexing the RNA of the active (immunostimulatory) composition with cationic or polycationic compounds as defined herein preferably provides adjuvant properties and confers a stabilizing effect to the at least one RNA of the active (immunostimulatory) composition.
  • Particularly such preferred, such cationic or polycationic compounds are selected from cationic or polycationic peptides or proteins, including protamine, nucleoline, spermin or spermidine, or other cationic peptides or proteins, such as poly-L-lysine (PLL), poly-arginine, basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, Tat, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs, PpT620, prolin-rich peptides, arginine-rich peptides, lysine-rich peptides, MPG-peptide(s), Pep-1, L-oligomers, Calcitonin peptide(s), Antennapedia-derived peptides (particularly from Drosophila antennapedia), pAntp, pIsl, FGF, Lactoferrin, Transportan, Buforin-2, Bac715-24, SynB, SynB(1), pVEC, hCT-derived peptides, SAP, protamine, spermine, spermidine, or histones. Further preferred cationic or polycationic compounds may include cationic polysaccharides, for example chitosan, polybrene, cationic polymers, e.g. polyethyleneimine (PEI), cationic lipids, e.g. DOTMA: [1-(2,3-sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride, DMRIE, di-C14-amidine, DOTIM, SAINT, DC-Chol, BGTC, CTAP, DOPC, DODAP, DOPE: Dioleyl phosphatidylethanol-amine, DOSPA, DODAB, DOIC, DMEPC, DOGS: Dioctadecylamidoglicylspermin, DIMRI: Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide, DOTAP: dioleoyloxy-3-(trimethylammonio)propane, DC-6-14: 0,0-ditetradecanoyl-N-(α-trimethylammonioacetyl)diethanolamine chloride, CLIP1: rac-[(2,3-dioctadecyloxypropyl)(2-hydroxyethyl)]-dimethylammonium chloride, CLIP6: rac-[2(2,3-dihexadecyloxypropyl-oxymethyloxy)ethyl]trimethylammonium, CLIPS: rac-[2(2,3 -dihexadecyloxypropyl-oxysuccinyloxy)ethyl]-trimethylammonium, oligofectamine, or cationic or polycationic polymers, e.g. modified polyaminoacids, such as 13-aminoacid-polymers or reversed polyamides, etc., modified polyethylenes, such as PVP (poly(N-ethyl-4-vinylpyridinium bromide)), etc., modified acrylates, such as pDMAEMA (poly(dimethylaminoethyl methylacrylate)), etc., modified Amidoamines such as pAMAM (poly(amidoamine)), etc., modified polybetaaminoester (PBAE), such as diamine end modified 1,4 butanediol diacrylate-co-5-amino-1-pentanol polymers, etc., dendrimers, such as polypropylamine dendrimers or pAMAM based dendrimers, etc., polyimine(s), such as PEI: poly(ethyleneimine), poly(propyleneimine), etc., polyallylamine, sugar backbone based polymers, such as cyclodextrin based polymers, dextran based polymers, Chitosan, etc., silan backbone based polymers, such as PMOXA-PDMS copolymers, etc., Blockpolymers consisting of a combination of one or more cationic blocks (e.g. selected of a cationic polymer as mentioned above) and of one or more hydrophilic- or hydrophobic blocks (e.g. polyethyleneglycole); etc.
  • Additionally, preferred cationic or polycationic proteins or peptides, which can be used as an adjuvant by complexing the at least one RNA of the active (immunostimulatory) composition, may be selected from following proteins or peptides having the following total formula (I): (Arg)l (SEQ ID NO: 39); (Lys)m (SEQ ID NO: 40); (His)n (SEQ ID NO: 41); (Orn)o;(Xaa)x, wherein 1+m+n+o+x=8-15, and 1, m, n or o independently of each other may be any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15, provided that the overall content of Arg, Lys, His and Orn represents at least 50% of all amino acids of the oligopeptide; and Xaa may be any amino acid selected from native (=naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x may be any number selected from 0, 1, 2, 3 or 4, provided, that the overall content of Xaa does not exceed 50% of all amino acids of the oligopeptide. Particularly preferred oligoarginines in this context are e.g. Arg7 (SEQ ID NO: 42), Arg8 (SEQ ID NO: 43), Arg9 (SEQ ID NO: 44), Arg7 (SEQ ID NO: 42), H3R9 (SEQ ID NO: 45), R9H3 (SEQ ID NO: 46), H3R9H3 (SEQ ID NO: 47), YSSR9SSY (SEQ ID NO: 48), (RKH)4 (SEQ ID NO: 49), Y(RKH)2R (SEQ ID NO: 50), etc.
  • Suitable adjuvants may furthermore be selected from nucleic acids having the formula (II): GlXmGn, (SEQ ID NO: 51) wherein: G is guanosine, uracil or an analogue of guanosine or uracil;
  • X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above-mentioned nucleotides; 1 is an integer from 1 to 40, wherein when 1=1 G is guanosine or an analogue thereof, when 1>1 at least 50% of the nucleotides are guanosine or an analogue thereof; m is an integer and is at least 3; wherein when m=3 X is uracil or an analogue thereof, when m>3 at least 3 successive uracils or analogues of uracil occur; n is an integer from 1 to 40, wherein when n=1 G is guanosine or an analogue thereof, when n>1 at least 50% of the nucleotides are guanosine or an analogue thereof.
  • Other suitable adjuvants may furthermore be selected from nucleic acids having the formula (III): ClXmCn (SEQ ID NO: 52), wherein: C is cytosine, uracil or an analogue of cytosine or uracil; X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above-mentioned nucleotides; 1 is an integer from 1 to 40, wherein when 1=1 C is cytosine or an analogue thereof, when 1>1 at least 50% of the nucleotides are cytosine or an analogue thereof; m is an integer and is at least 3; wherein when m=3 X is uracil or an analogue thereof, when m>3 at least 3 successive uracils or analogues of uracil occur; n is an integer from 1 to 40, wherein when n=1 C is cytosine or an analogue thereof, when n>1 at least 50% of the nucleotides are cytosine or an analogue thereof.
  • According to one preferred embodiment, the present invention may furthermore provide a vaccine containing the active (immunostimulatory) composition according to the invention. The inventive vaccine may additionally contain a pharmaceutically acceptable carrier and/or further auxiliary substances and additives and/or adjuvants. According to a particularly preferred embodiment, the antigens encoded by the at least one RNA of the active (immunostimulatory) composition, contained in the inventive vaccine, are selected from the above mentioned groups or subgroups. According to an even more preferred embodiment, the protein antigens are selected from any of the antigens of the following subgroup comprising NY-ESO1 [accession number NM_001327], hTERT [accession number NM_198253], survivin [accession number AF077350], 5T4 [accession number NM_006670] and WT1 [accession number NM_000378], and/or from any of the antigens of the following subgroup comprising MAGE-C1 and MAGE-C2, as defined herein, and/or from any of the antigens of the following subgroup comprising MAGE-A2 and MAGE-A3, as defined herein.
  • The inventive vaccine typically comprises a safe and effective amount of the at least one RNA of the active (immunostimulatory) composition as defined above encoding at least two antigens as defined above, more preferably encoding at least two antigens selected from any of the above goups or subgroups, most preferably in any of the indicated combinations. As used herein, “safe and effective amount” means an amount of the at least one RNA of the active (immunostimulatory) composition in the vaccine as defined above, that is sufficient to significantly induce a positive modification of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition to be treated, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma. At the same time, however, a “safe and effective amount” is small enough to avoid serious side-effects, that is to say to permit a sensible relationship between advantage and risk. The determination of these limits typically lies within the scope of sensible medical judgment. In relation to the inventive vaccine, the expression “safe and effective amount” preferably means an amount of the RNA (and thus of the encoded at least two antigens) that is suitable for stimulating the adaptive immune system in such a manner that no excessive or damaging immune reactions are achieved but, preferably, also no such immune reactions below a measurable level. Such a “safe and effective amount” of the at least one RNA of the active (immunostimulatory) composition in the vaccine as defined above may furthermore be selected in dependence of the type of RNA, e.g. monocistronic, bi- or even multicistronic RNA, since a bi- or even multicistronic RNA may lead to a significantly higher expression of the encoded antigen(s) than use of an equal amount of a monocistronic RNA. A “safe and effective amount” of the at least one RNA of the active (immunostimulatory) composition as defined above, which is contained in the inventive vaccine, will furthermore vary in connection with the particular condition to be treated and also with the age and physical condition of the patient to be treated, the severity of the condition, the duration of the treatment, the nature of the accompanying therapy, of the particular pharmaceutically acceptable carrier used, and similar factors, within the knowledge and experience of the accompanying doctor. The vaccine according to the invention can be used according to the invention for human and also for veterinary medical purposes, as a pharmaceutical composition or as a vaccine.
  • The vaccine according to the invention typically contains a pharmaceutically acceptable carrier. The expression “pharmaceutically acceptable carrier” as used herein preferably includes the liquid or non-liquid basis of the inventive vaccine. If the inventive vaccine is provided in liquid form, the carrier will typically be pyrogen-free water; isotonic saline or buffered (aqueous) solutions, e.g. phosphate-, citrate-buffered solutions, etc.. Particularly for injection of the inventive vaccine, water or preferably a buffer, more preferably an aqueous buffer, may be used, containing a sodium salt, preferably at least 50 mM of a sodium salt, a calcium salt, preferably at least 0,01 mM of a calcium salt, and optionally a potassium salt, preferably at least 3 mM of a potassium salt. According to a preferred embodiment, the sodium, calcium and, optionally, potassium salts may occur in the form of their halogenides, e.g. chlorides, iodides, or bromides, in the form of their hydroxides, carbonates, hydrogen carbonates, or sulfates, etc. Without being limited thereto, examples of sodium salts include e.g. NaCl, NaI, NaBr, Na2CO3, NaHCO3, Na2SO4, examples of the optional potassium salts include e.g. KCl, KI, KBr, K2CO3, KHCO3, K2SO4, and examples of calcium salts include e.g. CaCl2, CaI2, CaBr2, CaCO3, CaSO4, Ca(OH)2. Furthermore, organic anions of the aforementioned cations may be contained in the buffer.
  • According to a more preferred embodiment, the buffer suitable for injection purposes as defined above, may contain salts selected from sodium chloride (NaCl), calcium chloride (CaCl2) and optionally potassium chloride (KCl), wherein further anions may be present additional to the chlorides. CaCl2 can also be replaced by another salt like KCl. Typically, the salts in the injection buffer are present in a concentration of at least 50 mM sodium chloride (NaCl), at least 3 mM potassium chloride (KCl) and at least 0,01 mM calcium chloride (CaCl2). The injection buffer may be hypertonic, isotonic or hypotonic with reference to the specific reference medium, i.e. the buffer may have a higher, identical or lower salt content with reference to the specific reference medium, wherein preferably such concentrations of the afore mentioned salts may be used, which do not lead to damage of cells due to osmosis or other concentration effects. Reference media are e.g. in “in vivo” methods occurring liquids such as blood, lymph, cytosolic liquids, or other body liquids, or e.g. liquids, which may be used as reference media in “in vitro” methods, such as common buffers or liquids. Such common buffers or liquids are known to a skilled person. Ringer-Lactate solution is particularly preferred as a liquid basis.
