US20100291156A1 - 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
US20100291156A1
US20100291156A1 US12/682,213 US68221308A US2010291156A1 US 20100291156 A1 US20100291156 A1 US 20100291156A1 US 68221308 A US68221308 A US 68221308A US 2010291156 A1 US2010291156 A1 US 2010291156A1
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
US12/682,213
Other languages
English (en)
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
Individual
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 Individual filed Critical Individual
Assigned to CUREVAC GMBH reassignment CUREVAC GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOERR, INGMAR, LANDER, THOMAS, PROBST, JOCHEN, BARNER, MARIJKE
Publication of US20100291156A1 publication Critical patent/US20100291156A1/en
Assigned to CUREVAC AG reassignment CUREVAC AG CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: CUREVAC GMBH
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.
  • 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.
  • 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 (CD8 + ) may form a CTL-response. Cytotoxic T cells (CD8 + ) 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-based immunotherapy of cancer “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.
  • 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).
  • 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.
  • 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).
  • 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.
  • 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., O. 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 family (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.
  • 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.
  • 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-A2.1-binding 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-C1”—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.
  • 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.
  • 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-C1 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 protein, 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.
  • 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.
  • 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-
  • 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 at, 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-hexadecy
  • 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, Biochim
  • 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.
  • RNAases ribonucleases
  • 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).
  • 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
  • the so-called poly-A tail is typically 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, preferably about 10 to 100 adenosine nucleotides, more preferably about 20 to 100 adenosine nucleotides or even more preferably about 40 to 80 adenosine nucleotides.
  • 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, preferably about 10 to 100 cytosine nucleotides, more preferably about 20 to 70 cytosine nucleotides or even more preferably about 20 to 60 or even 10 to 40 cytosine nucleotides.
  • 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 determined 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 at, 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 at, 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)CCAN x CCC(U/A)Py x UC(C/U)CC (SEQ ID NO: 28), which is contained in the 3′UTR of the very stable RNA which codes for globin, (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, NY, 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 GeneBank accession number X65327
  • 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 compartment, preferably the cell surface, the endoplasmic reticulum (ER) or the endosomal-lysosomal compartment.
  • 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 Lampl, Tapasin, Erp57, Calretikulin, Calnexin, and further membrane associated proteins or of proteins associated with the endoplasmic reticulum (ER) or the endosomal-lysosomal compartment.
  • 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-
  • MPLTM (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); MURAMETIDETM (Nac-Mur-L-Ala-D-Gln-OCH 3 ); MURAPALMITINETM and D-MURAPALMITINETM (Nac-Mur-L-Thr-D-isoGln-s
  • 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 O,O-ditetradecanoyl-N-( ⁇ -trimethylammonioacetyl)diethanolamine chloride
  • CLIP1 rac-[(2,3-dioctadecyloxypropyl)(2-hydroxyethyl)]
  • modified polyaminoacids such as ⁇ -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.
  • 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 , Arg 8 , Arg 9 , Arg 7 , H 3 R 9 , R 9 H 3 , H 3 R 9 H 3 , YSSR 9 SSY, (RKH) 4 , Y(RKH) 2 R, etc.
  • 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 groups 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. 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.
  • 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, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, or due to its binding affinity (as ligands) to murine Toll-like receptors TLR1, TLR2, TLR3, TLR4, TLR5, 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, TLR5, 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 alia, 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.
  • FIG. 10 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. 10B ), C) and D) show a comparison of the corresponding regions 325-408:
  • FIG. 12 depicts a RNA sequence (SEQ ID NO: 12) (starting sequence based on the wildtype) encoding CEA (CEA (carcinoembryonic antigen) HsCEACAM5); GC Content: 52.20%; Length: 2109 bp.
  • CEA CEA (carcinoembryonic antigen) HsCEACAM5)
  • 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.
  • Survivin baculoviral IAP repeat-containing 5, BIRC5
  • 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-ESO-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.
  • 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 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.
  • 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.
  • 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.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Oncology (AREA)
  • Pulmonology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
US12/682,213 2007-10-09 2008-10-08 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc) Abandoned US20100291156A1 (en)

Applications Claiming Priority (3)

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)
EPPCTEP2007008770 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)

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2008/008503 A-371-Of-International WO2009046974A2 (en) 2007-10-09 2008-10-08 Composition for treating lung cancer, particularly of non-small lung cancers (nsclc)

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/750,664 Continuation US9352028B2 (en) 2007-10-09 2013-01-25 Composition for treating lung cancer, particularly of non-small lung cancers (NSCLC)

Publications (1)

Publication Number Publication Date
US20100291156A1 true US20100291156A1 (en) 2010-11-18

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 Expired - Fee Related US9352028B2 (en) 2007-10-09 2013-01-25 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)
US15/145,540 Abandoned US20160317636A1 (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 After (5)

Application Number Title Priority Date Filing Date
US13/750,664 Expired - Fee Related US9352028B2 (en) 2007-10-09 2013-01-25 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)
US15/145,540 Abandoned US20160317636A1 (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)

Country Status (20)

Country Link
US (6) US20100291156A1 (pt)
EP (2) EP3375454B1 (pt)
JP (3) JP5139529B2 (pt)
CN (1) CN101820899B (pt)
AU (1) AU2008309930B2 (pt)
BR (1) BRPI0818318B8 (pt)
CA (1) CA2696768C (pt)
CY (1) CY1120168T1 (pt)
DK (1) DK2197481T3 (pt)
ES (1) ES2668926T3 (pt)
HR (1) HRP20180698T1 (pt)
HU (1) HUE037295T2 (pt)
LT (1) LT2197481T (pt)
MX (1) MX2010003766A (pt)
NO (1) NO2197481T3 (pt)
PL (1) PL2197481T3 (pt)
PT (1) PT2197481T (pt)
RU (1) RU2526510C2 (pt)
SI (1) SI2197481T1 (pt)
WO (2) WO2009046738A1 (pt)

