WO2023001895A1 - Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma - Google Patents

Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma Download PDF

Info

Publication number
WO2023001895A1
WO2023001895A1 PCT/EP2022/070373 EP2022070373W WO2023001895A1 WO 2023001895 A1 WO2023001895 A1 WO 2023001895A1 EP 2022070373 W EP2022070373 W EP 2022070373W WO 2023001895 A1 WO2023001895 A1 WO 2023001895A1
Authority
WO
WIPO (PCT)
Prior art keywords
cell
years
treatment
patients
recombinant
Prior art date
Application number
PCT/EP2022/070373
Other languages
French (fr)
Inventor
Leander Grode
Original Assignee
Vakzine Projekt Management Gmbh
Serum Institute Of India Ltd.
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
Priority claimed from EP21187253.6A external-priority patent/EP4122491A1/en
Priority claimed from US17/667,784 external-priority patent/US20220160858A1/en
Application filed by Vakzine Projekt Management Gmbh, Serum Institute Of India Ltd. filed Critical Vakzine Projekt Management Gmbh
Priority to IL310225A priority Critical patent/IL310225A/en
Priority to AU2022313491A priority patent/AU2022313491A1/en
Priority to EP22751731.5A priority patent/EP4373519A1/en
Priority to CA3224671A priority patent/CA3224671A1/en
Publication of WO2023001895A1 publication Critical patent/WO2023001895A1/en

Links

Classifications

    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/04Mycobacterium, e.g. Mycobacterium tuberculosis
    • 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
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/522Bacterial cells; Fungal cells; Protozoal cells avirulent or attenuated
    • 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
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • A61K2039/585Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation wherein the target is cancer
    • 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

Definitions

  • the invention relates to a recombinant Mycobacterium cell for use as an immunotherapeutic agent in the therapy of bladder carcinoma, particularly in the second-line therapy of non-muscle-invasive bladder carcinoma.
  • Urothelial bladder carcinoma is the 5th most common cancer. In the United States, about 75.000 new cases are diagnosed each year 4.5% of all new cancers, and approximately 15.600 deaths are expected. In Germany, about 16.000 new cases are diagnosed each year. Because a recurrence of disease is likely in bladder carcinoma, patients must undergo surveillance for an extended period.
  • bladder carcinomas begin in transitional epithelial cells that make up the inner lining of the bladder. As these tumors grow, they can invade the surrounding connective tissue and muscle. In advanced disease, tumors spread beyond the bladder to nearby lymph nodes or pelvic organs or metastasize to more distant organs such as lung, liver, and bone.
  • the overall 5-year survival rate for bladder carcinoma is 77%, and this rate has not changed significantly over the last 10 years.
  • the 5-year relative survival rates for patients with tumors restricted to the inner layer of the bladder are 96% and 69%, respectively.
  • the rates drop to 34% for those with disease that has spread locally beyond the bladder and to 6% with distant metastases.
  • Recurrence and progression to muscle-invasive disease may lead to additional chemotherapeutical, surgical, and radio-oncological interventions including cisplatin-based chemotherapy, transurethral resection of the bladder (TURB), cystectomy and chemo-radiotherapy.
  • Recurrent bladder carcinoma may be treated with combination therapy regimens, including gemcitabine plus cisplatin or methotrexate, vinblastine, doxorubicin plus cisplatin.
  • An alternative option for recurrent patients is a re-treatment with conventional BCG therapy (Yates et al, Eur Urol 62 (2012), 1088-1096).
  • an object of the present invention to provide an improved treatment for patients failing to respond to first-line conventional BCG therapy as these patients are at high risk of cancer progression.
  • An improved treatment of these high-risk patients will increase bladder preservation rates and consequently improve quality of life and decrease health costs.
  • a recombinant BCG strain expressing a phagolysosomal escape domain is described in WO 99/101496, the content of which is herein incorporated by reference.
  • the phagolysosomal escape domain enables the strain to escape from the phagosome of infected host cells by perforating the membrane of the phagosome.
  • a urease-deficient recombinant strain was developed. This strain is disclosed in WO 2004/094469, the content of which is herein incorporated.
  • WO 2012/085101 discloses that a recombinant BCG strain expressing membrane-perforating listeriolysin (Hly) of Listeria monocytogenes and devoid of urease C induces superior protection against aerogenic challenge with Mycobacterium tuberculosis ( Mtb ) as compared to parental BCG in a preclinical model. Further, it is shown that both the recombinant and the parenteral strain induce marked Th1 immune responses, whilst only the recombinant BCG strain elicits are profound Th17 response in addition.
  • listeriolysin Hly
  • Mtb Mycobacterium tuberculosis
  • WO 2016/177717 discloses that a recombinant urease-deficient and listeriolysin-expressing recombinant BCG strain induces a superior immune response compared to the parenteral BCG in an animal model. Further, the start of a human clinical phase I/ll trial using recombinant BCG as an immunotherapeutic agent in patients after a first standard BCG therapy is reported.
  • a first aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject, e.g., a human subject suffering from bladder carcinoma, particularly from non-muscle- invasive bladder carcinoma as a second-line therapy.
  • a subject e.g., a human subject suffering from bladder carcinoma, particularly from non-muscle- invasive bladder carcinoma as a second-line therapy.
  • the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject as a second-line therapy a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain for use in the immunotherapeutic treatment of bladder carcinoma wherein the recombinant Mycobacterium cell is administered to a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma as a second-line therapy.
  • the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject as a second-line therapy a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a further aspect of the present invention is a recombinant Mycobacterium cell which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain for use as an immunotherapeutic agent in the treatment of bladder carcinoma, in particular recurrent bladder carcinoma, wherein the subject to be treated has relapsed and/or has progressed after a first treatment of bladder carcinoma, particularly after a first treatment with conventional BCG therapy.
  • the present invention further relates to a method for the immunotherapy of bladder carcinoma in a subject in need thereof, particularly of non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma, wherein the subject has relapsed and/or has progressed after a first treatment of bladder carcinoma, particularly after a first treatment with conventional BCG therapy, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma, wherein disease recurrence is inhibited for a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years.