  • However, one or more compatible solid or liquid fillers or diluents or encapsulating compounds may be used as well, which are suitable for administration to a person. The term “compatible” as used herein means that the constituents of the inventive vaccine are capable of being mixed with the at least one RNA of the active (immunostimulatory) composition, encoding at least two antigens as defined above, in such a manner that no interaction occurs which would substantially reduce the pharmaceutical effectiveness of the inventive vaccine under typical use conditions. Pharmaceutically acceptable carriers, fillers and diluents must, of course, have sufficiently high purity and sufficiently low toxicity to make them suitable for administration to a person to be treated. Some examples of compounds which can be used as pharmaceutically acceptable carriers, fillers or constituents thereof are sugars, such as, for example, lactose, glucose and sucrose; starches, such as, for example, corn starch or potato starch; cellulose and its derivatives, such as, for example, sodium carboxymethylcellulose, ethylcellulose, cellulose acetate; powdered tragacanth; malt; gelatin; tallow; solid glidants, such as, for example, stearic acid, magnesium stearate; calcium sulfate; vegetable oils, such as, for example, groundnut oil, cottonseed oil, sesame oil, olive oil, corn oil and oil from theobroma; polyols, such as, for example, polypropylene glycol, glycerol, sorbitol, mannitol and polyethylene glycol; alginic acid.
  • The choice of a pharmaceutically acceptable carrier is determined in principle by the manner in which the inventive vaccine is administered. The inventive vaccine can be administered, for example, systemically or locally. Routes for systemic administration in general include, for example, transdermal, oral, parenteral routes, including subcutaneous, intravenous, intramuscular, intraarterial, intradermal and intraperitoneal injections and/or intranasal administration routes. Routes for local administration in general include, for example, topical administration routes but also intradermal, transdermal, subcutaneous, or intramuscular injections or intralesional, intracranial, intrapulmonal, intracardial, and sublingual injections.
  • More preferably, vaccines may be administered by an intradermal, subcutaneous, or intramuscular route. Compositions/vaccines are therefore preferably formulated in liquid or solid form. The suitable amount of the inventive vaccine to be administered can be determined by routine experiments with animal models. Such models include, without implying any limitation, rabbit, sheep, mouse, rat, dog and non-human primate models. Preferred unit dose forms for injection include sterile solutions of water, physiological saline or mixtures thereof. The pH of such solutions should be adjusted to about 7.4. Suitable carriers for injection include hydrogels, devices for controlled or delayed release, polylactic acid and collagen matrices. Suitable pharmaceutically acceptable carriers for topical application include those which are suitable for use in lotions, creams, gels and the like. If the inventive vaccine is to be administered perorally, tablets, capsules and the like are the preferred unit dose form. The pharmaceutically acceptable carriers for the preparation of unit dose forms which can be used for oral administration are well known in the prior art. The choice thereof will depend on secondary considerations such as taste, costs and storability, which are not critical for the purposes of the present invention, and can be made without difficulty by a person skilled in the art.
  • The inventive vaccine can additionally contain one or more auxiliary substances in order to further increase the immunogenicity. A synergistic action of the at least one RNA of the active (immunostimulatory) composition as defined above and of an auxiliary substance, which may be optionally also contained in the inventive vaccine as described above, is preferably achieved thereby. Depending on the various types of auxiliary substances, various mechanisms can come into consideration in this respect. For example, compounds that permit the maturation of dendritic cells (DCs), for example lipopolysaccharides, TNF-alpha or CD40 ligand, form a first class of suitable auxiliary substances. In general, it is possible to use as auxiliary substance any agent that influences the immune system in the manner of a “danger signal” (LPS, GP96, etc.) or cytokines, such as GM-CFS, which allow an immune response produced by the immune-stimulating adjuvant according to the invention to be enhanced and/or influenced in a targeted manner. Particularly preferred auxiliary substances are cytokines, such as monokines, lymphokines, interleukins or chemokines, that—additional to induction of the adaptive immune response by the encoded at least two antigens—promote the innate immune response, such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, IL-21, IL-22, IL-23, IL-24, IL-25, IL-26, IL-27, IL-28, IL-29, IL-30, IL-31, IL-32, IL-33, INF-alpha, IFN-beta, INF-gamma, GM-CSF, G-CSF, M-CSF, LT-beta or TNF-alpha, growth factors, such as hGH.
  • Further additives which may be included in the inventive vaccine are emulsifiers, such as, for example, Tween®; wetting agents, such as, for example, sodium lauryl sulfate; colouring agents; taste-imparting agents, pharmaceutical carriers; tablet-forming agents; stabilizers; antioxidants; preservatives.
  • The inventive vaccine can also additionally contain any further compound, which is known to be immune-stimulating due to its binding affinity (as ligands) to human Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLRS, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLRS, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12 or TLR13.
  • Another class of compounds, which may be added to an inventive vaccine in this context, may be CpG nucleic acids, in particular CpG-RNA or CpG-DNA. A CpG-RNA or CpG-DNA can be a single-stranded CpG-DNA (ss CpG-DNA), a double-stranded CpG-DNA (dsDNA), a single-stranded CpG-RNA (ss CpG-RNA) or a double-stranded CpG-RNA (ds CpG-RNA). The CpG nucleic acid is preferably in the form of CpG-RNA, more preferably in the form of single-stranded CpG-RNA (ss CpG-RNA). The CpG nucleic acid preferably contains at least one or more (mitogenic) cytosine/guanine dinucleotide sequence(s) (CpG motif(s)). According to a first preferred alternative, at least one CpG motif contained in these sequences, that is to say the C (cytosine) and the G (guanine) of the CpG motif, is unmethylated. All further cytosines or guanines optionally contained in these sequences can be either methylated or unmethylated. According to a further preferred alternative, however, the C (cytosine) and the G (guanine) of the CpG motif can also be present in methylated form.
  • According to a further preferred object of the present invention, the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein, may be used (for the preparation of a vaccine according to the present invention) for the treatment of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma.
  • According to a further preferred object of the present invention, the inventive vaccine or the at least one RNA encoding at least two (preferably) different antigens as defined herein may be used for the treatment of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma.
  • In this context also included in the present invention are methods of treating lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma, by administering to a patient in need thereof a pharmaceutically effective amount of an inventive vaccine, or a pharmaceutically effective amount of an inventive active (immunostimulatory) composition. Such a method typically comprises an optional first step of preparing the inventive active (immunostimulatory) composition, or the inventive vaccine, and a second step, comprising administering (a pharmaceutically effective amount of) said inventive active (immunostimulatory) composition or said inventive vaccine to a patient in need thereof. A patient in need thereof will be typically selected from any mammal. In the context of the present invention, a mammal is preferably selected from the group comprising, without being limited thereto, e.g. goat, cattle, swine, dog, cat, donkey, monkey, ape, a rodent such as a mouse, hamster, rabbit and, particularly, human, wherein the mammal typically suffers from lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including, without being restricted thereto, squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma or a condition related thereto.
  • The invention relates also to the use of the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein (for the preparation of an inventive vaccine), preferably for eliciting an immune response in a mammal, preferably for the treatment of lung cancer, more preferably for the treatment of a non-small-cell lung cancer (NSCLC) related condition as defined herein.
  • Similarly, the invention also relates also to the use of the inventive vaccine per se or the at least one RNA encoding at least two (preferably) different antigens as defined herein for eliciting an adaptive immune response in a mammal, preferably for the treatment of lung cancer, more preferably of a non-small-cell lung cancer (NSCLC) related condition as defined herein.
  • Prevention or treatment of lung cancer in a patient in need thereof, preferably of a non-small-cell lung cancer (NSCLC) related condition as defined herein, may be carried out by administering the inventive active (immunostimulatory) composition and/or the inventive vaccine at once or in a time staggered manner, e.g. as a kit of parts, each part containing at least one preferably different antigen. For administration, preferably any of the administration routes may be used as defined above. E.g., one may treat lung cancer, preferably a non-small-cell lung cancer (NSCLC) related condition as defined herein, by inducing or enhancing an adaptive immune response on the basis of at least two (specifically selected) antigens encoded by the at least one RNA of the inventive active (immunostimulatory) composition. Administering of the inventive active (immunostimulatory) composition and/or the inventive vaccine may then occur prior, concurrent and/or subsequent to administering another inventive inventive active (immunostimulatory) composition and/or inventive vaccine as defined herein which may contain another combination of RNAs encoding different antigens, wherein each antigen encoded by the at least one RNA of the inventive active (immunostimulatory) composition may preferably be suitable for the therapy of lung cancer, more preferably for the treatment of a non-small-cell lung cancer (NSCLC) related condition as defined herein. In this context, a therapy as defined herein may also comprise the modulation of a disease associated to lung cancer, preferably a disease associated to non-small-cell lung cancer (NSCLC) as defined herein.
  • According to one further embodiment, the present invention furthermore comprises the use of the active (immunostimulatory) composition (for the preparation of an (inventive) vaccine) for modulating, preferably to induce or enhance, an immune response in a mammal as defined above, more preferably to support the treatment of lung cancer, especially NSCLC as defined herein. In this context, support of the treatment of lung cancer, especially NSCLC as defined herein, may be any combination of a conventional cancer therapy for lung cancer, especially for NSCLC as defined herein, such as radiation therapy, chemotherapy, proton therapy, hormonal therapy, antibody therapy, adjuvant therapies, therapies including other vaccines than an inventive vaccine, therapies including kinase inhibitors or small nucleotides, etc., or some combination of these, and a therapy using the inventive active (immunostimulatory) composition or the inventive vaccine as defined herein. Support of the treatment of lung cancer, especially NSCLC as defined herein, may be also envisaged in any of the other embodiments defined herein.
  • Administration of the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein or the inventive vaccine may be carried out in a time staggered treatment. A time staggered treatment may be e.g. administration of the inventive active (immunostimulatory) composition or the at least one RNA encoding at least two (preferably) different antigens as defined herein or the inventive vaccine prior, concurrent and/or subsequent to a therapy of lung cancer, especially NSCLC, e.g. by administration of the inventive active (immunostimulatory) composition or vaccine prior, concurrent and/or subsequent to a therapy or an administration of a therapeutic suitable for the treatment of lung cancer, especially of NSCLC as defined herein. Such time staggered treatment may be carried out using e.g. a kit, preferably a kit of parts as defined below.
  • Time staggered treatment may additionally or alternatively also comprise an administration of the inventive active (immunostimulatory) composition or vaccine, preferably of the at least one RNA encoding at least two (preferably different) antigens as defined above, in a form, wherein the at least one RNA encoding at least two (preferably different) antigens as defined above, preferably forming part of the inventive active (immunostimulatory) composition or vaccine, is administered parallel, prior or subsequent to another at least one RNA encoding at least two (preferably different) antigens as defined above, preferably forming part of the same inventive active (immunostimulatory) composition or vaccine. Preferably, the administration (of all at least one RNAs) occurs within an hour, more preferably within 30 minutes, even more preferably within 15, 10, 5, 4, 3, or 2 minutes or even within 1 minute. Such time staggered treatment may be carried out using e.g. a kit, preferably a kit of parts as defined below.