Cited By (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US8703906B2 (en) 2009-09-03 2014-04-22 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8968746B2 (en) 2010-07-30 2015-03-03 Curevac Gmbh Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9234013B2 (en) 2010-08-13 2016-01-12 Curevac Ag 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
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US20160168227A1 (en) * 2013-08-21 2016-06-16 Curevac Ag Composition and vaccine for treating lung cancer
US9402887B2 (en) 2007-10-09 2016-08-02 Curevac Ag Composition for treating prostate cancer (PCa)
US9421255B2 (en) 2011-02-21 2016-08-23 Curevac Ag Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9447431B2 (en) 2012-02-15 2016-09-20 Curevac Ag 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
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
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
US9518989B2 (en) 2013-02-12 2016-12-13 Texas Tech University System Composition and method for diagnosis and immunotherapy of lung cancer
US9572874B2 (en) 2008-09-30 2017-02-21 Curevac Ag Composition comprising a complexed (M)RNA and a naked mRNA for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US9616084B2 (en) 2009-12-09 2017-04-11 Curevac Ag Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
US9623095B2 (en) 2011-03-02 2017-04-18 Curevac Ag Vaccination in newborns and infants
US9669089B2 (en) 2012-02-15 2017-06-06 Curevac Ag 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
US9683233B2 (en) 2012-03-27 2017-06-20 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
US9688729B2 (en) 2013-08-21 2017-06-27 Curevac Ag Respiratory syncytial virus (RSV) vaccine
US9737595B2 (en) 2010-12-29 2017-08-22 Curevac Ag Combination of vaccination and inhibition of MHC class I restricted antigen presentation
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US9890391B2 (en) 2012-03-27 2018-02-13 Curevac Ag RNA vector with an open reading frame, an albumin 3′-UTR, and a histone stem loop
US9974845B2 (en) 2013-02-22 2018-05-22 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US10010592B2 (en) 2012-02-15 2018-07-03 Curevac Ag 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
US10047375B2 (en) 2013-12-30 2018-08-14 Curevac Ag Artificial nucleic acid molecules
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10080809B2 (en) 2012-03-27 2018-09-25 Curevac Ag Artificial nucleic acid molecules comprising a 5′TOP UTR
US10111967B2 (en) 2007-09-04 2018-10-30 Curevac Ag Complexes of RNA and cationic peptides for transfection and for immunostimulation
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
US10232024B2 (en) 2012-02-15 2019-03-19 Curevac Ag 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
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US10293058B2 (en) 2015-04-22 2019-05-21 Curevac Ag RNA containing composition for treatment of tumor diseases
US10293060B2 (en) 2013-08-21 2019-05-21 Curevac Ag Method for increasing expression of RNA-encoded proteins
US10307472B2 (en) 2014-03-12 2019-06-04 Curevac Ag Combination of vaccination and OX40 agonists
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10369216B2 (en) 2014-04-01 2019-08-06 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US10501768B2 (en) 2015-07-13 2019-12-10 Curevac Ag Method of producing RNA from circular DNA and corresponding template DNA
US10517827B2 (en) 2015-05-20 2019-12-31 Curevac Ag Dry powder composition comprising long-chain RNA
US10588959B2 (en) 2013-08-21 2020-03-17 Curevac Ag Combination vaccine
US10648017B2 (en) 2013-12-30 2020-05-12 Curevac Real Estate Gmbh Methods for RNA analysis
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
US10653768B2 (en) 2015-04-13 2020-05-19 Curevac Real Estate Gmbh Method for producing RNA compositions
US10653712B2 (en) 2016-09-14 2020-05-19 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US10682426B2 (en) 2013-08-21 2020-06-16 Curevac Ag Rabies vaccine
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US10729654B2 (en) 2015-05-20 2020-08-04 Curevac Ag Dry powder composition comprising long-chain RNA
US10760070B2 (en) 2015-05-29 2020-09-01 Curevac Real Estate Gmbh Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US10780054B2 (en) 2015-04-17 2020-09-22 Curevac Real Estate Gmbh Lyophilization of RNA
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10837039B2 (en) 2014-06-10 2020-11-17 Curevac Real Estate Gmbh Methods and means for enhancing RNA production
US10898584B2 (en) 2013-11-01 2021-01-26 Curevac Ag Modified RNA with decreased immunostimulatory properties
CN112402596A (zh) * 2019-08-22 2021-02-26 上海细胞治疗集团有限公司 多肽组合物及疫苗
US10988754B2 (en) 2017-07-04 2021-04-27 Cure Vac AG Nucleic acid molecules
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11141476B2 (en) 2016-12-23 2021-10-12 Curevac Ag MERS coronavirus vaccine
US11141474B2 (en) 2016-05-04 2021-10-12 Curevac Ag Artificial nucleic acid molecules encoding a norovirus antigen and uses thereof
US11149278B2 (en) 2014-12-12 2021-10-19 Curevac Ag Artificial nucleic acid molecules for improved protein expression
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
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US11254951B2 (en) 2014-12-30 2022-02-22 Curevac Ag Artificial nucleic acid molecules
US11279923B2 (en) 2016-11-28 2022-03-22 Curevac Ag Method for purifying RNA
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
US11357856B2 (en) 2017-04-13 2022-06-14 Acuitas Therapeutics, Inc. Lipids for delivery of active agents
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA vaccines
US11384375B2 (en) 2015-04-30 2022-07-12 Curevac Ag Immobilized poly(n)polymerase
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
US11464847B2 (en) 2016-12-23 2022-10-11 Curevac Ag Lassa virus vaccine
US11464836B2 (en) 2016-12-08 2022-10-11 Curevac Ag RNA for treatment or prophylaxis of a liver disease
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
US11524066B2 (en) 2016-12-23 2022-12-13 CureVac SE Henipavirus vaccine
US11525158B2 (en) 2017-12-21 2022-12-13 CureVac SE Linear double stranded DNA coupled to a single support or a tag and methods for producing said linear double stranded DNA
US11542490B2 (en) 2016-12-08 2023-01-03 CureVac SE RNAs for wound healing
US11559570B2 (en) 2015-05-15 2023-01-24 CureVac SE Prime-boost regimens involving administration of at least one mRNA construct
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
US11602557B2 (en) 2017-08-22 2023-03-14 Cure Vac SE Bunyavirales vaccine
US11608513B2 (en) 2015-05-29 2023-03-21 CureVac SE Method for adding cap structures to RNA using immobilized enzymes
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11684665B2 (en) 2015-12-22 2023-06-27 CureVac SE Method for producing RNA molecule compositions
US11690910B2 (en) 2012-01-31 2023-07-04 CureVac SE Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
US11692002B2 (en) 2017-11-08 2023-07-04 CureVac SE RNA sequence adaptation
US11697816B2 (en) 2013-12-30 2023-07-11 CureVac SE Artificial nucleic acid molecules
US11723967B2 (en) 2016-02-17 2023-08-15 CureVac SE Zika virus vaccine
US11739335B2 (en) 2017-03-24 2023-08-29 CureVac SE Nucleic acids encoding CRISPR-associated proteins and uses thereof
US11820728B2 (en) 2017-04-28 2023-11-21 Acuitas Therapeutics, Inc. Carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
US11931406B2 (en) 2017-12-13 2024-03-19 CureVac SE Flavivirus vaccine
US11976019B2 (en) 2020-07-16 2024-05-07 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
US11975064B2 (en) 2011-03-02 2024-05-07 CureVac SE Vaccination with mRNA-coded antigens
WO2024097894A1 (en) * 2022-11-04 2024-05-10 RNAimmune, Inc. Compositions and methods of ribonucleic acid vaccines encoding nye-so-1
US12036277B2 (en) 2011-03-02 2024-07-16 CureVac SE Vaccination with mRNA-coded antigens