  • the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a still further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma wherein overall survival is increased in a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years, or of at least 4 years.
  • the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain wherein overall survival is increased in a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years, or of at least 4 years.
  • a still further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • a phagolysosomal escape domain for use in a method for improving the life quality of a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent thereby reducing or avoiding cystectomy in said subject.
  • the present invention further relates to a method for improving the life quality of a subject suffering from bladder carcinoma, particularly from non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
  • the present invention relates to a second-line therapy with rBCG in combination with at least one of:
  • the present invention relates to an improvement of life quality comprising reducing or the need of cystectomy in combination with at least one of:
  • the present invention relates to a second-line therapy in combination with:
  • second-line immunotherapy refers to the treatment of a subject, particularly a human subject, after failure of a first-line therapy.
  • first-line therapy particularly means disease recurrence and/or disease progression including progression of the primary tumor and/or formation of metastases after first-line therapy. More particularly, the term “failure” means disease recurrence.
  • first-line therapy particularly refers to a first-line therapy including an immunotherapy, and more particularly to a first-line immunotherapy comprising administration of standard BCG without a phagolysosomal escape domain as described herein, such as previously approved standard BCG as described in Rentsch, C A, Eur Urol 2014, PMID: 24674149.
  • second-line immunotherapy comprises administration of a recombinant Mycobacterium cell as described herein after a single “first-line therapy” cycle or after multiple, e.g., 2, 3 or more “first-line therapy” cycles, particularly a single cycle or multiple cycles comprising administration of BCG without a phagolysosomal escape domain.
  • the recombinant Mycobacterium cell is administered as a second-line treatment.
  • the individual to be treated may have received a first treatment, in particular of bladder carcinoma treatment, selected from the group of standard BCG treatment, chemotherapy including systemic and/or intravesical chemotherapy, bladder surgery, radiation and any combination thereof.
  • the first-line treatment may include cisplatin-based chemotherapy, in particular cisplatin-based chemotherapy followed by bladder surgery and/or radiation therapy, concomitant chemotherapy and standard BCG.
  • the individual to be treated has received standard BCG treatment as a first treatment of bladder carcinoma and/or underwent cystectomy or another local treatment or systemic chemotherapy.
  • the subject to be treated according to the present invention is suffering from bladder carcinoma, particularly non-muscle invasive bladder carcinoma (NMIBC) including carcinoma in situ (CIS).
  • NMIBC non-muscle invasive bladder carcinoma
  • CIS carcinoma in situ
  • the subject is suffering from recurrent NMBIC.
  • the subject has a recurrent high-grade NMIBC for progression (e.g., score 7-23) based on the European Organization for Research and Treatment.
  • the subject has undergone to a transurethral resection of the bladder tumor (TURBT) before therapy start.
  • the subject has a tumor-free bladder state at therapy start, e.g., as confirmed by transurethral resection of the bladder (TURB) and biopsy.
  • the subject is a patient with a high grade NMIBC tumor present at 6 months or both at 3 months and 6 months after start of a previous cycle of standard BCG therapy and worsening of the disease under standard BCG therapy including at least one of higher number of recurrences, higher tumor category or higher grade, and appearances of CIS, including low grade tumors, optionally in spite of an initial response.
  • the subject is a smoker.
  • a recombinant Mycobacterium cell as described herein as a second-line therapy is highly effective in reducing disease occurrence for an extended time period, e.g., a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years or of at least 4 years after treatment start. 3 years after treatment start, the median time of disease recurrence is already 1.3 years and will probably increase further. At this time, no tumor recurrence was observed in about 49% of the patients.
  • the therapy of the present invention provides a disease-free time period of at least 1 year, particularly of at least 2 years, of at least 3 years, or of at least 4 years for at least 30% of the patients and particularly for at least 45% of the patients.
  • the treatment is highly efficient in stopping disease recurrence in a sub-group of patients, who did not suffer from disease recurrence in a time period up to 2 years after treatment start.
  • administration of a recombinant Mycobacterium cell as described herein is highly effective in increasing the overall survival in an extended time period, e.g., a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years or of at least 4 years. 3 years after start of the treatment, the overall survival rate was between 70% and 80%.
  • the therapy of the invention is suitable in avoiding cystectomy in a large number of patients resulting in a high improvement of life quality.
  • cystectomy may be avoided for a time period of at least a 1 year, particularly for a time period of at least 2 years, of at least 3 years or of at least 4 years in at least 30% of the patients and particularly for at least 45% of the patients.
  • the immunotherapeutic agent is a live recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising (a) a domain capable of eliciting an immune response and (b) a phagolysosomal escape domain.
  • the domain capable of eliciting an immune response is preferably an immunogenic peptide or polypeptide from a pathogen or an immunogenic fragment thereof.
  • the Mycobacterium cell is preferably an M. bovis cell, an M. tuberculosis cell, particularly an attenuated M. tuberculosis cell or other Mycobacteria, e.g. M. microti, M. smegmatis, M. canettii, M. marinum or M. fortuitum. More preferably, the cell is an attenuated recombinant M. bovis (BCG) cell, particularly an M. bovis BCG cell, more particularly a recombinant M. bovis BCG cell from strain Danish subtype Prague (Brosch et al., Proc. Natl. Acad. Sci. USA, 104 (2007), 5396-5601).
  • BCG attenuated recombinant M. bovis
  • the Mycobacterium cell is recombinant urease-deficient.
  • the ureC sequence of the Mycobacterium cell is inactivated (AUrec), e.g., by constructing a suicide vector containing a ureC gene disrupted by a selection marker gene, e.g., the hygromycin gene, transforming the target cell with the vector and screening for selection marker-positive cells having a urease negative phenotype.
  • the selection marker gene i.e. , the hygromycin gene, is subsequently inactivated.
  • the cell is a selection marker-free recombinant Mycobacterium cell.
  • the cell is selection marker-free recombinant BCG strain Danish subtype Prague characterized as recombinant BCG AUrec::Hly+.