  • According to a final embodiment, the present invention also provides kits, particularly kits of parts, comprising the active inventive (immunostimulatory) composition, and/or the inventive vaccine, and optionally technical instructions with information on the administration and dosage of the inventive active (immunostimulatory) composition and/or the inventive vaccine. The technical instructions may contain information about administration and dosage of the inventive active (immunostimulatory) composition, and/or the inventive vaccine. Such kits, preferably kits of parts, may applied e.g. for any of the above mentioned applications or uses, preferably for the use of at least one inventive active (immunostimulatory) composition (for the preparation of an inventive vaccine) for the treatment of lung cancer, especially of NSCLC as defined herein. The kits may also be applied for the use of at least one inventive active (immunostimulatory) composition (for the preparation of an inventive vaccine) for the treatment of lung cancer, preferably NSCLC as defined herein, wherein the inventive active (immunostimulatory) composition) and/or the vaccine due to the encoded at least two antigens may be capable to induce or enhance an immune response in a mammal as defined above. Such kits may further be applied for the use of at least one inventive active (immunostimulatory) composition, (for the preparation of an inventive vaccine) for modulating, preferably for eliciting, e.g. to induce or enhance, an immune response in a mammal as defined above, and preferably to support treatment of lung cancer, especially of NSCLC. Kits of parts, as a special form of kits, may contain one or more identical or different active inventive (immunostimulatory) compositions and/or one or more identical or different inventive vaccines in different parts of the kit. Kits of parts may also contain an (e.g. one) active inventive (immunostimulatory) composition, an (e.g. one) inventive vaccine and/or the at least one RNA encoding at least one antigen as defined above in different parts of the kit, e.g. each part of the kit containing at least one RNA encoding a preferably different antigen. Additionally, a combination of both types of kits of parts is possible. Kits of parts may be used, e.g. when a time staggered treatment is envisaged, e.g. when using different formulations and/or increasing concentrations of the active inventive (immunostimulatory) composition, the inventive vaccine and/or the at least one RNA encoding at least one antigens as defined above during the same treatment in vivo. Kits of parts may also be used when a separated formulation or administration of the different antigens of the inventive active (immunostimulatory) composition (i.e. in parts) is envisaged or necessary (e.g. for technical reasons), but e.g. a combined presence of the different antigens in vivo is still to be achieved. Particularly kits of parts as a special form of kits are envisaged, wherein each part of the kit contains at least one preferably different antigen as defined above, all parts of the kit of parts preferably forming the active inventive (immunostimulatory) composition or the inventive vaccine as defined herein. Such specific kits of parts may particularly be suitable, e.g. if different antigens are formulated separately as different parts of the kits, but are then administered at once together or in a time staggered manner to the mammal in need thereof. In the latter case administration of all of the different parts of such a kit typically occurs within a short time limit, such that all antigens are present in the mammal at about the same time subsequent to administration of the last part of the kit. Any of the above kits may be used in a treatment as defined above.
  • Advantages of the Present Invention
  • The present invention provides an active (immunostimulatory) composition for the treatment of lung cancer, particularly of non-small lung cancer (NSCLC), wherein the composition comprises at least one RNA, preferably a mRNA, encoding at least two (preferably different) antigens capable of eliciting an (adaptive) immune response in a mammal wherein the antigens are selected from the group consisting of hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1, or MAGE-C2. Such an active (immunostimulatory) composition allows efficient treatment of lung cancer, particularly of non-small lung cancer (NSCLC), or supplementary treatment when using conventional therapies. It furthermore avoids the problem of uncontrolled propagation of the introduced DNA sequences by the use of RNA as an approach for curative methods. RNA as used in the inventive active (immunostimulatory) composition has additional considerable advantages over DNA expression systems e.g. in immune response, immunization or vaccination. These advantages include, inter alfa, that RNA introduced into a cell is not integrated into the genome. This avoids the risk of mutation of this gene, which otherwise may be completely or partially inactivated or give rise to misinformation. It further avoids other risks of using DNA as an agent to induce an immune response (e.g. as a vaccine) such as the induction of pathogenic anti-DNA antibodies in the patient into whom the foreign DNA has been introduced, so bringing about a (possibly fatal) immune response. In contrast, no anti-RNA antibodies have yet been detected.
  • BRIEF DESCRIPTION OF DRAWINGS
  • The following Figures are intended to illustrate the invention further. They are not intended to limit the subject matter of the invention thereto.
  • FIG. 1: depicts a RNA sequence (SEQ ID NO: 1) (starting sequence based on the wildtype) encoding MUC1 (HsMUC1-5xVNTR (The wildtype sequence normally shows 40 tandem repeats. These were—for cloning reasons—reduced to 5 tandem repeats). GC content: 61.27%; length: 1668 bp).
  • FIG. 2: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 2) encoding MUC1 (HsMUC1 GC-5xVNTR, 1. GC maximized, 2. Codon usage) GC content: 73.56%; length 1668 bp. Difference to basic sequence (FIG. 1 (SEQ ID NO: 1)): 398/1668 bases=23.86%.
  • FIG. 3: depicts a RNA sequence (SEQ ID NO: 3) (starting sequence based on the wildtype) encoding 5T4 (Hs5T4 (trophoblast glycoprotein TPBG); GC content: 61.60%; length: 1263 bp.
  • FIG. 4: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 4) encoding 5T4 (Hs5T4 GC, 1. GC-maximized, 2. Codon usage); GC content: 70.47%; length 1263 bp. Difference to basic sequence (FIG. 3 (SEQ ID NO: 3)): 247/1263 Bases=19.56%.
  • FIG. 5: depicts a RNA sequence (SEQ ID NO: 5) (starting sequence based on the wildtype) encoding Her-2/neu (HsHer2/neu (v-erb-b2 erythroblastic leukemia viral oncogene homolog 2)); GC content: 60.78% ; length: 3768 bp.
  • FIG. 6: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 6) encoding Her-2/neu (HsHer2/neu GC, 1. GC-maximized, 2. Codon usage); GC content: 70.54%; length 3768 bp. Difference to basic sequence (FIG. 5 (SEQ ID NO: 5)): 772/3768 Bases=20.49%.
  • FIG. 7: depicts a RNA sequence (SEQ ID NO: 7) (starting sequence based on the wildtype) encoding hTERT (HsTERT (telomerase reverse transcriptase); GC Content: 66.08% ; Length: 3399 bp.
  • FIG. 8: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 8) encoding hTERT (HsTERT GC, 1. GC-maximized, 2. Codon usage); GC Content: 72.96%; Length 3399 bp, Difference to basic sequence (FIG. 7 (SEQ ID NO: 7)): 566/3399 Bases=16.65%.
  • FIG. 9: depicts a RNA sequence (SEQ ID NO: 9) (starting sequence based on the wildtype) encoding WT1 (HsWT1 (Wilms tumor 1)); GC Content: 61.78% ; Length: 1554 bp.
  • FIGS. 10A-D: FIG. 10 A) depicts a RNA sequence (SEQ ID NO: 10) encoding WT1 (HsWT1 (Wilms tumor 1)) showing a sequence with a reduced GC content in region 325-408 of said sequence compared to the corresponding region of the wildtype sequence.
      • FIGS. 10 B), C) and D) show a comparison of the corresponding regions 325-408:
      • in B) the wildtype sequence according to FIG. 9 (SEQ ID NO: 53),
      • in C) the GC-maximized sequence according to FIG. 11 (SEQ ID NOS: 54 and 55, respectively, in order of appearance),
      • and
      • in D) the GC-reduced sequence according to FIG. 10 (SEQ ID NOS: 56 and 57, respectively, in order of appearance),
      • which all show a different GC-pattern.
  • FIG. 11: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 11) encoding WT1 (HsWT1 GC, 1. GC-maximized, 2. Codon usage); GC Content: 72.59%; Length 1554 bp. Difference to basic sequence (FIG. 9 (SEQ ID NO: 9)): 322/1554 Bases=20.72%.
  • FIG. 12: depicts a RNA sequence (SEQ ID NO: 12) (starting sequence based on the wildtype) encoding CEA (CEA (carcinoembryonic antigen) HsCEACAMS); GC Content: 52.20% ; Length: 2109 bp.
  • FIG. 13: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 13) encoding CEA (CEACAMS GC, 1. GC-maximized, 2. Codon usage, already in place); GC Content: 66.24%; Length 2109 bp. Difference to basic sequence (FIG. 12 (SEQ ID NO: 12)): 495/2109 Bases=23.47%.
  • FIG. 14: depicts a RNA sequence (SEQ ID NO: 14) (starting sequence based on the wildtype) encoding MAGE-A2 (HsMAGE-A2 (melanoma antigen family A, 2) HsMAGE-A2B). GC Content: 55.87% ; Length: 945 bp.
  • FIG. 15: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 15) encoding MAGE-A2 (HsMAGE-A2B GC, 1. GC-maximized, 2. Codon usage); GC Content: 68.57%; Length 945 bp. Difference to basic sequence (FIG. 14 (SEQ ID NO: 14)): 187/945 Bases=19.79%.
  • FIG. 16: depicts a RNA sequence (SEQ ID NO: 16) (starting sequence based on the wildtype) encoding MAGE-A3 (MAGE-A3 (melanoma antigen family A, 3) MAGE-A3) GC Content: 56.30% ; Length: 945 bp.
  • FIG. 17: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 17) encoding MAGE-A3 (MAGE-A3 GC, 1.GC-maximized, 2. Codon usage, already known GC-Enrichment); GC Content: 69.00%; Length 945 bp. Difference to basic sequence (FIG. 16 (SEQ ID NO: 16)): 190/945 Bases=20.11%.
  • FIG. 18: depicts a RNA sequence (SEQ ID NO: 18) (starting sequence based on the wildtype) encoding Survivin (Survivin (baculoviral IAP repeat-containing 5, BIRC5) HsSurvivin(wt)); GC Content: 52.68% ; Length: 429 bp.
  • FIG. 19: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 19) encoding Survivin (HsSurvivin(GC), 1. GC-maximized, 2. Codon Usage, already known GC-Enrichment); GC Content: 65.27%; Length: 429 bp. Difference to basic sequence (FIG. 18 (SEQ ID NO: 18)): 72/429 Bases=16.78%.
  • FIG. 20: depicts a RNA sequence (SEQ ID NO: 20) (starting sequence based on the wildtype) encoding NY-ESO-1 (Homo sapiens NY-ESO-1 (NY-ESO-1(wt)); GC-Content 67.4%.
  • FIG. 21: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 21) encoding NY-ESO-1 (NY-E SO-1(GC), GC-Content 79.56%, (already known GC-Enrichment); Difference to wt (FIG. 20 (SEQ ID NO: 20)): 112/543 Bases, 20.63%.
  • FIG. 22: depicts a RNA sequence (SEQ ID NO: 22) (starting sequence based on the wildtype) encoding MAGE-C1 (HsMAGEC1 (melanoma antigen family C, 1) HsMAGEC1(wt)) GC Content: 51.86%; Length: 3429 bp.
  • FIG. 23: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 23) encoding MAGE-C1 (HsMAGEC1(GC), 1. GC-maximized, 2. Codon usage). GC Content: 68.73%; Length 3429 bp. Difference to basic sequence (FIG. 22 (SEQ ID NO: 22)): 964/3429 Bases =28.11%
  • FIG. 24: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 24) encoding a truncated MAGE-C1 (HsMAGEC1(GC), 1. GC-maximized, 2. Codon usage). In comparison to the basic sequence (FIG. 22 (SEQ ID NO: 22)) the repeat regions were deleted and the sequence according to FIG. 24, following an initial start codon (ATG), starts at aa 613 of the GC-maximized wildtype sequence (FIG. 23 (SEQ ID NO: 23)).
  • FIG. 25: depicts a RNA sequence (SEQ ID NO: 25) (starting sequence based on the wildtype) encoding MAGE-C2 (HsMAGE-C2 (melanoma antigen family C, 2)HsMAGE-C2); GC Content: 50.81% ; Length: 1122 bp.
  • FIG. 26: depicts a (GC) stabilized RNA sequence (SEQ ID NO: 26) encoding MAGE-C2 (HsMAGE-C2 GC, 1. GC-maximized, 2. Codon usage); GC Content: 66.58%; Length 1122 bp, Difference to basic sequence (FIG. 25 (SEQ ID NO: 25)): 264/1122 Bases =23.53%.