Families Citing this family (38)

* 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
US9724404B2 (en) 2009-04-13 2017-08-08 INSERM (Institut National de la Santé et de la Recherche Médicale) HPV particles and uses thereof
EP2281579A1 (en) * 2009-08-05 2011-02-09 BioNTech AG Vaccine composition comprising 5'-Cap modified RNA
CN101623503B (zh) * 2009-08-06 2011-07-27 中国人民解放军第三军医大学 生存素与路易斯寡糖的复合物及其制备方法和应用
US10196637B2 (en) 2011-06-08 2019-02-05 Nitto Denko Corporation Retinoid-lipid drug carrier
AU2012291101B2 (en) 2011-07-29 2016-04-07 Riken Cell for use in immunotherapy which contains modified nucleic acid construct encoding Wilms tumor gene product or fragment thereof, method for producing said cell, and said nucleic acid construct
US10316332B2 (en) 2011-07-29 2019-06-11 Riken Cell for immunotherapy, including modified nucleic acid construct encoding Wilms tumor gene product
CA2841016A1 (en) 2013-02-05 2014-08-05 Nitto Denko Corporation Wt1 peptide cancer vaccine composition for transdermal administration
RU2697443C2 (ru) * 2013-02-05 2019-08-14 Нитто Денко Корпорейшн Препарат противораковой вакцины, содержащий пептид wt1, в форме ленты трансдермального введения
BR122021025267B1 (pt) 2013-02-22 2023-12-05 Curevac Ag Partes de kit e composição farmacêutica compreendendo um inibidor da via de pd-1
EP2971165A4 (en) 2013-03-15 2016-11-23 Moderna Therapeutics Inc DISSOLUTION OF DNA FRAGMENTS IN MRNA MANUFACTURING METHODS
SG11201510751YA (en) * 2013-08-21 2016-03-30 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
WO2015042325A1 (en) 2013-09-18 2015-03-26 Aura Biosciences, Inc. Virus-like particle conjugates for diagnosis and treatment of tumors
WO2016077125A1 (en) 2014-11-10 2016-05-19 Moderna Therapeutics, Inc. Alternative nucleic acid molecules containing reduced uracil content and uses thereof
EP3218508A4 (en) * 2014-11-10 2018-04-18 Modernatx, Inc. Multiparametric nucleic acid optimization
US11866754B2 (en) 2015-10-16 2024-01-09 Modernatx, Inc. Trinucleotide mRNA cap analogs
US20190218546A1 (en) 2015-10-16 2019-07-18 Modernatx, Inc. Mrna cap analogs with modified phosphate linkage
CA3024744A1 (en) * 2016-05-17 2017-11-23 Genecentric Therapeutics, Inc. Methods for subtyping of lung squamous cell carcinoma
US20190194760A1 (en) 2016-05-25 2019-06-27 Curevac Ag Novel biomarkers
CA3029813A1 (en) 2016-06-13 2017-12-21 Torque Therapeutics, Inc. Methods and compositions for promoting immune cell function
AU2017368050A1 (en) 2016-11-29 2019-06-20 Puretech Lyt, Inc. Exosomes for delivery of therapeutic agents
CA3051252A1 (en) 2017-02-01 2018-08-09 Modernatx, Inc. Immunomodulatory therapeutic mrna compositions encoding activating oncogene mutation peptides
KR20190124750A (ko) 2017-02-28 2019-11-05 사노피 치료적 rna
WO2018167320A1 (en) 2017-03-17 2018-09-20 Curevac Ag Rna vaccine and immune checkpoint inhibitors for combined anticancer therapy
US10034951B1 (en) 2017-06-21 2018-07-31 New England Biolabs, Inc. Use of thermostable RNA polymerases to produce RNAs having reduced immunogenicity
US11524033B2 (en) 2017-09-05 2022-12-13 Torque Therapeutics, Inc. Therapeutic protein compositions and methods of making and using the same
KR20200071081A (ko) 2017-10-19 2020-06-18 큐어백 아게 신규 인공 핵산 분자
WO2020067453A1 (ja) * 2018-09-28 2020-04-02 大日本住友製薬株式会社 注射用組成物
WO2020072914A1 (en) 2018-10-04 2020-04-09 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed rna
US11072808B2 (en) 2018-10-04 2021-07-27 New England Biolabs, Inc. Methods and compositions for increasing capping efficiency of transcribed RNA
RU2696872C1 (ru) * 2019-04-11 2019-08-07 Федеральное государственное бюджетное научное учреждение "Томский национальный исследовательский медицинский центр" Российской академии наук" (Томский НИМЦ) Способ прогнозирования неблагоприятного исхода при немелкоклеточном раке легкого
CN110456079B (zh) * 2019-09-20 2020-06-02 四川大学华西医院 Tapbp自身抗体检测试剂在制备肺癌筛查试剂盒中的用途
CN111471635B (zh) * 2020-04-13 2022-02-15 江南大学 一种提高枯草芽孢杆菌核酸含量的方法
CN113444730A (zh) * 2021-03-17 2021-09-28 昆明市延安医院 一种原发性肝细胞klotho基因转导干细胞筛选构建方法
KR20230053998A (ko) 2021-10-15 2023-04-24 (주)지노믹트리 mRNA 발현을 위한 유전자 구조체
CN115998851A (zh) * 2022-12-28 2023-04-25 四川康德赛医疗科技有限公司 一种个体化mRNA组合物、载体、mRNA疫苗及其应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US20050032730A1 (en) * 2001-06-05 2005-02-10 Florian Von Der Mulbe Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
US20050250723A1 (en) * 2002-07-03 2005-11-10 Ingmar Hoerr Immunostimulation by chemically modified RNA
US20080025944A1 (en) * 2004-09-02 2008-01-31 Cure Vac Gmbh Combination Therapy for Immunostimulation
US20080171711A1 (en) * 2004-07-21 2008-07-17 Curevac Gmbh Mrna Mixture For Vaccinating Against Tumoral Diseases