  • the domain capable of eliciting an immune response is preferably selected from immunogenic peptides or polypeptides from M. bovis, M. tuberculosis or M. leprae or from immunogenic fragments thereof having a length of at least 6, preferably at least 8 amino acids, more preferably at least 9 amino acids and e.g., up to 20 amino acids.
  • suitable antigens are Ag85B (p30) from M. tuberculosis, Ag85B (a-antigen) from M. bovis BCG, Ag85A from M.
  • the domain capable of eliciting an immune response is selected from non-Mycobacterium polypeptides. More preferably, the immunogenic domain is derived from the antigen Ag85B. Most preferably, the immunogenic domain comprises the sequence from aa. 41 to aa.51 in SEQ ID No.2.
  • the recombinant nucleic acid molecule further comprises a phagolysosomal escape domain, i.e., a polypeptide domain which provides for an escape of the fusion polypeptide from the phagolysosome into the cytosol of mammalian cells.
  • a phagolysosomal escape domain i.e., a polypeptide domain which provides for an escape of the fusion polypeptide from the phagolysosome into the cytosol of mammalian cells.
  • the phagolysosomal escape domain is a Listeria phagolysosomal escape domain, which is described in US 5,733,151, herein incorporated by reference. More preferably, the phagolysosomal escape domain is derived from the listeriolysin gene (Hly) of L. monocytogenes.
  • the phagolysosomal domain is encoded by a nucleic acid molecule selected from: (a) a nucleotide sequence comprising nucleotides 211 - 1722 as shown in SEQ ID No.1, (b) a nucleotide sequence which encodes the same amino acid sequence as the sequence from (a), and (c) a nucleotide sequence hybridizing under stringent conditions with the sequence from (a) or (b).
  • a nucleic acid molecule selected from: (a) a nucleotide sequence comprising nucleotides 211 - 1722 as shown in SEQ ID No.1, (b) a nucleotide sequence which encodes the same amino acid sequence as the sequence from (a), and (c) a nucleotide sequence hybridizing under stringent conditions with the sequence from (a) or (b).
  • the present invention also comprises nucleic acid sequences hybridizing therewith.
  • hybridization is used as defined in Sambrook et al. (Molecular Cloning. A laboratory manual, Cold Spring Harbor Laboratory Press (1989), 1.101 -1.104).
  • hybridization is used if a positive hybridization signal can still be observed after washing for one hour with 1 X SSC and 0.1 % SDS at 55°C, preferably at 62° C and more preferably at 68°C, particularly for 1 hour in 0.2 X SSC and 0.1 % SDS at 55°C, preferably at 62°C and more preferably at 68°C.
  • a sequence hybridizing with a nucleotide sequence as per SEQ ID No.1 under such washing conditions is a phagolysosomal escape domain encoding nucleotide sequence preferred by the subject invention.
  • a nucleotide sequence encoding a phagolysosomal escape domain as described above may be directly obtained from a Listeria organism or from any recombinant source e.g., a recombinant E.coli cell containing the corresponding Listeria nucleic acid molecule, or a variant thereof as described above.
  • the recombinant nucleic acid molecule encoding for a fusion polypeptide contains a signal peptide encoding sequence.
  • the signal sequence is a signal sequence active in Mycobacteria, preferably in M. bovis, e.g., a native M. bovis signal sequence.
  • a preferred example of a suitable signal sequence is the nucleotide sequence coding for the Ag85B signal peptide, which is depicted in SEQ ID No.1 from nucleotide 1 to 120.
  • a peptide linker be provided between the immunogenic domain and the phagolysosomal escape domain.
  • said peptide linker has a length of from 5 to 50 amino acids. More preferably, a sequence encoding a linker as shown in SEQ ID No.1 from nucleotide 154 to 210 or a sequence corresponding thereto as regards the degeneration of the genetic code.
  • the nucleic acid may be located on a recombinant vector.
  • the recombinant vector is a prokaryotic vector, i.e. , a vector containing elements for replication or/and genomic integration in prokaryotic cells.
  • the recombinant vector carries the nucleic acid molecule of the present invention operatively linked with an expression control sequence.
  • the expression control sequence is preferably an expression control sequence active in Mycobacteria, particularly in M. bovis.
  • the vector can be an extrachromosomal vector or a vector suitable for integration into the chromosome. Examples of such vectors are known to the man skilled in the art and, for instance, given in Sambrook et at. supra.
  • the immunotherapeutic agent of the present invention is suitable for the treatment of bladder carcinoma, e.g., non-invasive bladder carcinoma, e.g. non-invasive papillary carcinoma in situ (T a ), non-invasive carcinoma in situ (Tcis), tumor invading subepithelial connective tissue (Ti), tumor invading superficial muscle (inner half) ⁇ h a ), tumor invading deep muscle (outer half) (T 2b ), tumor invading perivesical tissue (T3 including T3a and T3 b ), tumor invading prostate, uterus or vagina (T4a), and tumor invading pelvic wall or abdominal wall (T4 b ).
  • non-invasive bladder carcinoma e.g. non-invasive papillary carcinoma in situ (T a ), non-invasive carcinoma in situ (Tcis), tumor invading subepithelial connective tissue (Ti), tumor invading superficial muscle (inner half) ⁇ h a ), tumor invading
  • the tumor is a superficial tumor or carcinoma in situ (Tcis), non-invasive papillary carcinoma (T a ), or a tumor invading subepithelial connective tissue (Ti).
  • the immunotherapeutic treatment is suitable in the treatment of primary bladder carcinoma and/or in the treatment of recurring bladder carcinoma.
  • the immunotherapeutic agent is locally administered to the tumor site, i.e., to the site of a primary tumor before surgery or after surgery and optionally after chemotherapy.
  • the agent is preferably administered by vesicular instillation into the urinary bladder.
  • the immunotherapeutic agent is administered to the subject to be treated in an effective dose.
  • the dose for an administration may be about 10 6 to 10 10 viable units (CFU), e.g., about 10 7 to 5 x 10 9 or 10 8 to 3 x 10 9 viable units.
  • the dose for an administration is about 2 x 10 9 viable units (CFU).
  • the immunotherapeutic agent is administered several times, e.g., at least 3 times or at least 5 times up to 30 times, particularly about 15 times, at predetermined times during the treatment.