  • FIG. 27 shows the presence of IgG1 antibodies specific for the tumour antigen NY-ESO-1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 28: shows the presence of IgG2a antibodies specific for the tumour antigen NY-ESO-1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 29: shows the presence of IgG1 antibodies specific for the tumour antigen MAGE-C1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 30: shows the presence of IgG2a antibodies specific for the tumour antigen MAGE-C1 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 31 shows the presence of IgG1 antibodies specific for the tumour antigen MAGE-C2 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 32: shows the presence of IgG2a antibodies specific for the tumour antigen MAGE-C2 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 33: shows the induction of antigen-specific T-lymphocytes directed against the tumour antigen 5T4 in mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1,MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • FIG. 34: shows the induction of antigen-specific T-lymphocytes directed against the tumour antigen NY-ESO-1 iin mice which were vaccinated with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4) formulated with protamine at a mass ratio of 4:1.
  • EXAMPLES
  • The following examples are intended to illustrate the invention further. They are not intended to limit the subject matter of the invention thereto.
  • 1. Preparation of Encoding Plasmids:
  • In the following experiment DNA sequences, corresponding to the respective mRNA sequences end encoding the antigens
      • hTERT,
      • WT1,
      • MAGE-A2,
      • 5T4,
      • MAGE-A3,
      • MUC1,
      • Her-2/neu,
      • NY-ESO-1,
      • CEA,
      • Survivin,
      • MAGE-C1, or
      • MAGE-C2.
  • respectively, were prepared and used for in vitro transcription and transfection experiments. Thereby, the DNA sequence corresponding to the native antigen encoding mRNA was increased in GC-content and codon-optimized. Then, the coding sequence was transferred into an RNActive construct (CureVac GmbH, Tubingen, Germany), which has been modified with a poly-A-tag and a poly-C-tag (A70-C30).
  • 2. In Vitro Transcription:
  • Based on the recombinant plasmid DNA obtained in Example 1 the RNA sequences were prepared by in vitro transcription. Therefore, the recombinant plasmid DNA was linearized and subsequently in vitro transcribed using the T7 RNA polymerase. The DNA template was then degraded by DNase I digestion, and the RNA was recovered by LiCl precipitation and further cleaned by HPLC extraction (PUREMessenger®, CureVac GmbH, Tubingen, Germany).
  • 3. Complexation with Protamine
  • For transfection of the RNA into cells and organisms the RNA obtained by in vitro transcription was preferably complexed, more preferably with protamine upon mixing the RNA with protamine.
  • 4. Vaccination Experiments
  • For vaccination the RNA obtained by the in vitro transcription experiment as shown above (see Experiment 2) was transfected into mice (Mice: C57 BL/6), preferably when complexed with protamine (see Experiment 3). Transfection occurred in different groups, wherein 5 mice (C57 BL/6) per group were immunized intradermally 8 times within 3 weeks with the inventive mRNA cocktail, i.e. a mixture of mRNA complexed with protamine, wherein the RNA codes for at least two of the antigens hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1, or MAGE-C2.
  • 5. Detection of an Antigen-Specific Immune Response (B-Cell Immune Response):
  • Detection of an antigen-specific immune response (B-cell immune response) was carried out by detecting antigen-specific antibodies. Therefore, blood samples were taken from the vaccinated mice one week after the last vaccination and sera were prepared. MaxiSorb plates (Nalgene Nunc International) were coated with the antigenic protein as encoded by the mRNA-Cocktail (0.5 μg/well). After blocking with 1× PBS containing 0.05% Tween-20 and 1% BSA the plates were incubated with diluted mouse serum (1:30, 1:90, 1:270, 1:810). Subsequently a biotin-coupled secondary antibody (Anti-mouse-IgG2a Pharmingen) was added. After washing, the plate was incubated with Horseradish peroxidase-streptavidin and subsequently the conversion of the ABTS substrate (2,2′-azino-bis(3-ethyl-benzthiazoline-6-sulfonic acid) was measured.
  • 6. Detection of an Antigen-Specific Cellular Immune Response (T Cell Immune Response) by ELISPOT:
  • 2 weeks after the last vaccination mice were sacrificed, the spleens were removed and the splenocytes were isolated. The splenocytes were restimulated for 7 days in the presence of peptides from the above antigens (peptide library) or coincubated with dendritic cells generated from bone marrow cells of native syngeneic mice, which are electroporated with RNA coding for the antigen. To determine an antigen-specific cellular immune response INFgamma secretion was measured after re-stimulation. For detection of INFgamma a coat multiscreen plate (Millipore) was incubated overnight with coating buffer 0.1 M carbonate-bicarbonate buffer pH 9.6, 10.59 g/l Na2CO3, 8.4 g/l NaHCO3) comprising antibody against INFγ (BD Pharmingen, Heidelberg, Germany). Stimulators and effector cells were incubated together in the plate in the ratio of 1:20 for 24 h. The plate was washed with 1× PBS and incubated with a biotin-coupled secondary antibody. After washing with 1× PBS/0.05% Tween-20 the substrate (5-Bromo-4-Cloro-3-Indolyl Phosphate/Nitro Blue Tetrazolium Liquid Substrate System from Sigma Aldrich, Taufkirchen, Germany) was added to the plate and the conversion of the substrate could be detected visually.
  • 7. Tumor Challenge:
  • Immunization:
  • One week after the last immunization 1 Mio B16 melanoma cells or TRAMP-C1 cells were injected subcutaneously in the mice. Within 2 weeks (B16) or 7 weeks (TRAMP-C1), respectively, tumour volume was determined
  • 8. Preparation of a mRNA Vaccine
  • A particular example of the inventive active (immunostimulatory) composition, comprising a combination of several antigens for the use as a vaccine for the treatment of non-small cell lung cancer (NSCLC) was prepared in the following according to the above disclosure. The exemplary inventive active (immunostimulatory) composition consisted of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4, according to SEQ ID NOs: 4, 19, 21, 24 and 26 (GC-enriched sequences)) formulated with protamine at a mass ratio of 4:1.
  • Vaccination C57BL/6 mice were vaccinated intradermally with the mRNA vaccine consisting of 5 components, each containing mRNA coding for one NSCLC related antigen (NY-ESO-1, MAGE-C1, MAGE-C2, Survivin and 5T4, according to SEQ ID NOs: 4, 19, 21, 24 and 26 (GC-enriched sequences)) formulated with protamine (64 μg per antigen per cycle, divided into 4 injections/cycle). Control vaccination was performed using the corresponding total doses of RNA coding for LacZ (control mRNA lacZ). The vaccination comprised three immunization cycles ( week 1, 3, and 5). The groups, number of mice and mouse strains are indicated in the following table:
  • Groups Mouse strain Number of mice
    mRNA vaccine C57BL/6 10
    5 for Elispot and 5 for
    antibody detection in serum
    by ELISA
    Control mRNA lacZ C57BL/6 5
    3 for Elispot and all 5 for
    antibody detection in serum
    by ELISA
  • Detection of antigen-specific antibodies
  • 6 days after last vaccination blood samples (200 μl) were taken retro-orbitally and serum was analyzed for the presence of antigen specific antibody subtypes IgG1 and IgG2a using ELISA. 96-well ELISA plates were coated with recombinant protein (10 pg/m1 in coating buffer, incubation at 37° C. for 4 h) and blocked with 200 μl blocking buffer per well over night at 4° C. Subsequently, the samples were incubated with serum pooled from each group of mice and titrated in dilutions ranging from 1:3 to 1:48 for 4 hours at room temperature. After incubation with a specific antibody (1:300 in blocking buffer) against mouse IgG1 or IgG2a and incubation with a HRP-coupled secondary antibody (1:500 in blocking buffer), TMB-substrate was added. The colorimetric reaction was measured at 450 nm using an ELISA reader (Tecan Deutschland GmbH, Crailsheim, Germany).
  • ELISPOT
  • For the detection of cytotoxic T-lymphocyte (CTL) responses the analysis of the secretion of the effector cytokine IFN-γ in response to a specific stimulus can be visualized at a single cell level using the ELISPOT technique.
  • Splenocytes from antigen-vaccinated and control mice were isolated 6 days after last vaccination and then transferred into 96-well ELISPOT plates coated with an anti-IFN-γ capture antibody (10 μg/ml). The cells were then stimulated for 24 hours at 37° C. either with relevant antigen-derived peptide library or with the HIV-derived library or the solvent of the peptides, DMSO, or incubated in pure medium as a control. All libraries were used at a concentration of 1 μg/peptide/ml. After the incubation period the cells were washed out of the plate and the IFN-γ secreted by the cells was detected using a biotinylated secondary antibody against murine IFN-γ (1 μg/ml), followed by streptavidin-AKP. Spots were visualized using BCIP/NBT substrate and counted using an automated ELISPOT reader (Immunospot Analyzer, CTL Analyzers LLC).
  • Statistical Analysis
  • Statistical analysis was performed using Graph Pad Prism 5.01 (GraphPad Software, Inc.). All results were expressed as the mean (or median)±standard error of means. For Elispot assays, due to the fact that the basal activation is strongly individual dependent, a background correction was performed individually per mouse by subtraction of the number of spots in medium wells from all other values. Two-tailed Mann-Whitney tests were used to analyze difference between the test groups with a significance level of 5%.
  • Results and Discussion
  • Mice were vaccinated with the mRNA vaccine containing five components as defined above, particularly GC-enriched mRNAs coding for the NSCLC-associated antigens NY-ESO-1, MAGE-C2, MAGE-C1, Survivin and 5T4, (according to SEQ ID NOs: 4, 19, 21, 24 and 26 (GC-enriched sequences)) each formulated separately with the cationic peptide protamine at a mass ratio of 4:1. Control mice were treated with irrelevant RNA coding for LacZ formulated with protamine at the same ratio as the mRNA vaccine.
  • Using serum isolated from blood drawn from the antigen-vaccinated and control mice, we tested the induction of specific antibodies against the antigens. For three of the five analyzed proteins, MAGE-C1, MAGE-C2 and NY-ESO-1, we detected antigen specific antibodies in serum of mice vaccinated with the mRNA vaccine demonstrating that the mRNAs are functional and immunogenic in vivo. Proteins required for detection of antibodies were produced in E. coli. As production of proteins in E.coli can influence post-translational modifications and these are not well described for the used antigens, this could account for the lack of response seen for the remaining proteins.
  • Next the activation of cytotoxic T-cells in response to the administration of the mRNA vaccine was analyzed. IFN-γ is the main mediator of Th1 responses and secreted by activated CTLs. Therefore the presence of antigen-specific cytotoxic T-cells in splenocytes from vaccinated mice was investigated using the ELISPOT technique. As an antigenic stimulus for splenocytes restricted peptide libraries were used. Because distinct epitopes of the used human antigens for mouse MHC (H-2Kb and H-2Db in C57BL/6 mice) are not known, we had to use a hypothetical selection of peptides selected due to potential binding affinity by search of the SYFPEITHI database. Out of peptide libraries (15mers with 11 amino acids overlap) spanning the whole sequences of the proteins, those 15mers containing the hypothetically best epitopes were selected and pooled up to a maximum of 18 peptides. However, these selections might not necessarily contain the correct epitopes so that the detection of immune responses with the help of these tools can easily yield false negative results. Nevertheless, the stimulation with two of these libraries, originating from NY-ESO-1 and 5T4, led to high IFN-γ secretion in splenocytes from mice vaccinated with the mRNA vaccine and not in splenocytes from control mice, vaccinated with mRNA coding for irrelevant protein β-galctosidase. None of the splenocytes reacted to the HIV-derived control peptide library. The number of IFN-γ spots by splenocytes incubated in medium alone represents the basal activation of the freshly isolated cells. Due to the fact that the basal activation is strongly individual dependent, the background correction was performed individually by subtraction of the number of spots in medium wells from all other values.
  • The results of these experiments are shown in FIGS. 27 to 34.