Family Cites Families (26)

* 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
US4500707A (en) 1980-02-29 1985-02-19 University Patents, Inc. Nucleosides useful in the preparation of polynucleotides
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4415732A (en) 1981-03-27 1983-11-15 University Patents, Inc. Phosphoramidite compounds and processes
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
US4373071A (en) 1981-04-30 1983-02-08 City Of Hope Research Institute Solid-phase synthesis of polynucleotides
US4401796A (en) 1981-04-30 1983-08-30 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
US5262530A (en) 1988-12-21 1993-11-16 Applied Biosystems, Inc. Automated system for polynucleotide synthesis and purification
US5047524A (en) 1988-12-21 1991-09-10 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 (de) 1999-09-09 2006-04-06 Curevac Gmbh Transfer von mRNAs unter Verwendung von polykationischen Verbindungen
EP1330467B1 (en) 2000-11-03 2007-05-02 Washington University Modified, hydroxy-substituted aromatic structures having cytoprotective activity
US7955845B2 (en) * 2001-11-20 2011-06-07 Dana Farber Cancer Institute, Inc. Modified antigen-presenting cells
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
DE102005023170A1 (de) 2005-05-19 2006-11-23 Curevac Gmbh Optimierte Formulierung für mRNA
EP1906997A2 (en) * 2005-07-11 2008-04-09 Globeimmune, Inc. Compositions and methods for eliciting an immune response to escape mutants of targeted therapies
RU2345788C2 (ru) * 2005-09-02 2009-02-10 ООО "Научно-практический центр медико-биологических проблем "ГОРМЕЗИС" Способ лазерной вакцинации больных с метастатическими формами рака
JP2010507361A (ja) 2006-07-31 2010-03-11 キュアバック ゲーエムベーハー 具体的には免疫刺激剤/アジュバントとしての、一般式(I):GlXmGn、または一般式(II):ClXmCnで表される核酸
DE102006061015A1 (de) 2006-12-22 2008-06-26 Curevac Gmbh Verfahren zur Reinigung von RNA im präparativen Maßstab mittels HPLC
DE102007001370A1 (de) 2007-01-09 2008-07-10 Curevac Gmbh RNA-kodierte Antikörper
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

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
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
US20050032730A1 (en) * 2001-06-05 2005-02-10 Florian Von Der Mulbe Pharmaceutical composition containing a stabilised mRNA optimised for translation in its coding regions
US20110269950A1 (en) * 2001-06-05 2011-11-03 Curevac Gmbh PHARMACEUTICAL COMPOSITION CONTAINING A STABILISED mRNA OPTIMISED FOR TRANSLATION IN ITS CODING REGIONS
US20050250723A1 (en) * 2002-07-03 2005-11-10 Ingmar Hoerr Immunostimulation by chemically modified RNA
US20080171711A1 (en) * 2004-07-21 2008-07-17 Curevac Gmbh Mrna Mixture For Vaccinating Against Tumoral Diseases
US20080025944A1 (en) * 2004-09-02 2008-01-31 Cure Vac Gmbh Combination Therapy for Immunostimulation

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
Godelaine et al. A new tumor-specific antigen encoded by MAGE-C2 and presented to cytolytic T lymphocytes by HLA-B44. Cancer Immunology, Immunotherapy: CII, Vol. 56, No. 6, pages 753-759, June 2007, published online November 10, 2006. *
Jungbluth et al. CT7 (MAGE-C1) antigen expression in normal and neoplastic tissues. International Journal of Cancer, Vol. 99, pages 839-845, 2002. *
Kobayashi et al. Peptide epitope identification for tumor-reactive CD4 T cells. Current Opinion in Immunology, Vol. 20, pages 221-227, May 2008. *
Mitchell et al. mRNA turnover. Current Opinion in Cell Biology, Vol. 13, No. 3, pages 320-325, 2001. *
Mulryan et al. Attenuated recombinant vaccinia virus expressing oncofetal antigen (Tumor-associated antigen) 5T4 induces active therapy of established tumors. Molecular Cancer Therapeutics, Vol. 1, pages 1129-1137, 2002. *
Pesole et al. UTRdb and UTRsite: specialized databases of sequences and functional elements of 5' and 3' untranslated regions of eukaryotic mRNAs. Update 2002. Nucleic Acids Research, Vol. 30, No. 1, pages 335-340, 2002. *
Ross, Control of messenger RNA stability in higher eukaryotes. TIG, Vol. 12, No. 5, pages 171-175, 1996. *
Tajima et al. Identification of an epitope from the epithelial cell adhesion molecule eliciting HLA-A*2402-restricted cytotoxic T-lymphocyte responses. Tissue Antigens, Vol. 64, pages 650-659, 2004. *
Tourriere et al., mRNA degradation machines in eukaryotic cells. Biochimie, Vol. 84, pages 821-837, 2002. *
Wilusz et al., Bringing the role of mRNA decay in the control of gene expression into focus. TIG, Vol. 20, No. 10, pages 491-497, 2004. *
Xiang et al. A DNA vaccine targeting survivin combines apoptosis with suppression of angiogenesis in lung tumor eradication. Cancer Research, Vol. 65, No. 2, pages 553-561, 2005. *