  • the immunotherapeutic agent is usually provided as a pharmaceutical preparation, which comprises the recombinant Mycobacterial cell in solid form, e.g., a lyophilized or cryoconserved preparation, which is reconstituted with a suitable liquid carrier before use.
  • a pharmaceutical preparation which comprises the recombinant Mycobacterial cell in solid form, e.g., a lyophilized or cryoconserved preparation, which is reconstituted with a suitable liquid carrier before use.
  • the preparation may be provided in liquid form, e.g., as suspension.
  • the immunotherapeutic agent of the invention is administered for the treatment of carcinoma in situ.
  • a standard schedule may comprise weekly administration of the agent for at least 4, e.g., 4, 5, 6, 7 or 8 weeks as an induction therapy.
  • the induction therapy should not start until 2- 3 weeks after primary tumor surgery.
  • administration may continue using maintenance therapy for at least 6 months or at least 1 year.
  • the immunotherapeutic agent is administered in an induction therapy in the prophylactic treatment of tumor recurrence.
  • therapy may start about 2-3 weeks after biopsy of the tumor site and be repeated, e.g., at weekly intervals for at least 4, e.g., 4, 5, 6, 7 or 8 weeks. In intermediate and high-risk tumors this may be followed by maintenance therapy.
  • Maintenance therapy may comprise long-term therapy, e.g., 6, 9 or 12 months therapy or even longer with treatments at monthly intervals.
  • maintenance therapy may comprise 2, 3 or 4 administrations at weekly intervals, at month 3, 6, 12, 18, 24, 30 and 36.
  • the immunotherapeutic agent is used for the treatment of non-muscle invasive bladder cancer in patients with recurrence after standard BCG therapy.
  • the immunotherapeutic agent is administered into the bladder according to a schedule involving weekly instillations during an induction phase with e.g., 6 weekly instillations, a first maintenance phase after about 3 months with e.g., 3 weekly instillations, a second maintenance phase after about 6 months with e.g., 3 instillations and a third maintenance phase after about 12 months with e.g., 3 instillations.
  • the administration as immunotherapeutic agent of the recombinant Mycobacterium cell as described above may be combined with further anti tumor therapy, e.g., radiation and/or chemotherapy. Further, the immunotherapy as described above may be combined with a non-tumor site specific administration of the recombinant Mycobacterium cell in order to provide a general stimulation of the immune system. This non-site specific administration may be effected as described in WO 2012/085101 , e.g., before surgery of the primary tumor.
  • the agent is preferably administered to a human subject in a dose of about 1-10 x 10 5 , preferably about 2-8 x 10 5 cells.
  • the agent is preferably administered as a single dose, e.g., by injection. Subcutaneous injection is preferred. Further, it is preferred to administer the agent without adjuvant.
  • rBCG recombinant M. bovis (BCG) Danish subtype Prague with an inactivated ureC sequence (AUrec) and without functional selection marker gene which expresses an Ag85B/Flly fusion protein as shown in SEQ ID No.2 (Hly+).
  • BCG recombinant M. bovis
  • AUrec inactivated ureC sequence
  • Example 1 Phase I/ll open label clinical trial assessing safety and efficacy of intravesical instillation of recombinant BCG (rBCG) in human patients with recurrent non-muscle invasive bladder cancer after standard BCG therapy
  • Phase I Phase I was conducted to determine safety, tolerability, and the recommended phase II dose of intravesical rBCG instillations in patients with recurrence of non-muscle-invasive bladder cancer after TURB and standard BCG therapy.
  • Phase II was conducted to investigate the efficacy, safety, tolerability and immunogenicity of intravesical rBCG instillations in patients with recurrence of non-muscle-invasive bladder cancer after TURB in standard BCG therapy.
  • Clinical protocol rBCG was administered into bladder in 15 weekly instillations (induction phase: instillations 1-6; maintenance 3 months: instillations 7-9; maintenance 6 months: instillations 10-12, maintenance 12 months: instillations 13-15).
  • the primary endpoint of the phase I was dose limiting toxicity (DLT) of intravesical rBCG instillations in patients with recurrence after standard BCG therapy in non-muscle invasive bladder cancer (NMIBC).
  • DLT dose limiting toxicity
  • NMIBC non-muscle invasive bladder cancer
  • the DLT period corresponds to 3 instillations plus 1 week and covers acute toxicities induced by treatment.
  • Patients were treated in two cohorts of three, following the rules of a 3 + 3 design (dose de-escalation rules: if patients treated at dose level 1 show signs of DLT, dose of instilled rBCG will be reduced to level -1 , which is 10 times lower than level 1 ).
  • the dose levels were as follows:
  • progression score 7-2 - Recurrent high-grade NMIBC for progression (progression score 7-23) based on the European Organization for Research and Treatment of Cancer scoring system, failing BCG therapy (Babjuk M, Eur Urol 2008 PMID 18468779), for whom radical cystectomy or re-induction with standard BCG is indicated.
  • UC urothelial carcinoma
  • Phase II was conducted with dose level 1: 1- 19.2 x 10 8 CFU of rBCG.
  • the active agent was administered in 50 ml drug solution containing dextran, glucose, 0.9% sodium chloride, 0.025% Tween 80 and water for injection.
  • the treatment was safe and well tolerated, with only 5% of patients unable to tolerate adequate induction therapy. No dose-limiting toxicity occurred and no grade 3 or 4 adverse events were observed.
  • the trial was designed as a multicentre, open-label, single-arm, phase I/ll study and conducted in compliance with the current version of the declaration of Helsinki, the ICH-GCP, and with national legal and regulatory requirements. Written informed consent was obtained from all patients prior to enrolment.
  • Prior therapy was defined as one previous cycle of intravesical BCG (induction phase >5 instillations ⁇ BCG maintenance).
  • bladder wash cytology had to be negative, except for patients with pure or concomitant CIS and imaging without evidence of metastatic disease.