Claims (24)

1. Active (immunostimulatory) composition comprising at least one RNA encoding at least two different antigens,
a) wherein at least one of these at least two antigens is (are) selected from:
NY-ESO-1,
MAGE-C1, and/or
MAGE-C2; and
b) wherein the further antigen(s) is (are) selected from at least one antigen selected from the following group:
hTERT,
WT1,
MAGE-A2,
5T4,
MAGE-A3,
MUC1,
Her-2/neu,
NY-ESO-1,
CEA,
Survivin,
MAGE-C1, and/or
MAGE-C2.
2. Active (immunostimulatory) composition according to claim 1, wherein at least one of these at least two different antigens according to a) is selected from
MAGE-C1, and/or
MAGE-C2.
3. Active (immunostimulatory) composition according to claim 1 or 2, wherein the further antigen(s) according to b) is (are) exclusively selected from the following combinations of antigens:
hTERT and WT1; or
hTERT and MAGE-A2; or
hTERT and 5T4; or
hTERT and MAGE-A3; or
hTERT and MUC1; or
hTERT and Her-2/neu; or
hTERT and NY-ESO-1; or
hTERT and CEA; or
hTERT and Survivin; or
hTERT and MAGE-C1; or
hTERT and MAGE-C2; or
WT1 and MAGE-A2; or
WT1 and 5T4; or
WT1 and MAGE-A3; or
WT1 and MUC1; or
WT1 and Her-2/neu; or
WT1 and NY-ESO-1; or
WT1 and CEA; or
WT1 and Survivin; or
WT1 and MAGE-C1; or
WT1 and MAGE-C2; or
MAGE-A2 and 5T4; or
MAGE-A2 and MAGE-A3; or
MAGE-A2 and MUC1; or
MAGE-A2 and Her-2/neu; or
MAGE-A2 and NY-ESO-1; or
MAGE-A2 and CEA; or
MAGE-A2 and Survivin; or
MAGE-A2 and MAGE-C1; or
MAGE-A2 and MAGE-C2; or
5T4 and MAGE-A3; or
5T4 and MUC1; or
5T4 and Her-2/neu; or
5T4 and NY-ESO-1; or
5T4 and CEA; or
5T4 and Survivin; or
5T4 and MAGE-C1; or
5T4 and MAGE-C2; or
MAGE-A3 and MUC1; or
MAGE-A3 and Her-2/neu; or
MAGE-A3 and NY-ESO-1; or
MAGE-A3 and CEA; or
MAGE-A3 and Survivin; or
MAGE-A3 and MAGE-C1
MAGE-A3 and MAGE-C2
MUC1 and Her-2/neu; or
MUC1 and NY-ESO-1; or
MUC1 and CEA; or
MUC1 and Survivin; or
MUC1 and MAGE-C1; or
MUC1 and MAGE-C2; or
HER-2/NEU and NY-ESO-1; or
HER-2/NEU and CEA; or
HER-2/NEU and Survivin; or
HER-2/NEU and MAGE-C 1; or
HER-2/NEU and MAGE-C2; or
NY-ESO-1 and CEA; or
NY-ESO-1 and Survivin; or
NY-ESO-1 and MAGE-C1; or
NY-ESO-1 and MAGE-C2; or
CEA and Survivin; or
CEA and MAGE-C1; or
CEA and MAGE-C2; or
Survivin and MAGE-C 1; or
Survivin and MAGE-C2; or
MAGE-C1 and MAGE-C2;
or
hTERT, WT1 and MAGE-A2; or
hTERT, WT1 and 5T4; or
hTERT, WT1 and MAGE-A3; or
hTERT, WT1 and MUC1; or
hTERT, WT1 and Her-2/neu; or
hTERT, WT1 and NY-ESO-1; or
hTERT, WT1 and CEA; or
hTERT, WT1 and Survivin; or
hTERT, WT1 and MAGE-C1; or
hTERT, WT1 and MAGE-C2; or
WT1, MAGE-A2 and 5T4; or
WT1, MAGE-A2 and MAGE-A3; or
WT1, MAGE-A2 and MUC1; or
WT1, MAGE-A2 and Her-2/neu; or
WT1, MAGE-A2 and NY-ESO-1; or
WT1, MAGE-A2 and CEA; or
WT1, MAGE-A2 and Survivin; or
WT1, MAGE-A2 and MAGE-C1; or
WT1, MAGE-A2 and MAGE-C2; or
MAGE-A2, 5T4 and MAGE-A3; or
MAGE-A2, 5T4 and MUC1; or
MAGE-A2, 5T4 and Her-2/neu; or
MAGE-A2, 5T4 and NY-ESO-1; or
MAGE-A2, 5T4 and CEA; or
MAGE-A2, 5T4 and Survivin; or
MAGE-A2, 5T4 and MAGE-C1; or
MAGE-A2, 5T4 and MAGE-C2; or
5T4, MAGE-A3 and MUC1; or
5T4, MAGE-A3 and Her-2/neu; or
5T4, MAGE-A3 and NY-ESO-1; or
5T4, MAGE-A3 and CEA; or
5T4, MAGE-A3 and Survivin; or
5T4, MAGE-A3 and MAGE-C1; or
5T4, MAGE-A3 and MAGE-C2; or
MAGE-A3, MUC1 and Her-2/neu; or
MAGE-A3, MUC1 and NY-ESO-1; or
MAGE-A3, MUC1 and CEA; or
MAGE-A3, MUC1 and Survivin; or
MAGE-A3, MUC1 and MAGE-C1; or
MAGE-A3, MUC1 and MAGE-C2; or
MUC1, Her-2/neu and NY-ESO-1; or
MUC1, Her-2/neu and CEA; or
MUC1, Her-2/neu and Survivin; or
MUC1, Her-2/neu and MAGE-C1; or
MUC1, Her-2/neu and MAGE-C2; or
HER-2/NEU, NY-ESO-1 and CEA; or
HER-2/NEU, NY-ESO-1 and Survivin; or
HER-2/NEU, NY-ESO-1 and MAGE-C1; or
HER-2/NEU, NY-ESO-1 and MAGE-C2; or
NY-ESO-1, CEA and Survivin; or
NY-ESO-1, CEA and MAGE-C1; or
NY-ESO-1, CEA and MAGE-C2; or
CEA, Survivin and MAGE-C 1; or
CEA, Survivin and MAGE-C2; or
Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2 and 5T4; or
hTERT, WT1, MAGE-A2 and MAGE-A3; or
hTERT, WT1, MAGE-A2 and MUC1; or
hTERT, WT1, MAGE-A2 and Her-2/neu; or
hTERT, WT1, MAGE-A2 and NY-ESO-1; or
hTERT, WT1, MAGE-A2 and CEA; or
hTERT, WT1, MAGE-A2 and Survivin; or
hTERT, WT1, MAGE-A2 and MAGE-C1; or
hTERT, WT1, MAGE-A2 and MAGE-C2; or
WT1, MAGE-A2, 5T4 and MAGE-A3; or
WT1, MAGE-A2, 5T4 and MUC1; or
WT1, MAGE-A2, 5T4 and Her-2/neu; or
WT1, MAGE-A2, 5T4 and NY-ESO-1; or
WT1, MAGE-A2, 5T4 and CEA; or
WT1, MAGE-A2, 5T4 and Survivin; or
WT1, MAGE-A2, 5T4 and MAGE-C1; or
WT1, MAGE-A2, 5T4 and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3 and MUC1; or
MAGE-A2, 5T4, MAGE-A3 and Her-2/neu; or
MAGE-A2, 5T4, MAGE-A3 and NY-ESO-1; or
MAGE-A2, 5T4, MAGE-A3 and CEA; or
MAGE-A2, 5T4, MAGE-A3 and Survivin; or
MAGE-A2, 5T4, MAGE-A3 and MAGE-C1; or
MAGE-A2, 5T4, MAGE-A3 and MAGE-C2; or
5T4, MAGE-A3, MUC1, and Her-2/neu; or
5T4, MAGE-A3, MUC1 and NY-ESO-1; or
5T4, MAGE-A3, MUC1 and CEA; or
5T4, MAGE-A3, MUC1 and Survivin; or
5T4, MAGE-A3, MUC1 and MAGE-C1; or
5T4, MAGE-A3, MUC1 and MAGE-C2; or
MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
MAGE-A3, MUC1, Her-2/neu and CEA; or
MAGE-A3, MUC1, Her-2/neu and Survivin; or
MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
MUC1, Her-2/neu, NY-ESO-1 and CEA; or
MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
HER-2/NEU, NY-ESO-1, CEA and Survivin; or
HER-2/NEU, NY-ESO-1, CEA and MAGE-C1; or
HER-2/NEU, NY-ESO-1, CEA and MAGE-C2; or
NY-ESO-1, CEA, Survivin and MAGE-C1; or
NY-ESO-1, CEA, Survivin and MAGE-C2; or
CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4 and MAGE-A3; or
hTERT, WT1, MAGE-A2, 5T4 and MUC1; or
hTERT, WT1, MAGE-A2, 5T4 and Her-2/neu; or
hTERT, WT1, MAGE-A2, 5T4 and NY-ESO-1; or
hTERT, WT1, MAGE-A2, 5T4 and CEA; or
hTERT, WT1, MAGE-A2, 5T4 and Survivin; or
hTERT, WT1, MAGE-A2, 5T4 and MAGE-C1; or
hTERT, WT1, MAGE-A2, 5T4 and MAGE-C2; or
WT1, MAGE-A2, 5T4, MAGE-A3 and MUC1; or
WT1, MAGE-A2, 5T4, MAGE-A3 and Her-2/neu; or
WT1, MAGE-A2, 5T4, MAGE-A3 and NY-ESO-1; or
WT1, MAGE-A2, 5T4, MAGE-A3 and CEA; or
WT1, MAGE-A2, 5T4, MAGE-A3 and Survivin; or
WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C1; or
WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3, MUC1 and Her-2/neu; or
MAGE-A2, 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
MAGE-A2, 5T4, MAGE-A3, MUC1 and CEA; or
MAGE-A2, 5T4, MAGE-A3, MUC1 and Survivin; or
MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C1; or
MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C2; or
5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
HER-2/NEU, NY-ESO-1, CEA, Survivin and MAGE-C1; or
HER-2/NEU, NY-ESO-1, CEA, Survivin and MAGE-C2; or
NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and MUC1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and Her-2/neu; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and NY-ESO-1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and CEA; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and Survivin; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3 and MAGE-C2; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Her-2/neu; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and CEA; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Survivin; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
HER-2/NEU, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Her-2/neu; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and NY-ESO-1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and CEA; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and Survivin; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1 and MAGE-C2; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and NY-ESO-1;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and CEA; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and Survivin; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C1;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu and MAGE-C2;
or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin;
or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin;
or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1;
or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2;
or
MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and CEA; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and Survivin; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1 and MAGE-C2; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C1; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and Survivin; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA and MAGE-C2; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C 1; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C 1; or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin and MAGE-C2; or
WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2;
or
hTERT, WT1, MAGE-A2, 5T4, MAGE-A3, MUC1, Her-2/neu, NY-ESO-1, CEA, Survivin, MAGE-C1 and MAGE-C2.
4. Active (immunostimulatory) composition according to any of claims 1 to 3, wherein the at least one RNA comprises a length of 250 to 20000 nucleotides.
5. Active (immunostimulatory) composition according to any of claims 1 to 4, wherein the at least one RNA is a mRNA.
6. Active (immunostimulatory) composition according to any of claims 1 to 5, wherein the at least one RNA is a monocistronic, bicistronic or even multicistronic RNA.
7. Active (immunostimulatory) composition according to claim 5, wherein the at least two antigens are each encoded by a monocistronic RNA.
8. Active (immunostimulatory) composition according to claim 6, wherein the at least two antigens are encoded by a mixture of monocistronic, bicistronic and/or even multicistronic RNAs.
9. Active (immunostimulatory) composition according to any of claims 1 to 8, wherein the at least one RNA comprises a RNA selected from RNAs encoding a fragment, a variant or an epitope of an antigen as defined in any of claims 1 to 3.
10. Active (immunostimulatory) composition according to any of claims 1 to 9, wherein the at least one RNA comprises a RNA selected from RNAs being identical or at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, or 80% identical to the RNA sequences according to SEQ ID NOs: 1, 3, 5, 7, 9, 12, 14, 16, 18, 20, 22 or 25.
11. Active (immunostimulatory) composition according to any of claims 1 to 10, wherein the at least one RNA is a modified RNA, in particular a stabilized mRNA.
12. Active (immunostimulatory) composition according to claim 11, wherein the G/C content of the coding region of the at least one RNA is increased compared to the G/C content of the coding region of the wild-type RNA, the coded amino acid sequence of the at least one RNA preferably not being modified compared to the encoded amino acid sequence of the wild-type RNA.
13. Active (immunostimulatory) composition according to any of claims 11 to 12, wherein the A/U content in the environment of the ribosome binding site of the at least one RNA is increased compared with the A/U content in the environment of the ribosome binding site of the wild-type RNA.
14. Active (immunostimulatory) composition according to any of claims 11 to 13, wherein the coding region and/or the 5′ and/or 3′ untranslated region of the modified mRNA is modified compared to the wild-type RNA such that it contains no destabilizing sequence elements, the coded amino acid sequence of the modified mRNA preferably not being modified compared to the wild-type RNA.
15. Active (immunostimulatory) composition according to any of claims 11 to 14, wherein the modified mRNA has a 5′ cap structure and/or a poly(A) tail, preferably of 10 to 200 adenosine nucleotides, and/or a poly(C) tail, preferably of 10 to 200 cytosine nucleotides, and/or at least one IRES and/or at least one 5′ and/or 3′ stabilizing sequence.
16. Active (immunostimulatory) composition according to any of claims 1 to 15, wherein the at least one RNA, preferably all mRNAs, comprises a RNA selected from RNAs being identical or at least 80% identical to the RNA sequences according to SEQ ID NOs: 2, 4, 6, 8, 10, 11, 13, 15, 17, 19, 21, 23, 24 or 26.
17. Active (immunostimulatory) composition according to any of claims 1 to 16, wherein the at least one RNA is complexed with one or more polycations, preferably with protamine or oligofectamine, most preferably with protamine.
18. Active (immunostimulatory) composition according to any of claims 1 to 17, wherein the active composition additionally comprises at least one adjuvant.
19. Active (immunostimulatory) composition according to claim 18, wherein the at least one adjuvant is selected from the group consisting of:
cationic or polycationic compounds, comprising cationic or polycationic peptides or proteins, including protamine, nucleoline, spermin or spermidine, poly-L-lysine (PLL), poly-arginine, basic polypeptides, cell penetrating peptides (CPPs), including HIV-binding peptides, Tat, HIV-1 Tat (HIV), Tat-derived peptides, Penetratin, VP22 derived or analog peptides, HSV VP22 (Herpes simplex), MAP, KALA or protein transduction domains (PTDs, PpT620, prolin-rich peptides, arginine-rich peptides, lysine-rich peptides, MPG-peptide(s), Pep-1, L-oligomers, Calcitonin peptide(s), Antennapedia-derived peptides (particularly from Drosophila antennapedia), pAntp, pIsl, FGF, Lactoferrin, Transportan, Buforin-2, Bac715-24, SynB, SynB(1), pVEC, hCT-derived peptides, SAP, protamine, spermine, spermidine, or histones, cationic polysaccharides, including chitosan, polybrene, cationic polymers, including polyethyleneimine (PEI), cationic lipids, including DOTMA: [1-(2,3 -sioleyloxy)propyl)]-N,N,N-trimethylammonium chloride, DMRIE, di-C14-amidine, DOTIM, SAINT, DC-Chol, BGTC, CTAP, DOPC, DODAP, DOPE: Dioleyl phosphatidylethanol-amine, DOSPA, DODAB, DOIC, DMEPC, DOGS: Dioctadecylamidoglicylspermin, DSL Dimyristo-oxypropyl dimethyl hydroxyethyl ammonium bromide, DOTAP: dioleoyloxy-3-(trimethylammonio)propane, DC-6-14: 0,0-ditetradecanoyl-N-(α-trimethylammonioacetyl)diethanolamine chloride, CLIP1: rac-[(2,3-dioctadecyloxypropyl)(2-hydroxyethyl)]-dimethylammonium chloride, CLIP6: rac-[2(2,3-dihexadecyloxypropyl-oxymethyloxy)ethyl]trimethylammonium, CLIPS: rac-[2(2,3-dihexadecyl oxypropyl-oxy succinyl oxy)ethyl]-trimethyl ammonium, oligofectamine, or cationic or polycationic polymers, including modified polyaminoacids, including 13-aminoacid-polymers or reversed polyamides, modified polyethylenes, including PVP (poly(N-ethyl-4-vinylpyridinium bromide)), modified acrylates, including pDMAEMA (poly(dimethylaminoethyl methylacrylate)), modified Amidoamines including pAMAM (poly(amidoamine)), modified polybetaaminoester (PBAE), including diamine end modified 1,4 butanediol diacrylate-co-5-amino-1-pentanol polymers, dendrimers, including polypropylamine dendrimers or pAMAM based dendrimers, polyimine(s), including PEI: poly(ethyleneimine), poly(propyleneimine), polyallylamine, sugar backbone based polymers, including cyclodextrin based polymers, dextran based polymers, Chitosan, etc., silan backbone based polymers, such as PMOXA-PDMS copolymers, etc., Blockpolymers consisting of a combination of one or more cationic blocks selected of a cationic polymer as mentioned before, and of one or more hydrophilic- or hydrophobic blocks (e.g polyethyleneglycole);
or
cationic or polycationic proteins or peptides, selected from following proteins or peptides having the following total formula (I): (Arg)l;(Lys)m;(His)n;(Orn)o;(Xaa)x, wherein 1+m+n+o+x=8-15, and 1, m, n or o independently of each other may be any number selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15, provided that the overall content of Arg, Lys, His and Orn represents at least 50% of all amino acids of the oligopeptide; and Xaa may be any amino acid selected from native (=naturally occurring) or non-native amino acids except of Arg, Lys, His or Orn; and x may be any number selected from 0, 1, 2, 3 or 4, provided, that the overall content of Xaa does not exceed 50% of all amino acids of the oligopeptide; or
nucleic acids having the formula (II): GAmGn, wherein: G is guanosine, uracil or an analogue of guanosine or uracil; X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above-mentioned nucleotides; 1 is an integer from 1 to 40, wherein when 1=1 G is guanosine or an analogue thereof, when 1>1 at least 50% of the nucleotides are guanosine or an analogue thereof; m is an integer and is at least 3; wherein when m=3 X is uracil or an analogue thereof, when m>3 at least 3 successive uracils or analogues of uracil occur; n is an integer from 1 to 40, wherein when n=1 G is guanosine or an analogue thereof, when n>1 at least 50% of the nucleotides are guanosine or an analogue thereof;
or
nucleic acids having the formula (III): ClXmCn, wherein: C is cytosine, uracil or an analogue of cytosine or uracil; X is guanosine, uracil, adenosine, thymidine, cytosine or an analogue of the above-mentioned nucleotides; 1 is an integer from 1 to 40, wherein when 1=1 C is cytosine or an analogue thereof, when 1>1 at least 50% of the nucleotides are cytosine or an analogue thereof; m is an integer and is at least 3; wherein when m=3 X is uracil or an analogue thereof, when m>3 at least 3 successive uracils or analogues of uracil occur; n is an integer from 1 to 40, wherein when n=1 C is cytosine or an analogue thereof, when n>1 at least 50% of the nucleotides are cytosine or an analogue thereof.
20. Vaccine, comprising an active (immunostimulatory) composition according to any of claims 1 to 19.
21. Vaccine according to claim 20, wherein the active (immunostimulatory) composition according to any of claims 1 to 19 elicits an adaptive immune response.
22. Vaccine according to any of claims 20 and 21, wherein the vaccine further comprises a pharmaceutically acceptable carrier.
23. Use of an active (immunostimulatory) composition according to any of claims 1 to 19 for preparing a vaccine for the treatment of lung cancer, preferably of a non-small-cell lung cancer (NSCLC) related condition, more preferably of conditions related to the three main sub-types of non-small-cell lung cancer (NSCLC) including squamous cell lung carcinoma, adenocarcinoma and large cell lung carcinoma.
24. Kit, preferably kits of parts, comprising the active (immunostimulatory) composition according to any of claims 1 to 19, and/or a vaccine according to any of claims 20 to 22, and optionally technical instructions with information on the administration and dosage of the active (immunostimulatory) composition and/or the vaccine.
US15/985,443 2007-10-09 2018-05-21 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc) Abandoned US20180256694A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/985,443 US20180256694A1 (en) 2007-10-09 2018-05-21 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US17/378,954 US20220096616A1 (en) 2007-10-09 2021-07-19 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
PCT/EP2007/008770 WO2009046738A1 (en) 2007-10-09 2007-10-09 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
EPPCT/EP2007/008770 2007-10-09
PCT/EP2008/008503 WO2009046974A2 (en) 2007-10-09 2008-10-08 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US68221310A 2010-07-09 2010-07-09
US13/750,664 US9352028B2 (en) 2007-10-09 2013-01-25 Composition for treating lung cancer, particularly of non-small lung cancers (NSCLC)
US15/145,746 US20160346370A1 (en) 2007-10-09 2016-05-03 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US15/985,443 US20180256694A1 (en) 2007-10-09 2018-05-21 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/145,746 Division US20160346370A1 (en) 2007-10-09 2016-05-03 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US17/378,954 Division US20220096616A1 (en) 2007-10-09 2021-07-19 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Publications (1)