Cited By (222)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10111967B2 (en) 2007-09-04 2018-10-30 Curevac Ag Complexes of RNA and cationic peptides for transfection and for immunostimulation
US10434154B2 (en) 2007-10-09 2019-10-08 Curevac Ag Composition for treating prostate cancer (PCa)
US9402887B2 (en) 2007-10-09 2016-08-02 Curevac Ag Composition for treating prostate cancer (PCa)
US9572874B2 (en) 2008-09-30 2017-02-21 Curevac Ag Composition comprising a complexed (M)RNA and a naked mRNA for providing or enhancing an immunostimulatory response in a mammal and uses thereof
US9907862B2 (en) 2009-09-03 2018-03-06 Curevac Ag Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US9314535B2 (en) 2009-09-03 2016-04-19 Curevac Ag Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US10751424B2 (en) 2009-09-03 2020-08-25 Curevac Ag Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US8703906B2 (en) 2009-09-03 2014-04-22 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US9616084B2 (en) 2009-12-09 2017-04-11 Curevac Ag Mannose-containing solution for lyophilization, transfection and/or injection of nucleic acids
US8968746B2 (en) 2010-07-30 2015-03-03 Curevac Gmbh Complexation of nucleic acids with disulfide-crosslinked cationic components for transfection and immunostimulation
US9447164B2 (en) 2010-08-06 2016-09-20 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9937233B2 (en) 2010-08-06 2018-04-10 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9181319B2 (en) 2010-08-06 2015-11-10 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8822663B2 (en) 2010-08-06 2014-09-02 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9839697B2 (en) 2010-08-13 2017-12-12 Curevac Ag 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
US9234013B2 (en) 2010-08-13 2016-01-12 Curevac Ag 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
US10653799B2 (en) 2010-08-13 2020-05-19 Curevac Ag 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
US9701965B2 (en) 2010-10-01 2017-07-11 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9657295B2 (en) 2010-10-01 2017-05-23 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US10064959B2 (en) 2010-10-01 2018-09-04 Modernatx, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US9334328B2 (en) 2010-10-01 2016-05-10 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US11458193B2 (en) 2010-12-29 2022-10-04 Curevac Ag Combination of vaccination and inhibition of MHC class I restricted antigen presentation
US9737595B2 (en) 2010-12-29 2017-08-22 Curevac Ag Combination of vaccination and inhibition of MHC class I restricted antigen presentation
US9421255B2 (en) 2011-02-21 2016-08-23 Curevac Ag Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
US10568958B2 (en) 2011-02-21 2020-02-25 Curevac Ag Vaccine composition comprising complexed immunostimulatory nucleic acids and antigens packaged with disulfide-linked polyethyleneglycol/peptide conjugates
US10596252B2 (en) 2011-03-02 2020-03-24 Curevac Ag Vaccination in newborns and infants
US11672856B2 (en) 2011-03-02 2023-06-13 CureVac SE Vaccination in newborns and infants
US10729761B2 (en) 2011-03-02 2020-08-04 Curevac Ag Vaccination in newborns and infants
US12036277B2 (en) 2011-03-02 2024-07-16 CureVac SE Vaccination with mRNA-coded antigens
US11975064B2 (en) 2011-03-02 2024-05-07 CureVac SE Vaccination with mRNA-coded antigens
US10172935B2 (en) 2011-03-02 2019-01-08 Curevac Ag Vaccination in newborns and infants
US9623095B2 (en) 2011-03-02 2017-04-18 Curevac Ag Vaccination in newborns and infants
US9533047B2 (en) 2011-03-31 2017-01-03 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US8710200B2 (en) 2011-03-31 2014-04-29 Moderna Therapeutics, Inc. Engineered nucleic acids encoding a modified erythropoietin and their expression
US9950068B2 (en) 2011-03-31 2018-04-24 Modernatx, Inc. Delivery and formulation of engineered nucleic acids
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US10022425B2 (en) 2011-09-12 2018-07-17 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US10751386B2 (en) 2011-09-12 2020-08-25 Modernatx, Inc. Engineered nucleic acids and methods of use thereof
US9428535B2 (en) 2011-10-03 2016-08-30 Moderna Therapeutics, Inc. Modified nucleosides, nucleotides, and nucleic acids, and uses thereof
US8664194B2 (en) 2011-12-16 2014-03-04 Moderna Therapeutics, Inc. Method for producing a protein of interest in a primate
US9186372B2 (en) 2011-12-16 2015-11-17 Moderna Therapeutics, Inc. Split dose administration
US8754062B2 (en) 2011-12-16 2014-06-17 Moderna Therapeutics, Inc. DLIN-KC2-DMA lipid nanoparticle delivery of modified polynucleotides
US9295689B2 (en) 2011-12-16 2016-03-29 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US9271996B2 (en) 2011-12-16 2016-03-01 Moderna Therapeutics, Inc. Formulation and delivery of PLGA microspheres
US8680069B2 (en) 2011-12-16 2014-03-25 Moderna Therapeutics, Inc. Modified polynucleotides for the production of G-CSF
US11690910B2 (en) 2012-01-31 2023-07-04 CureVac SE Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
US11110156B2 (en) 2012-02-15 2021-09-07 Curevac Ag 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
US10912826B2 (en) 2012-02-15 2021-02-09 Curevac Ag 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
US9447431B2 (en) 2012-02-15 2016-09-20 Curevac Ag 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
US10682406B2 (en) 2012-02-15 2020-06-16 Curevac Ag 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
US10232024B2 (en) 2012-02-15 2019-03-19 Curevac Ag 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
US9669089B2 (en) 2012-02-15 2017-06-06 Curevac Ag 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
US10799577B2 (en) 2012-02-15 2020-10-13 Curevac Ag 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
US10010592B2 (en) 2012-02-15 2018-07-03 Curevac Ag 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
US10166283B2 (en) 2012-02-15 2019-01-01 Curevac Ag 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
US10111968B2 (en) 2012-02-15 2018-10-30 Curevac Ag 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
US10610605B2 (en) 2012-02-15 2020-04-07 Curevac Ag 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
US10898589B2 (en) 2012-02-15 2021-01-26 Cure Vac AG 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
US10080809B2 (en) 2012-03-27 2018-09-25 Curevac Ag Artificial nucleic acid molecules comprising a 5′TOP UTR
US10738306B2 (en) 2012-03-27 2020-08-11 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
US9683233B2 (en) 2012-03-27 2017-06-20 Curevac Ag Artificial nucleic acid molecules for improved protein or peptide expression
US9890391B2 (en) 2012-03-27 2018-02-13 Curevac Ag RNA vector with an open reading frame, an albumin 3′-UTR, and a histone stem loop
US8999380B2 (en) 2012-04-02 2015-04-07 Moderna Therapeutics, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9114113B2 (en) 2012-04-02 2015-08-25 Moderna Therapeutics, Inc. Modified polynucleotides encoding citeD4
US9233141B2 (en) 2012-04-02 2016-01-12 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9061059B2 (en) 2012-04-02 2015-06-23 Moderna Therapeutics, Inc. Modified polynucleotides for treating protein deficiency