  • Exclusion criteria were stage > T2 urothelial carcinoma of the bladder, concomitant urothelial carcinoma of the upper urinary tract, the non-prostatic urethra, or evidence of metastatic disease
  • rBCG was provided as a formulated lyophilized powder of 1-19.2 c 10 8 CFU/vial of live Mycobacterium bovis BCG ureCwhly (SIIPL, Serum Institute of India Private Limited, Pune, India) and reconstituted in 50 ml of 0.9% saline water for intravesical application. Patients were scheduled for standard treatment of 6 weekly intravesical instillations followed by maintenance of 1 year (3 instillations at 3, 6 and 12 months after first instillation).
  • the primary endpoint of the phase II part was defined as the recurrence-free rate (RFR) in the bladder 60 weeks after registration.
  • RFR recurrence-free rate
  • a recurrence in the bladder associated with cancer in the prostatic urethra or evidence for cancer in the upper urinary tract were not considered as a recurrence in the bladder.
  • Predefined secondary endpoints included time from trial registration to recurrence in the bladder, time to progression, overall survival (OS), AEs, tolerability, and QoL.
  • Progression was defined as progression to muscle-invasive bladder cancer or progression to metastatic disease.
  • OS was calculated from registration until death from any cause.
  • Tolerability was defined as the proportion of patients finishing five instillations of induction within 12 weeks after treatment start. 2.1.2 Statistical analyses
  • a history of chemical exposure as a risk factor for bladder cancer was identified in 3 patients, while 21 patients (52.5%) had a smoking history with a median (range) of 38 (4.0-99.0) pack-years. Concomitant or pure CIS was present in 27 (67.5%) patients. Fourteen patients (35%) received BCG maintenance therapy. The median progression score was 16 (7-20) for those patients with and 16 (7-19) for those without previous BCG maintenance.
  • progression-free rate and OS rate were 76.3% [95% Cl 56.4, 88.0] and 92.9% [95% Cl 74.3, 98.2], respectively.
  • Treatment-related AEs are listed in Table 6.
  • the major grade 2 AE term was genitourinary tract infection with common uropathogenic bacteria in one third of the patients. Two patients had to undergo in-hospital antibiotic treatment for urogenital tract infection.
  • GU infection The most common AE was genitourinary (GU) infection with common bacteria, occurring in one third of the patients. Potential explanations are i) that lubricants containing antiseptics for instillations were not allowed, and ii) that asymptomatic patients with positive urine dipstick had to undergo antibiotic treatment when urine culture was positive. Importantly, the GU infections did not significantly impact on QoL of the patients (not shown). No grade 4 or 5 AE occurred, and tolerability (defined as patients receiving more than 4 instillations during induction) was 95.2%. The treatment can therefore be considered as safe and well tolerated.
  • rBCG has the potential to decrease the proportion of patients becoming intolerable to BCG treatment and may be combined with other agents such as check-point inhibitors in order to increase efficiency.
  • rBCG has a promising tolerability, safety, and QoL profile.
  • BCG Bacillus Calmette Guerin. Some patients received more than one treatment. One of the 13 patients undergoing cystectomy received surgery due to chronic bladder infection and not because of cancer recurrence.
  • Figure 1 Kaplan Meier plot for time to recurrence in the bladder after a follow- up time up to 2.9 years.
  • Figure 4 Kaplan Meier plot for time to recurrence in the bladder for the FAS (follow up to 4 years).
  • sequences listed as SEQ ID N0.1 and SEQ ID NO.2 are as follows:
  • SEQ ID NO. 1 Nucleotide sequence encoding a phagolysosomal domain (nt 211-1722) and a stop codon (nt 1879-1881) atgacagacg tgagccgaaa gattcgagct tggggacgcc gattgatgat cggcacggca gcggctgtag tccttccggg cctggtgggggg cttgccggcg gagcggcaac cgcgggcgcg ttctccggc cggc cggc cggc cggggc c cggggcgagtac ctgcagtctg caaagcaatc cgctgcaaat aaattgcact cagcaggaca aaaattgcact cagcaggaca aaaattgcact cag

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The invention relates to a recombinant Mycobacterium cell for use as an immunotherapeutic agent in the treatment of bladder carcinoma, particularly in the second-line treatment of non-muscle-invasive bladder carcinoma.

Description

Recombinant Mycobacterium as an Immunotherapeutic Agent for the Second-Line Therapy of Bladder Carcinoma
Description
The invention relates to a recombinant Mycobacterium cell for use as an immunotherapeutic agent in the therapy of bladder carcinoma, particularly in the second-line therapy of non-muscle-invasive bladder carcinoma.
Urothelial bladder carcinoma is the 5th most common cancer. In the United States, about 75.000 new cases are diagnosed each year 4.5% of all new cancers, and approximately 15.600 deaths are expected. In Germany, about 16.000 new cases are diagnosed each year. Because a recurrence of disease is likely in bladder carcinoma, patients must undergo surveillance for an extended period.
Most bladder carcinomas begin in transitional epithelial cells that make up the inner lining of the bladder. As these tumors grow, they can invade the surrounding connective tissue and muscle. In advanced disease, tumors spread beyond the bladder to nearby lymph nodes or pelvic organs or metastasize to more distant organs such as lung, liver, and bone.
The overall 5-year survival rate for bladder carcinoma is 77%, and this rate has not changed significantly over the last 10 years. When considered by stage, the 5-year relative survival rates for patients with tumors restricted to the inner layer of the bladder are 96% and 69%, respectively. The rates drop to 34% for those with disease that has spread locally beyond the bladder and to 6% with distant metastases.
In the treatment of bladder carcinoma, tumor recurrence is a major concern, even for patients with low-grade disease and requires extensive follow-up. Better treatments, such as novel immunotherapies, might reduce recurrence rates and improve the survival of patients with bladder carcinoma. For patients with non-muscle invasive bladder carcinoma, treatment usually involves a surgical removal of the tumor followed by chemotherapy, usually mitomycin C, within the bladder (so-called intravesical chemotherapy). After recovering from surgery, patients with a lower risk of disease progression may undergo surveillance or additional intravesical chemotherapy. Patients with moderate-to-high-grade disease often receive intravesical immunotherapy with an attenuated live bacterium Bacillus Calmette Guerin (BCG). This therapy is designated “conventional” or “standard” BCG therapy in the following. Conventional BCG therapy was the first FDA-approved immunotherapy and helps reduce the risk of bladder carcinoma recurrence by stimulating an immune response that targets the bacteria as well as any bladder carcinoma cells. Despite the proven efficacy of conventional BCG treatment, about 35 to 45% of patients experience disease recurrence by 5 years and in about 10 to 13% of patients the disease progresses of (Oddens et al., Eur Urol 63 (2013, 462-472).