Publication Number Publication Date
US20180256694A1 true US20180256694A1 (en) 2018-09-13

Family

ID=39590300

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/682,213 Abandoned US20100291156A1 (en) 2007-10-09 2008-10-08 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US13/750,664 Active 2029-06-27 US9352028B2 (en) 2007-10-09 2013-01-25 Composition for treating lung cancer, particularly of non-small lung cancers (NSCLC)
US15/145,540 Abandoned US20160317636A1 (en) 2007-10-09 2016-05-03 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US15/145,746 Abandoned US20160346370A1 (en) 2007-10-09 2016-05-03 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US15/985,443 Abandoned US20180256694A1 (en) 2007-10-09 2018-05-21 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US17/378,954 Pending US20220096616A1 (en) 2007-10-09 2021-07-19 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US12/682,213 Abandoned US20100291156A1 (en) 2007-10-09 2008-10-08 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US13/750,664 Active 2029-06-27 US9352028B2 (en) 2007-10-09 2013-01-25 Composition for treating lung cancer, particularly of non-small lung cancers (NSCLC)
US15/145,540 Abandoned US20160317636A1 (en) 2007-10-09 2016-05-03 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US15/145,746 Abandoned US20160346370A1 (en) 2007-10-09 2016-05-03 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Family Applications After (1)