US9050297B2 (en) 2012-04-02 2015-06-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding aryl hydrocarbon receptor nuclear translocator
US9089604B2 (en) 2012-04-02 2015-07-28 Moderna Therapeutics, Inc. Modified polynucleotides for treating galactosylceramidase protein deficiency
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9827332B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of proteins
US9220755B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins associated with blood and lymphatic disorders
US9828416B2 (en) 2012-04-02 2017-11-28 Modernatx, Inc. Modified polynucleotides for the production of secreted proteins
US9255129B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides encoding SIAH E3 ubiquitin protein ligase 1
US9814760B2 (en) 2012-04-02 2017-11-14 Modernatx, Inc. Modified polynucleotides for the production of biologics and proteins associated with human disease
US9220792B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. Modified polynucleotides encoding aquaporin-5
US9254311B2 (en) 2012-04-02 2016-02-09 Moderna Therapeutics, Inc. Modified polynucleotides for the production of proteins
US9782462B2 (en) 2012-04-02 2017-10-10 Modernatx, Inc. Modified polynucleotides for the production of proteins associated with human disease
US9095552B2 (en) 2012-04-02 2015-08-04 Moderna Therapeutics, Inc. Modified polynucleotides encoding copper metabolism (MURR1) domain containing 1
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9587003B2 (en) 2012-04-02 2017-03-07 Modernatx, Inc. Modified polynucleotides for the production of oncology-related proteins and peptides
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
US9303079B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9301993B2 (en) 2012-04-02 2016-04-05 Moderna Therapeutics, Inc. Modified polynucleotides encoding apoptosis inducing factor 1
US9675668B2 (en) 2012-04-02 2017-06-13 Moderna Therapeutics, Inc. Modified polynucleotides encoding hepatitis A virus cellular receptor 2
US9107886B2 (en) 2012-04-02 2015-08-18 Moderna Therapeutics, Inc. Modified polynucleotides encoding basic helix-loop-helix family member E41
US9221891B2 (en) 2012-04-02 2015-12-29 Moderna Therapeutics, Inc. In vivo production of proteins
US9216205B2 (en) 2012-04-02 2015-12-22 Moderna Therapeutics, Inc. Modified polynucleotides encoding granulysin
US10501512B2 (en) 2012-04-02 2019-12-10 Modernatx, Inc. Modified polynucleotides
US9149506B2 (en) 2012-04-02 2015-10-06 Moderna Therapeutics, Inc. Modified polynucleotides encoding septin-4
US9192651B2 (en) 2012-04-02 2015-11-24 Moderna Therapeutics, Inc. Modified polynucleotides for the production of secreted proteins
US9597380B2 (en) 2012-11-26 2017-03-21 Modernatx, Inc. Terminally modified RNA
US9518989B2 (en) 2013-02-12 2016-12-13 Texas Tech University System Composition and method for diagnosis and immunotherapy of lung cancer
RU2718988C2 (ru) * 2013-02-22 2020-04-15 Куревак Аг Комбинация противораковой рнк-вакцины и ингибитора пути pd-1 и ее применение
US11458195B2 (en) 2013-02-22 2022-10-04 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US10434158B2 (en) 2013-02-22 2019-10-08 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US10117920B2 (en) 2013-02-22 2018-11-06 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US9974845B2 (en) 2013-02-22 2018-05-22 Curevac Ag Combination of vaccination and inhibition of the PD-1 pathway
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
US10150797B2 (en) 2013-08-21 2018-12-11 Curevac Ag Respiratory syncytial virus (RSV) vaccine
US11739125B2 (en) 2013-08-21 2023-08-29 Cure Vac SE Respiratory syncytial virus (RSV) vaccine
US10682426B2 (en) 2013-08-21 2020-06-16 Curevac Ag Rabies vaccine
US11266735B2 (en) 2013-08-21 2022-03-08 Curevac Ag Combination vaccine
US10799602B2 (en) 2013-08-21 2020-10-13 Curevac Ag Method for increasing expression of RNA-encoded proteins
US10588959B2 (en) 2013-08-21 2020-03-17 Curevac Ag Combination vaccine
US11965000B2 (en) 2013-08-21 2024-04-23 CureVac SE Respiratory syncytial virus (RSV) vaccine
US9688729B2 (en) 2013-08-21 2017-06-27 Curevac Ag Respiratory syncytial virus (RSV) vaccine
US11034729B2 (en) 2013-08-21 2021-06-15 Curevac Ag Respiratory syncytial virus (RSV) vaccine
US11369694B2 (en) 2013-08-21 2022-06-28 Curevac Ag Rabies vaccine
US20160168227A1 (en) * 2013-08-21 2016-06-16 Curevac Ag Composition and vaccine for treating lung cancer
US10293060B2 (en) 2013-08-21 2019-05-21 Curevac Ag Method for increasing expression of RNA-encoded proteins
US10815291B2 (en) 2013-09-30 2020-10-27 Modernatx, Inc. Polynucleotides encoding immune modulating polypeptides
US10323076B2 (en) 2013-10-03 2019-06-18 Modernatx, Inc. Polynucleotides encoding low density lipoprotein receptor
US10898584B2 (en) 2013-11-01 2021-01-26 Curevac Ag Modified RNA with decreased immunostimulatory properties
US11697816B2 (en) 2013-12-30 2023-07-11 CureVac SE Artificial nucleic acid molecules
US10047375B2 (en) 2013-12-30 2018-08-14 Curevac Ag Artificial nucleic acid molecules
US10648017B2 (en) 2013-12-30 2020-05-12 Curevac Real Estate Gmbh Methods for RNA analysis
US11110157B2 (en) 2014-03-12 2021-09-07 Curevac Ag Combination of vaccination and OX40 agonists
US10307472B2 (en) 2014-03-12 2019-06-04 Curevac Ag Combination of vaccination and OX40 agonists
US11110166B2 (en) 2014-04-01 2021-09-07 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
US10369216B2 (en) 2014-04-01 2019-08-06 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
US10709779B2 (en) 2014-04-23 2020-07-14 Modernatx, Inc. Nucleic acid vaccines
US10022435B2 (en) 2014-04-23 2018-07-17 Modernatx, Inc. Nucleic acid vaccines
US9872900B2 (en) 2014-04-23 2018-01-23 Modernatx, Inc. Nucleic acid vaccines
US10837039B2 (en) 2014-06-10 2020-11-17 Curevac Real Estate Gmbh Methods and means for enhancing RNA production
US11149278B2 (en) 2014-12-12 2021-10-19 Curevac Ag Artificial nucleic acid molecules for improved protein expression
US11761009B2 (en) 2014-12-12 2023-09-19 CureVac SE Artificial nucleic acid molecules for improved protein expression
US11286492B2 (en) 2014-12-12 2022-03-29 Curevac Ag Artificial nucleic acid molecules for improved protein expression
US11345920B2 (en) 2014-12-12 2022-05-31 Curevac Ag Artificial nucleic acid molecules for improved protein expression
US11254951B2 (en) 2014-12-30 2022-02-22 Curevac Ag Artificial nucleic acid molecules
US10653768B2 (en) 2015-04-13 2020-05-19 Curevac Real Estate Gmbh Method for producing RNA compositions
US11744886B2 (en) 2015-04-13 2023-09-05 CureVac Manufacturing GmbH Method for producing RNA compositions
US11446250B2 (en) 2015-04-17 2022-09-20 Curevac Real Estate Gmbh Lyophilization of RNA
US10780054B2 (en) 2015-04-17 2020-09-22 Curevac Real Estate Gmbh Lyophilization of RNA
US11491112B2 (en) 2015-04-17 2022-11-08 CureVac Manufacturing GmbH Lyophilization of RNA
US10293058B2 (en) 2015-04-22 2019-05-21 Curevac Ag RNA containing composition for treatment of tumor diseases
US10869935B2 (en) 2015-04-22 2020-12-22 Curevac Ag RNA containing composition for treatment of tumor diseases
US10918740B2 (en) 2015-04-22 2021-02-16 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
US11384375B2 (en) 2015-04-30 2022-07-12 Curevac Ag Immobilized poly(n)polymerase