Recurrence and progression to muscle-invasive disease may lead to additional chemotherapeutical, surgical, and radio-oncological interventions including cisplatin-based chemotherapy, transurethral resection of the bladder (TURB), cystectomy and chemo-radiotherapy. Recurrent bladder carcinoma may be treated with combination therapy regimens, including gemcitabine plus cisplatin or methotrexate, vinblastine, doxorubicin plus cisplatin. An alternative option for recurrent patients is a re-treatment with conventional BCG therapy (Yates et al, Eur Urol 62 (2012), 1088-1096). The earlier the BCG failure, the more probable is the failure of a second BCG cycle (Gallagher et al., Urology 71 (2008), 297-301). A second-line conventional BCG immunotherapy after initial BCG failure in non-muscle-invasive bladder cancer, however, was found to be not effective (Di Lorenzo et al., Cancer 2010, doi: 10.1002/cncr.24914). 87.5% patients failed to respond to BCG re-induction at one year. 37.5% of the patients had to undergo cystectomy and 40% underwent radiation therapy plus systemic chemotherapy after one year. The poor outcome in these patients failing to respond to conventional BCG therapy reflects the unmet medical need for improved bladder-sparing treatments after conventional BCG or other intravesical treatment failure. Thus, better treatment options are not only needed for a first-line therapy but also for patients with recurrence after a first course of standard BCG therapy.
Thus, it was an object of the present invention to provide an improved treatment for patients failing to respond to first-line conventional BCG therapy as these patients are at high risk of cancer progression. An improved treatment of these high-risk patients will increase bladder preservation rates and consequently improve quality of life and decrease health costs.
A recombinant BCG strain expressing a phagolysosomal escape domain is described in WO 99/101496, the content of which is herein incorporated by reference. The phagolysosomal escape domain enables the strain to escape from the phagosome of infected host cells by perforating the membrane of the phagosome. In order to provide an acidic phagosomal pH for optimal phagolysosomal escape activity, a urease-deficient recombinant strain was developed. This strain is disclosed in WO 2004/094469, the content of which is herein incorporated.
WO 2012/085101, the content of which is herein incorporated, discloses that a recombinant BCG strain expressing membrane-perforating listeriolysin (Hly) of Listeria monocytogenes and devoid of urease C induces superior protection against aerogenic challenge with Mycobacterium tuberculosis ( Mtb ) as compared to parental BCG in a preclinical model. Further, it is shown that both the recombinant and the parenteral strain induce marked Th1 immune responses, whilst only the recombinant BCG strain elicits are profound Th17 response in addition.
WO 2016/177717 discloses that a recombinant urease-deficient and listeriolysin-expressing recombinant BCG strain induces a superior immune response compared to the parenteral BCG in an animal model. Further, the start of a human clinical phase I/ll trial using recombinant BCG as an immunotherapeutic agent in patients after a first standard BCG therapy is reported.
Despite the discouraging results of Di Lorenzo et al. , supra, using conventional BCG therapy as a second-line treatment, the present inventors have surprisingly observed a high overall survival and a low disease recurrence rate even 3 or 4 years after the start of therapy using a urease-deficient and listeriolysin-expressing recombinant BCG as a second-line BCG immunotherapy, particularly after initial BCG failure in non-muscle-invasive bladder cancer.
A first aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject, e.g., a human subject suffering from bladder carcinoma, particularly from non-muscle- invasive bladder carcinoma as a second-line therapy.
Accordingly, the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject as a second-line therapy a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain. A further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use in the immunotherapeutic treatment of bladder carcinoma wherein the recombinant Mycobacterium cell is administered to a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma as a second-line therapy.
Accordingly, the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject as a second-line therapy a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain.
A further aspect of the present invention is a recombinant Mycobacterium cell which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use as an immunotherapeutic agent in the treatment of bladder carcinoma, in particular recurrent bladder carcinoma, wherein the subject to be treated has relapsed and/or has progressed after a first treatment of bladder carcinoma, particularly after a first treatment with conventional BCG therapy.
Accordingly, the present invention further relates to a method for the immunotherapy of bladder carcinoma in a subject in need thereof, particularly of non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma, wherein the subject has relapsed and/or has progressed after a first treatment of bladder carcinoma, particularly after a first treatment with conventional BCG therapy, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain.
A further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma, wherein disease recurrence is inhibited for a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years.
Accordingly, the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain, wherein disease recurrence is inhibited for a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years. A still further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma wherein overall survival is increased in a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years, or of at least 4 years.
Accordingly, the present invention further relates to a method for the immunotherapeutic treatment of bladder carcinoma, particularly of non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain, wherein overall survival is increased in a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years, or of at least 4 years.
A still further aspect of the present invention relates to a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use in a method for improving the life quality of a subject suffering from bladder carcinoma, particularly from non-muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent thereby reducing or avoiding cystectomy in said subject.
Accordingly, the present invention further relates to a method for improving the life quality of a subject suffering from bladder carcinoma, particularly from non- muscle-invasive bladder carcinoma, more particularly from recurrent bladder carcinoma in a subject, particularly in a human subject, comprising administering to said subject a recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain, and avoiding or reducing the need of cystectomy in said subject. It should be noted, that two or more of the features of the above-indicated aspects may be combined with each other.