Application Number Title Priority Date Filing Date
US17/378,954 Pending US20220096616A1 (en) 2007-10-09 2021-07-19 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Country Status (20)

Country Link
US (6) US20100291156A1 (en)
EP (2) EP2197481B1 (en)
JP (3) JP5139529B2 (en)
CN (1) CN101820899B (en)
AU (1) AU2008309930B2 (en)
BR (1) BRPI0818318B8 (en)
CA (1) CA2696768C (en)
CY (1) CY1120168T1 (en)
DK (1) DK2197481T3 (en)
ES (1) ES2668926T3 (en)
HR (1) HRP20180698T1 (en)
HU (1) HUE037295T2 (en)
LT (1) LT2197481T (en)
MX (1) MX2010003766A (en)
NO (1) NO2197481T3 (en)
PL (1) PL2197481T3 (en)
PT (1) PT2197481T (en)
RU (1) RU2526510C2 (en)
SI (1) SI2197481T1 (en)
WO (2) WO2009046738A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11667910B2 (en) 2015-05-29 2023-06-06 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration

Families Citing this family (136)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7276489B2 (en) * 2002-10-24 2007-10-02 Idera Pharmaceuticals, Inc. Modulation of immunostimulatory properties of oligonucleotide-based compounds by optimal presentation of 5′ ends
US9393315B2 (en) 2011-06-08 2016-07-19 Nitto Denko Corporation Compounds for targeting drug delivery and enhancing siRNA activity
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
WO2009046739A1 (en) 2007-10-09 2009-04-16 Curevac Gmbh Composition for treating prostate cancer (pca)
WO2010037408A1 (en) * 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
CN107252489A (en) 2009-04-13 2017-10-17 法国健康和医学研究院 HPV particles and application thereof
EP2281579A1 (en) * 2009-08-05 2011-02-09 BioNTech AG Vaccine composition comprising 5'-Cap modified RNA
CN101623503B (en) * 2009-08-06 2011-07-27 中国人民解放军第三军医大学 Composite of survivin and sialylLewis and preparation method and application thereof
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
WO2011069529A1 (en) 2009-12-09 2011-06-16 Curevac Gmbh Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
BR112013002298A2 (en) 2010-07-30 2016-05-24 Curevac Gmbh nucleic acid complexation with disulfide crosslinked cationic components for transfection and immune stimulation.
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2012019630A1 (en) 2010-08-13 2012-02-16 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded protein
ES2862955T3 (en) 2010-10-01 2021-10-08 Modernatx Inc Manipulated nucleic acids and methods of using them
WO2012089225A1 (en) 2010-12-29 2012-07-05 Curevac Gmbh Combination of vaccination and inhibition of mhc class i restricted antigen presentation
WO2012116715A1 (en) 2011-03-02 2012-09-07 Curevac Gmbh Vaccination in newborns and infants
WO2012113413A1 (en) 2011-02-21 2012-08-30 Curevac Gmbh Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
US10316332B2 (en) 2011-07-29 2019-06-11 Riken Cell for immunotherapy, including modified nucleic acid construct encoding Wilms tumor gene product
JPWO2013018778A1 (en) * 2011-07-29 2015-03-05 独立行政法人理化学研究所 Immunotherapeutic cells comprising a modified nucleic acid construct encoding a Wilms tumor gene product or fragment thereof, a method for producing the cell, and the nucleic acid construct
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
WO2013052523A1 (en) 2011-10-03 2013-04-11 modeRNA Therapeutics Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
CN104114572A (en) 2011-12-16 2014-10-22 现代治疗公司 Modified nucleoside, nucleotide, and nucleic acid compositions
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
WO2013120498A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded allergenic antigen or an autoimmune self-antigen
WO2013120500A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded tumour antigen
WO2013120499A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly (a) sequence or a polyadenylation signal for increasing the expression of an encoded pathogenic antigen
WO2013120497A1 (en) 2012-02-15 2013-08-22 Curevac Gmbh Nucleic acid comprising or coding for a histone stem-loop and a poly(a) sequence or a polyadenylation signal for increasing the expression of an encoded therapeutic protein
ES2654205T3 (en) 2012-03-27 2018-02-12 Curevac Ag Artificial nucleic acid molecules for enhanced protein or peptide expression
AU2013242403B2 (en) 2012-03-27 2018-10-18 Curevac Ag Artificial nucleic acid molecules
CN108929880A (en) 2012-03-27 2018-12-04 库瑞瓦格股份公司 Artificial nucleic acid molecule comprising 5 ' TOPUTR
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
EP2833920A2 (en) 2012-04-02 2015-02-11 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
PL2922554T3 (en) 2012-11-26 2022-06-20 Modernatx, Inc. Terminally modified rna
CN103961307B (en) 2013-02-05 2019-11-29 日东电工株式会社 For percutaneous administration of with WT1 peptide cancer vaccine composition
KR102050931B1 (en) * 2013-02-05 2019-12-02 닛토덴코 가부시키가이샤 Tape preparation of wt1 peptide cancer vaccine for transdermal administration
WO2014127006A1 (en) 2013-02-12 2014-08-21 Texas Tech University System Composition and method for diagnosis and immunotherapy of lung cancer
DK2958588T3 (en) * 2013-02-22 2017-11-20 Curevac Ag Combination of vaccination and inhibition of PD-1 pathway
EP3292873B1 (en) 2013-02-22 2019-05-01 CureVac AG Combination of vaccination and inhibition of the pd-1 pathway
US10077439B2 (en) 2013-03-15 2018-09-18 Modernatx, Inc. Removal of DNA fragments in mRNA production process
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
WO2015024665A1 (en) 2013-08-21 2015-02-26 Curevac Gmbh Rabies vaccine
CN105473157A (en) 2013-08-21 2016-04-06 库瑞瓦格股份公司 Combination vaccine
KR20160044566A (en) 2013-08-21 2016-04-25 큐어백 아게 Respiratory syncytial virus (RSV) vaccine
ES2702466T3 (en) 2013-08-21 2019-03-01 Curevac Ag Procedure to increase the expression of proteins encoded by RNA
MX2016002153A (en) * 2013-08-21 2017-03-01 Curevac Ag Composition and vaccine for treating prostate cancer.
EP3574916A1 (en) 2013-08-21 2019-12-04 CureVac AG Composition and vaccine for treating lung cancer
SG10201801429VA (en) * 2013-08-21 2018-03-28 Curevac Ag Composition and vaccine for treating lung cancer
PT3046583T (en) 2013-09-18 2019-05-30 Aura Biosciences Inc Virus-like particle conjugates for treatment of tumors
WO2015048744A2 (en) 2013-09-30 2015-04-02 Moderna Therapeutics, Inc. Polynucleotides encoding immune modulating polypeptides
EP3052521A1 (en) 2013-10-03 2016-08-10 Moderna Therapeutics, Inc. Polynucleotides encoding low density lipoprotein receptor
WO2015062738A1 (en) 2013-11-01 2015-05-07 Curevac Gmbh Modified rna with decreased immunostimulatory properties
WO2015101415A1 (en) 2013-12-30 2015-07-09 Curevac Gmbh Artificial nucleic acid molecules
BR112016014462A2 (en) 2013-12-30 2017-10-24 Curevac Ag artificial nucleic acid molecules
US11254951B2 (en) 2014-12-30 2022-02-22 Curevac Ag Artificial nucleic acid molecules
SG11201604198YA (en) 2013-12-30 2016-07-28 Curevac Ag Methods for rna analysis
US10307472B2 (en) 2014-03-12 2019-06-04 Curevac Ag Combination of vaccination and OX40 agonists
EP3129050A2 (en) 2014-04-01 2017-02-15 CureVac AG Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
SG11201608798YA (en) 2014-04-23 2016-11-29 Modernatx Inc Nucleic acid vaccines
BR112016026980B1 (en) 2014-06-10 2022-05-03 Curevac Real Estate Gmbh Method for synthesizing an RNA molecule of a given sequence
WO2016077125A1 (en) 2014-11-10 2016-05-19 Moderna Therapeutics, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
EP4324473A2 (en) 2014-11-10 2024-02-21 ModernaTX, Inc. Multiparametric nucleic acid optimization
EP3708668B1 (en) 2014-12-12 2022-07-27 CureVac AG Artificial nucleic acid molecules for improved protein expression
US10653768B2 (en) 2015-04-13 2020-05-19 Curevac Real Estate Gmbh Method for producing RNA compositions
EP3283125B1 (en) 2015-04-17 2021-12-29 CureVac Real Estate GmbH Lyophilization of rna
SG10201907164SA (en) 2015-04-22 2019-09-27 Curevac Ag Rna containing composition for treatment of tumor diseases
WO2016172219A1 (en) * 2015-04-24 2016-10-27 The Arizona Board Of Regents On Behalf Of The University Of Arizona Methods and compositions for inhibition of toll-like receptors (tlrs)-mediated inflammation
ES2897823T3 (en) 2015-04-30 2022-03-02 Curevac Ag Immobilized poly(N)polymerase
US11559570B2 (en) 2015-05-15 2023-01-24 CureVac SE Prime-boost regimens involving administration of at least one mRNA construct
SG11201708540VA (en) 2015-05-20 2017-12-28 Curevac Ag Dry powder composition comprising long-chain rna
CN107530448A (en) 2015-05-20 2018-01-02 库瑞瓦格股份公司 Include long-chain RNA dry powder composite
US11608513B2 (en) 2015-05-29 2023-03-21 CureVac SE Method for adding cap structures to RNA using immobilized enzymes
WO2017009376A1 (en) 2015-07-13 2017-01-19 Curevac Ag Method of producing rna from circular dna and corresponding template dna
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US11225682B2 (en) 2015-10-12 2022-01-18 Curevac Ag Automated method for isolation, selection and/or detection of microorganisms or cells comprised in a solution
ES2914225T3 (en) 2015-10-16 2022-06-08 Modernatx Inc Modified phosphate bond mRNA cap analogs
WO2017066797A1 (en) 2015-10-16 2017-04-20 Modernatx, Inc. Trinucleotide mrna cap analogs
EP3364950A4 (en) 2015-10-22 2019-10-23 ModernaTX, Inc. Tropical disease vaccines
EP3364981A4 (en) 2015-10-22 2019-08-07 ModernaTX, Inc. Human cytomegalovirus vaccine
EP4011451A1 (en) 2015-10-22 2022-06-15 ModernaTX, Inc. Metapneumovirus mrna vaccines
WO2017081110A1 (en) 2015-11-09 2017-05-18 Curevac Ag Rotavirus vaccines
SG11201804398XA (en) 2015-12-22 2018-07-30 Curevac Ag Method for producing rna molecule compositions
EP3394280A1 (en) 2015-12-23 2018-10-31 CureVac AG Method of rna in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
SG11201806340YA (en) 2016-02-17 2018-09-27 Curevac Ag Zika virus vaccine
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
WO2017186928A1 (en) 2016-04-29 2017-11-02 Curevac Ag Rna encoding an antibody
WO2017191274A2 (en) 2016-05-04 2017-11-09 Curevac Ag Rna encoding a therapeutic protein
WO2017191264A1 (en) 2016-05-04 2017-11-09 Curevac Ag Nucleic acid molecules and uses thereof
EP3458611B1 (en) * 2016-05-17 2023-11-08 Genecentric Therapeutics, Inc. Methods for subtyping of lung squamous cell carcinoma
US20190194760A1 (en) 2016-05-25 2019-06-27 Curevac Ag Novel biomarkers
EP3468613A1 (en) 2016-06-09 2019-04-17 CureVac AG Hybrid carriers for nucleic acid cargo
CN109715196A (en) 2016-06-13 2019-05-03 转矩医疗股份有限公司 For promoting the composition and method of immune cell function
CN116837052A (en) 2016-09-14 2023-10-03 摩登纳特斯有限公司 High-purity RNA composition and preparation method thereof
EP3528821A4 (en) 2016-10-21 2020-07-01 ModernaTX, Inc. Human cytomegalovirus vaccine
WO2018096179A1 (en) 2016-11-28 2018-05-31 Curevac Ag Method for purifying rna
EP3548005A4 (en) 2016-11-29 2020-06-17 Puretech Health LLC Exosomes for delivery of therapeutic agents
WO2018104540A1 (en) 2016-12-08 2018-06-14 Curevac Ag Rnas for wound healing
EP3808380A1 (en) 2016-12-08 2021-04-21 CureVac AG Rna for treatment or prophylaxis of a liver disease
EP3551193A4 (en) 2016-12-08 2020-08-19 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11524066B2 (en) 2016-12-23 2022-12-13 CureVac SE Henipavirus vaccine
EP3558354A1 (en) 2016-12-23 2019-10-30 CureVac AG Lassa virus vaccine
US11141476B2 (en) 2016-12-23 2021-10-12 Curevac Ag MERS coronavirus vaccine
MX2019009070A (en) * 2017-02-01 2019-10-30 Modernatx Inc Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides.
EP3582790A4 (en) 2017-02-16 2020-11-25 ModernaTX, Inc. High potency immunogenic compositions
UY37621A (en) 2017-02-28 2018-09-28 Sanofi Sa THERAPEUTIC ARN THAT CODIFIES CYTOKINS
CN110392577A (en) 2017-03-17 2019-10-29 库尔维科公司 For combining the RNA vaccine and immunologic test point inhibitor of anti-cancer therapies
CN110914433A (en) 2017-03-24 2020-03-24 库尔维科公司 Nucleic acids encoding CRISPR-associated proteins and uses thereof
US11357856B2 (en) 2017-04-13 2022-06-14 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
CN110799492B (en) 2017-04-28 2023-06-27 爱康泰生治疗公司 Novel carbonyl lipid and lipid nanoparticle formulations for delivery of nucleic acids
US10034951B1 (en) 2017-06-21 2018-07-31 New England Biolabs, Inc. Use of thermostable RNA polymerases to produce RNAs having reduced immunogenicity
US10988754B2 (en) 2017-07-04 2021-04-27 Cure Vac AG Nucleic acid molecules
US11602557B2 (en) 2017-08-22 2023-03-14 Cure Vac SE Bunyavirales vaccine
AU2018328209A1 (en) 2017-09-05 2020-04-23 Torque Therapeutics, Inc. Therapeutic protein compositions and methods of making and using the same
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
CA3073634A1 (en) 2017-10-19 2019-04-25 Curevac Ag Novel artificial nucleic acid molecules
US11692002B2 (en) 2017-11-08 2023-07-04 CureVac SE RNA sequence adaptation
EP3723796A1 (en) 2017-12-13 2020-10-21 CureVac AG Flavivirus vaccine
SG11202005760PA (en) 2017-12-21 2020-07-29 Curevac Ag Linear double stranded dna coupled to a single support or a tag and methods for producing said linear double stranded dna
AU2019349329A1 (en) * 2018-09-28 2021-04-29 Sumitomo Pharma Co., Ltd. Injectable composition
US11072808B2 (en) 2018-10-04 2021-07-27 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed RNA
CA3114892A1 (en) 2018-10-04 2020-04-09 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed rna
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
RU2696872C1 (en) * 2019-04-11 2019-08-07 Федеральное государственное бюджетное научное учреждение "Томский национальный исследовательский медицинский центр" Российской академии наук" (Томский НИМЦ) Method for predicting an unfavorable outcome in non-small-cell lung cancer
CN112402596B (en) * 2019-08-22 2023-12-08 上海细胞治疗集团有限公司 Polypeptide composition and vaccine
CN110456079B (en) * 2019-09-20 2020-06-02 四川大学华西医院 Application of TAPBP autoantibody detection reagent in preparation of lung cancer screening kit
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
CN111471635B (en) * 2020-04-13 2022-02-15 江南大学 Method for increasing content of nucleic acid in bacillus subtilis
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
BR112023012303A2 (en) 2020-12-22 2024-02-15 CureVac SE RNA VACCINE AGAINST SARS-COV-2 VARIANTS
CN113444730A (en) * 2021-03-17 2021-09-28 昆明市延安医院 Screening and constructing method of primary hepatocyte klotho gene transduction stem cells
CN115998851A (en) * 2022-12-28 2023-04-25 四川康德赛医疗科技有限公司 Individuation mRNA composition, vector, mRNA vaccine and application thereof