US11661634B2 (en) 2015-05-08 2023-05-30 CureVac Manufacturing GmbH Method for producing RNA
US11559570B2 (en) 2015-05-15 2023-01-24 CureVac SE Prime-boost regimens involving administration of at least one mRNA construct
US10729654B2 (en) 2015-05-20 2020-08-04 Curevac Ag Dry powder composition comprising long-chain RNA
US10517827B2 (en) 2015-05-20 2019-12-31 Curevac Ag Dry powder composition comprising long-chain RNA
US11179337B2 (en) 2015-05-20 2021-11-23 Curevac Ag Dry powder composition comprising long-chain RNA
US11534405B2 (en) 2015-05-20 2022-12-27 Curevac Ag Dry powder composition comprising long-chain RNA
US11433027B2 (en) 2015-05-20 2022-09-06 Curevac Ag Dry powder composition comprising long-chain RNA
US10760070B2 (en) 2015-05-29 2020-09-01 Curevac Real Estate Gmbh Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
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
US11608513B2 (en) 2015-05-29 2023-03-21 CureVac SE Method for adding cap structures to RNA using immobilized enzymes
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
US11274293B2 (en) 2015-05-29 2022-03-15 Curevac Real Estate Gmbh Method for producing and purifying RNA, comprising at least one step of tangential flow filtration
US10501768B2 (en) 2015-07-13 2019-12-10 Curevac Ag Method of producing RNA from circular DNA and corresponding template DNA
US10449244B2 (en) 2015-07-21 2019-10-22 Modernatx, Inc. Zika RNA vaccines
US10702597B2 (en) 2015-07-21 2020-07-07 Modernatx, Inc. CHIKV RNA vaccines
US11007260B2 (en) 2015-07-21 2021-05-18 Modernatx, Inc. Infectious disease vaccines
US11364292B2 (en) 2015-07-21 2022-06-21 Modernatx, Inc. CHIKV RNA 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
US10543269B2 (en) 2015-10-22 2020-01-28 Modernatx, Inc. hMPV RNA vaccines
US10933127B2 (en) 2015-10-22 2021-03-02 Modernatx, Inc. Betacoronavirus mRNA vaccine
US10716846B2 (en) 2015-10-22 2020-07-21 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10702600B1 (en) 2015-10-22 2020-07-07 Modernatx, Inc. Betacoronavirus mRNA vaccine
US11278611B2 (en) 2015-10-22 2022-03-22 Modernatx, Inc. Zika virus RNA vaccines
US11235052B2 (en) 2015-10-22 2022-02-01 Modernatx, Inc. Chikungunya virus RNA vaccines
US10517940B2 (en) 2015-10-22 2019-12-31 Modernatx, Inc. Zika virus RNA vaccines
US10124055B2 (en) 2015-10-22 2018-11-13 Modernatx, Inc. Zika virus RNA vaccines
US11872278B2 (en) 2015-10-22 2024-01-16 Modernatx, Inc. Combination HMPV/RSV RNA vaccines
US10702599B2 (en) 2015-10-22 2020-07-07 Modernatx, Inc. HPIV3 RNA vaccines
US10064935B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10238731B2 (en) 2015-10-22 2019-03-26 Modernatx, Inc. Chikagunya virus RNA vaccines
US10272150B2 (en) 2015-10-22 2019-04-30 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10383937B2 (en) 2015-10-22 2019-08-20 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US10064934B2 (en) 2015-10-22 2018-09-04 Modernatx, Inc. Combination PIV3/hMPV RNA vaccines
US10675342B2 (en) 2015-10-22 2020-06-09 Modernatx, Inc. Chikungunya virus RNA vaccines
US11484590B2 (en) 2015-10-22 2022-11-01 Modernatx, Inc. Human cytomegalovirus RNA vaccines
US11413346B2 (en) 2015-11-09 2022-08-16 Curevac Ag Rotavirus vaccines
US11786590B2 (en) 2015-11-09 2023-10-17 CureVac SE Rotavirus vaccines
US11684665B2 (en) 2015-12-22 2023-06-27 CureVac SE Method for producing RNA molecule compositions
US11248223B2 (en) 2015-12-23 2022-02-15 Curevac Ag Method of RNA in vitro transcription using a buffer containing a dicarboxylic acid or tricarboxylic acid or a salt thereof
US11723967B2 (en) 2016-02-17 2023-08-15 CureVac SE Zika virus vaccine
US11920174B2 (en) 2016-03-03 2024-03-05 CureVac SE RNA analysis by total hydrolysis and quantification of released nucleosides
US11596699B2 (en) 2016-04-29 2023-03-07 CureVac SE RNA encoding an antibody
US11078247B2 (en) 2016-05-04 2021-08-03 Curevac Ag RNA encoding a therapeutic protein
US11141474B2 (en) 2016-05-04 2021-10-12 Curevac Ag Artificial nucleic acid molecules encoding a norovirus antigen and uses thereof
US11478552B2 (en) 2016-06-09 2022-10-25 Curevac Ag Hybrid carriers for nucleic acid cargo
US10653712B2 (en) 2016-09-14 2020-05-19 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US11202793B2 (en) 2016-09-14 2021-12-21 Modernatx, Inc. High purity RNA compositions and methods for preparation thereof
US11541113B2 (en) 2016-10-21 2023-01-03 Modernatx, Inc. Human cytomegalovirus vaccine
US11197927B2 (en) 2016-10-21 2021-12-14 Modernatx, Inc. Human cytomegalovirus vaccine
US10695419B2 (en) 2016-10-21 2020-06-30 Modernatx, Inc. Human cytomegalovirus vaccine
US11279923B2 (en) 2016-11-28 2022-03-22 Curevac Ag Method for purifying RNA
US11464836B2 (en) 2016-12-08 2022-10-11 Curevac Ag RNA for treatment or prophylaxis of a liver disease
US11103578B2 (en) 2016-12-08 2021-08-31 Modernatx, Inc. Respiratory virus nucleic acid vaccines
US11542490B2 (en) 2016-12-08 2023-01-03 CureVac SE RNAs for wound healing
US11464847B2 (en) 2016-12-23 2022-10-11 Curevac Ag Lassa virus vaccine
US11141476B2 (en) 2016-12-23 2021-10-12 Curevac Ag MERS coronavirus vaccine
US11865084B2 (en) 2016-12-23 2024-01-09 CureVac SE MERS coronavirus vaccine
US11524066B2 (en) 2016-12-23 2022-12-13 CureVac SE Henipavirus vaccine
US10273269B2 (en) 2017-02-16 2019-04-30 Modernatx, Inc. High potency immunogenic zika virus compositions
US11739335B2 (en) 2017-03-24 2023-08-29 CureVac SE 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
US11820728B2 (en) 2017-04-28 2023-11-21 Acuitas Therapeutics, Inc. Carbonyl lipids and lipid nanoparticle formulations for delivery of nucleic acids
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
US11207398B2 (en) 2017-09-14 2021-12-28 Modernatx, Inc. Zika virus mRNA vaccines
US10653767B2 (en) 2017-09-14 2020-05-19 Modernatx, Inc. Zika virus MRNA vaccines
US11692002B2 (en) 2017-11-08 2023-07-04 CureVac SE RNA sequence adaptation
US11931406B2 (en) 2017-12-13 2024-03-19 CureVac SE Flavivirus vaccine
US11525158B2 (en) 2017-12-21 2022-12-13 CureVac SE Linear double stranded DNA coupled to a single support or a tag and methods for producing said linear double stranded DNA
US11351242B1 (en) 2019-02-12 2022-06-07 Modernatx, Inc. HMPV/hPIV3 mRNA vaccine composition
CN112402596A (zh) * 2019-08-22 2021-02-26 上海细胞治疗集团有限公司 多肽组合物及疫苗
US11964011B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11576966B2 (en) 2020-02-04 2023-02-14 CureVac SE Coronavirus vaccine
US11596686B2 (en) 2020-02-04 2023-03-07 CureVac SE Coronavirus vaccine
US11964012B2 (en) 2020-02-04 2024-04-23 CureVac SE Coronavirus vaccine
US11241493B2 (en) 2020-02-04 2022-02-08 Curevac Ag Coronavirus vaccine
US11471525B2 (en) 2020-02-04 2022-10-18 Curevac Ag Coronavirus vaccine
US11976019B2 (en) 2020-07-16 2024-05-07 Acuitas Therapeutics, Inc. Cationic lipids for use in lipid nanoparticles
US11406703B2 (en) 2020-08-25 2022-08-09 Modernatx, Inc. Human cytomegalovirus vaccine
US11918643B2 (en) 2020-12-22 2024-03-05 CureVac SE RNA vaccine against SARS-CoV-2 variants
US11872280B2 (en) 2020-12-22 2024-01-16 CureVac SE RNA vaccine against SARS-CoV-2 variants
WO2024097894A1 (en) * 2022-11-04 2024-05-10 RNAimmune, Inc. Compositions and methods of ribonucleic acid vaccines encoding nye-so-1