Thus, the present invention relates to a second-line therapy with rBCG in combination with at least one of:
- treatment of a subject who has relapsed and/or has progressed after a first treatment of bladder carcinoma, particularly after a first treatment with conventional BCG therapy,
- an inhibition of disease recurrence for a time period of at least 1 year, particularly for a time period of at least 2 years or of at least 3 years, or of at least 4 years,
- an increase of overall survival to a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years,
- and
- an improvement of life quality comprising reducing or the need of cystectomy. Further, the present invention relates to an improvement of life quality comprising reducing or the need of cystectomy in combination with at least one of:
- a second-line therapy for a subject having a disease recurrence and/or disease progression after a failed first-line therapy,
- an inhibition of disease recurrence for a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years,
- an increase of overall survival to a time period of at least 1 year, and particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years.
In a particular embodiment, the present invention relates to a second-line therapy in combination with:
- an inhibition of disease recurrence for a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years,
- an increase of overall survival to a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years, or of at least 4 years,
- and optionally an improvement of life quality comprising reducing or the need of cystectomy.
The present inventors have found that a second-line immunotherapy comprising administration of a recombinant Mycobacterium cell as described herein was highly effective in reducing disease occurrence and increasing overall survival. In this context, the term “second-line immunotherapy” refers to the treatment of a subject, particularly a human subject, after failure of a first-line therapy. The term “failure” particularly means disease recurrence and/or disease progression including progression of the primary tumor and/or formation of metastases after first-line therapy. More particularly, the term “failure” means disease recurrence. The term “first-line therapy” particularly refers to a first-line therapy including an immunotherapy, and more particularly to a first-line immunotherapy comprising administration of standard BCG without a phagolysosomal escape domain as described herein, such as previously approved standard BCG as described in Rentsch, C A, Eur Urol 2014, PMID: 24674149. The term “second-line immunotherapy” comprises administration of a recombinant Mycobacterium cell as described herein after a single “first-line therapy” cycle or after multiple, e.g., 2, 3 or more “first-line therapy” cycles, particularly a single cycle or multiple cycles comprising administration of BCG without a phagolysosomal escape domain.
According to a preferred embodiment of the invention the recombinant Mycobacterium cell is administered as a second-line treatment. According to such embodiments, the individual to be treated may have received a first treatment, in particular of bladder carcinoma treatment, selected from the group of standard BCG treatment, chemotherapy including systemic and/or intravesical chemotherapy, bladder surgery, radiation and any combination thereof. For example, the first-line treatment may include cisplatin-based chemotherapy, in particular cisplatin-based chemotherapy followed by bladder surgery and/or radiation therapy, concomitant chemotherapy and standard BCG. Preferably, the individual to be treated has received standard BCG treatment as a first treatment of bladder carcinoma and/or underwent cystectomy or another local treatment or systemic chemotherapy.
The subject to be treated according to the present invention is suffering from bladder carcinoma, particularly non-muscle invasive bladder carcinoma (NMIBC) including carcinoma in situ (CIS). In particular embodiments, the subject is suffering from recurrent NMBIC. In some embodiments, the subject has a recurrent high-grade NMIBC for progression (e.g., score 7-23) based on the European Organization for Research and Treatment. In further embodiments, the subject has undergone to a transurethral resection of the bladder tumor (TURBT) before therapy start. In some embodiments, the subject has a tumor-free bladder state at therapy start, e.g., as confirmed by transurethral resection of the bladder (TURB) and biopsy. In particular embodiments, the subject is a patient with a high grade NMIBC tumor present at 6 months or both at 3 months and 6 months after start of a previous cycle of standard BCG therapy and worsening of the disease under standard BCG therapy including at least one of higher number of recurrences, higher tumor category or higher grade, and appearances of CIS, including low grade tumors, optionally in spite of an initial response.
According to an especially preferred embodiment, the subject is a smoker.
Administration of a recombinant Mycobacterium cell as described herein as a second-line therapy is highly effective in reducing disease occurrence for an extended time period, e.g., a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years or of at least 4 years after treatment start. 3 years after treatment start, the median time of disease recurrence is already 1.3 years and will probably increase further. At this time, no tumor recurrence was observed in about 49% of the patients.
Thus, the therapy of the present invention provides a disease-free time period of at least 1 year, particularly of at least 2 years, of at least 3 years, or of at least 4 years for at least 30% of the patients and particularly for at least 45% of the patients.
Notably, in the time period between 2 and 3 years or between 2 and 4 years after treatment start, no new disease recurrence was observed. Thus, the treatment is highly efficient in stopping disease recurrence in a sub-group of patients, who did not suffer from disease recurrence in a time period up to 2 years after treatment start.
In contrast thereto, administration of a standard BCG as a second-line therapy was not effective at all as described by Di Lorenzo et al. supra, since 2 years after treatment start in only less than 5% of the patients no tumor recurrence was observed. This high clinical efficiency of the therapy of the present invention was found without the occurrence of clinically significant adverse events. Thus, the treatment translates into an overall survival benefit for the patients.
Thus, administration of a recombinant Mycobacterium cell as described herein is highly effective in increasing the overall survival in an extended time period, e.g., a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years or of at least 4 years. 3 years after start of the treatment, the overall survival rate was between 70% and 80%.
In particular embodiments, the therapy of the invention is suitable in avoiding cystectomy in a large number of patients resulting in a high improvement of life quality. For example, cystectomy may be avoided for a time period of at least a 1 year, particularly for a time period of at least 2 years, of at least 3 years or of at least 4 years in at least 30% of the patients and particularly for at least 45% of the patients.
The immunotherapeutic agent is a live recombinant Mycobacterium cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising (a) a domain capable of eliciting an immune response and (b) a phagolysosomal escape domain. The domain capable of eliciting an immune response is preferably an immunogenic peptide or polypeptide from a pathogen or an immunogenic fragment thereof.