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5132418A (en) 1980-02-29 1992-07-21 University Patents, Inc. Process for preparing polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4973679A (en) 1981-03-27 1990-11-27 University Patents, Inc. Process for oligonucleo tide synthesis using phosphormidite intermediates
US4668777A (en) 1981-03-27 1987-05-26 University Patents, Inc. Phosphoramidite nucleoside compounds
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
US4401796A (en) 1981-04-30 1983-08-30 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US5153319A (en) 1986-03-31 1992-10-06 University Patents, Inc. Process for preparing polynucleotides
US5804381A (en) * 1996-10-03 1998-09-08 Cornell Research Foundation Isolated nucleic acid molecule encoding an esophageal cancer associated antigen, the antigen itself, and uses thereof
US5047524A (en) 1988-12-21 1991-09-10 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5700642A (en) 1995-05-22 1997-12-23 Sri International Oligonucleotide sizing using immobilized cleavable primers
DE69923840T2 (en) 1999-09-09 2006-04-06 Curevac Gmbh Transfer of mRNAs using polycationic compounds
DE60128251T2 (en) 2000-11-03 2008-01-10 Washington University Oestrone derivatives with cell protective effect
EP1392341B1 (en) * 2001-06-05 2005-03-30 Curevac GmbH Pharmaceutical composition containing a stabilised mrna which is optimised for translation in the coding regions thereof
US7955845B2 (en) * 2001-11-20 2011-06-07 Dana Farber Cancer Institute, Inc. Modified antigen-presenting cells
DE10229872A1 (en) * 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
WO2005005615A2 (en) 2003-07-08 2005-01-20 Fox Chase Cancer Center Anti-mullerian inhibiting substance type ii receptor (misiir) immunoconjugates to detect and treat cancer
US7842289B2 (en) * 2003-12-24 2010-11-30 Aduro Biotech Recombinant nucleic acid molecules, expression cassettes, and bacteria, and methods of use thereof
DE102004035227A1 (en) * 2004-07-21 2006-02-16 Curevac Gmbh mRNA mixture for vaccination against tumor diseases
DE102004042546A1 (en) * 2004-09-02 2006-03-09 Curevac Gmbh Combination therapy for immune stimulation
DE102005023170A1 (en) 2005-05-19 2006-11-23 Curevac Gmbh Optimized formulation for mRNA
WO2007008780A2 (en) * 2005-07-11 2007-01-18 Globeimmune, Inc. Compositions and methods for eliciting an immune response to escape mutants of targeted therapies
RU2345788C2 (en) * 2005-09-02 2009-02-10 ООО "Научно-практический центр медико-биологических проблем "ГОРМЕЗИС" Laser vaccination method for patients suffering from metastatic cancer type
JP2010507361A (en) 2006-07-31 2010-03-11 キュアバック ゲーエムベーハー Specifically, a nucleic acid represented by the general formula (I): GlXmGn or the general formula (II): ClXmCn as an immunostimulant / adjuvant
DE102006061015A1 (en) 2006-12-22 2008-06-26 Curevac Gmbh Process for the purification of RNA on a preparative scale by HPLC
DE102007001370A1 (en) 2007-01-09 2008-07-10 Curevac Gmbh RNA-encoded antibodies
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
WO2009046739A1 (en) 2007-10-09 2009-04-16 Curevac Gmbh Composition for treating prostate cancer (pca)
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11667910B2 (en) 2015-05-29 2023-06-06 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US11760992B2 (en) 2015-05-29 2023-09-19 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US11834651B2 (en) 2015-05-29 2023-12-05 CureVac Manufacturing GmbH Method for producing and purifying RNA, comprising at least one step of tangential flow filtration

Also Published As

Publication number Publication date
US20100291156A1 (en) 2010-11-18
US20220096616A1 (en) 2022-03-31
PL2197481T3 (en) 2018-10-31
BRPI0818318B1 (en) 2020-10-13
HUE037295T2 (en) 2018-08-28
SI2197481T1 (en) 2018-07-31
CN101820899B (en) 2014-03-12
US20130202645A1 (en) 2013-08-08
RU2010118024A (en) 2011-11-20
JP6012423B2 (en) 2016-10-25
CN101820899A (en) 2010-09-01
JP6031131B2 (en) 2016-11-24
US20160346370A1 (en) 2016-12-01
WO2009046974A3 (en) 2009-06-18
CY1120168T1 (en) 2018-12-12
NO2197481T3 (en) 2018-07-14
LT2197481T (en) 2018-06-25
CA2696768A1 (en) 2009-04-16
WO2009046974A2 (en) 2009-04-16
US20160317636A1 (en) 2016-11-03
ES2668926T3 (en) 2018-05-23
EP2197481A2 (en) 2010-06-23
MX2010003766A (en) 2010-04-30
WO2009046738A1 (en) 2009-04-16
JP2015110613A (en) 2015-06-18
DK2197481T3 (en) 2018-05-22
EP3375454A1 (en) 2018-09-19
RU2526510C2 (en) 2014-08-20
US9352028B2 (en) 2016-05-31
AU2008309930B2 (en) 2013-02-21
PT2197481T (en) 2018-05-10
JP2013067633A (en) 2013-04-18
JP2010540672A (en) 2010-12-24
BRPI0818318B8 (en) 2021-05-25
AU2008309930A1 (en) 2009-04-16
HRP20180698T1 (en) 2018-06-29
CA2696768C (en) 2019-03-26
EP3375454B1 (en) 2021-04-14
EP2197481B1 (en) 2018-02-14
JP5139529B2 (en) 2013-02-06

Similar Documents

Publication Publication Date Title
US20220096616A1 (en) Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)
US20190365879A1 (en) COMPOSITION FOR TREATING PROSTATE CANCER (PCa)
AU2014310932B2 (en) Composition and vaccine for treating lung cancer
US20160166668A1 (en) Composition and vaccine for treating prostate cancer
EP3035955B1 (en) Composition and vaccine for treating lung cancer
EP2195015B1 (en) COMPOSITION FOR TREATING PROSTATE CANCER (PCa)

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: CUREVAC SE, GERMANY

Free format text: CHANGE OF NAME;ASSIGNOR:CUREVAC AG;REEL/FRAME:062522/0043

Effective date: 20220926