Also Published As

Publication number Publication date
MX2010003766A (es) 2010-04-30
WO2009046738A1 (en) 2009-04-16
HRP20180698T1 (hr) 2018-06-29
US20180256694A1 (en) 2018-09-13
US20130202645A1 (en) 2013-08-08
BRPI0818318B8 (pt) 2021-05-25
PL2197481T3 (pl) 2018-10-31
RU2526510C2 (ru) 2014-08-20
WO2009046974A2 (en) 2009-04-16
EP3375454B1 (en) 2021-04-14
CN101820899B (zh) 2014-03-12
EP2197481A2 (en) 2010-06-23
HUE037295T2 (hu) 2018-08-28
JP2015110613A (ja) 2015-06-18
DK2197481T3 (en) 2018-05-22
BRPI0818318B1 (pt) 2020-10-13
LT2197481T (lt) 2018-06-25
JP6031131B2 (ja) 2016-11-24
JP6012423B2 (ja) 2016-10-25
AU2008309930A1 (en) 2009-04-16
ES2668926T3 (es) 2018-05-23
JP2013067633A (ja) 2013-04-18
US20220096616A1 (en) 2022-03-31
AU2008309930B2 (en) 2013-02-21
US20160317636A1 (en) 2016-11-03
CA2696768A1 (en) 2009-04-16
US20160346370A1 (en) 2016-12-01
US9352028B2 (en) 2016-05-31
EP3375454A1 (en) 2018-09-19
CA2696768C (en) 2019-03-26
RU2010118024A (ru) 2011-11-20
CN101820899A (zh) 2010-09-01
CY1120168T1 (el) 2018-12-12
JP5139529B2 (ja) 2013-02-06
PT2197481T (pt) 2018-05-10
JP2010540672A (ja) 2010-12-24
SI2197481T1 (en) 2018-07-31
WO2009046974A3 (en) 2009-06-18
EP2197481B1 (en) 2018-02-14
NO2197481T3 (pt) 2018-07-14

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
AS Assignment

Owner name: CUREVAC GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BARNER, MARIJKE;PROBST, JOCHEN;LANDER, THOMAS;AND OTHERS;SIGNING DATES FROM 20100608 TO 20100614;REEL/FRAME:024574/0597

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: CUREVAC AG, GERMANY

Free format text: CHANGE OF NAME;ASSIGNOR:CUREVAC GMBH;REEL/FRAME:037115/0430

Effective date: 20150917

AS Assignment

Owner name: CUREVAC SE, GERMANY

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

Effective date: 20220926