The Mycobacterium cell is preferably an M. bovis cell, an M. tuberculosis cell, particularly an attenuated M. tuberculosis cell or other Mycobacteria, e.g. M. microti, M. smegmatis, M. canettii, M. marinum or M. fortuitum. More preferably, the cell is an attenuated recombinant M. bovis (BCG) cell, particularly an M. bovis BCG cell, more particularly a recombinant M. bovis BCG cell from strain Danish subtype Prague (Brosch et al., Proc. Natl. Acad. Sci. USA, 104 (2007), 5396-5601). In an especially preferred embodiment, the Mycobacterium cell is recombinant urease-deficient. In an especially preferred embodiment, the ureC sequence of the Mycobacterium cell is inactivated (AUrec), e.g., by constructing a suicide vector containing a ureC gene disrupted by a selection marker gene, e.g., the hygromycin gene, transforming the target cell with the vector and screening for selection marker-positive cells having a urease negative phenotype. In an even more preferred embodiment, the selection marker gene, i.e. , the hygromycin gene, is subsequently inactivated. In this embodiment, the cell is a selection marker-free recombinant Mycobacterium cell. Most preferably, the cell is selection marker-free recombinant BCG strain Danish subtype Prague characterized as recombinant BCG AUrec::Hly+. The domain capable of eliciting an immune response is preferably selected from immunogenic peptides or polypeptides from M. bovis, M. tuberculosis or M. leprae or from immunogenic fragments thereof having a length of at least 6, preferably at least 8 amino acids, more preferably at least 9 amino acids and e.g., up to 20 amino acids. Specific examples for suitable antigens are Ag85B (p30) from M. tuberculosis, Ag85B (a-antigen) from M. bovis BCG, Ag85A from M. tuberculosis and ESAT-6 from M. tuberculosis and fragments thereof. In other embodiments, the domain capable of eliciting an immune response is selected from non-Mycobacterium polypeptides. More preferably, the immunogenic domain is derived from the antigen Ag85B. Most preferably, the immunogenic domain comprises the sequence from aa. 41 to aa.51 in SEQ ID No.2.
The recombinant nucleic acid molecule further comprises a phagolysosomal escape domain, i.e., a polypeptide domain which provides for an escape of the fusion polypeptide from the phagolysosome into the cytosol of mammalian cells. Preferably, the phagolysosomal escape domain is a Listeria phagolysosomal escape domain, which is described in US 5,733,151, herein incorporated by reference. More preferably, the phagolysosomal escape domain is derived from the listeriolysin gene (Hly) of L. monocytogenes. Most preferably, the phagolysosomal domain is encoded by a nucleic acid molecule selected from: (a) a nucleotide sequence comprising nucleotides 211 - 1722 as shown in SEQ ID No.1, (b) a nucleotide sequence which encodes the same amino acid sequence as the sequence from (a), and (c) a nucleotide sequence hybridizing under stringent conditions with the sequence from (a) or (b).
Apart from the nucleotide sequence depicted in SEQ ID No.1 the present invention also comprises nucleic acid sequences hybridizing therewith. In the present invention, the term "hybridization" is used as defined in Sambrook et al. (Molecular Cloning. A laboratory manual, Cold Spring Harbor Laboratory Press (1989), 1.101 -1.104). In accordance with the present invention the term "hybridization" is used if a positive hybridization signal can still be observed after washing for one hour with 1 X SSC and 0.1 % SDS at 55°C, preferably at 62° C and more preferably at 68°C, particularly for 1 hour in 0.2 X SSC and 0.1 % SDS at 55°C, preferably at 62°C and more preferably at 68°C. A sequence hybridizing with a nucleotide sequence as per SEQ ID No.1 under such washing conditions is a phagolysosomal escape domain encoding nucleotide sequence preferred by the subject invention.
A nucleotide sequence encoding a phagolysosomal escape domain as described above may be directly obtained from a Listeria organism or from any recombinant source e.g., a recombinant E.coli cell containing the corresponding Listeria nucleic acid molecule, or a variant thereof as described above.
Preferably, the recombinant nucleic acid molecule encoding for a fusion polypeptide contains a signal peptide encoding sequence. More preferably, the signal sequence is a signal sequence active in Mycobacteria, preferably in M. bovis, e.g., a native M. bovis signal sequence. A preferred example of a suitable signal sequence is the nucleotide sequence coding for the Ag85B signal peptide, which is depicted in SEQ ID No.1 from nucleotide 1 to 120. Further, it is preferred that a peptide linker be provided between the immunogenic domain and the phagolysosomal escape domain. Preferably, said peptide linker has a length of from 5 to 50 amino acids. More preferably, a sequence encoding a linker as shown in SEQ ID No.1 from nucleotide 154 to 210 or a sequence corresponding thereto as regards the degeneration of the genetic code.
The nucleic acid may be located on a recombinant vector. Preferably, the recombinant vector is a prokaryotic vector, i.e. , a vector containing elements for replication or/and genomic integration in prokaryotic cells. Preferably, the recombinant vector carries the nucleic acid molecule of the present invention operatively linked with an expression control sequence. The expression control sequence is preferably an expression control sequence active in Mycobacteria, particularly in M. bovis. The vector can be an extrachromosomal vector or a vector suitable for integration into the chromosome. Examples of such vectors are known to the man skilled in the art and, for instance, given in Sambrook et at. supra.
The immunotherapeutic agent of the present invention is suitable for the treatment of bladder carcinoma, e.g., non-invasive bladder carcinoma, e.g. non-invasive papillary carcinoma in situ (Ta), non-invasive carcinoma in situ (Tcis), tumor invading subepithelial connective tissue (Ti), tumor invading superficial muscle (inner half) {ha), tumor invading deep muscle (outer half) (T2b), tumor invading perivesical tissue (T3 including T3a and T3b), tumor invading prostate, uterus or vagina (T4a), and tumor invading pelvic wall or abdominal wall (T4b). Particularly, the tumor is a superficial tumor or carcinoma in situ (Tcis), non-invasive papillary carcinoma (Ta), or a tumor invading subepithelial connective tissue (Ti). The immunotherapeutic treatment is suitable in the treatment of primary bladder carcinoma and/or in the treatment of recurring bladder carcinoma.
The immunotherapeutic agent is locally administered to the tumor site, i.e., to the site of a primary tumor before surgery or after surgery and optionally after chemotherapy. For the treatment of urothelial bladder carcinoma, the agent is preferably administered by vesicular instillation into the urinary bladder.
The immunotherapeutic agent is administered to the subject to be treated in an effective dose. For a human subject, the dose for an administration may be about 106 to 1010 viable units (CFU), e.g., about 107 to 5 x 109 or 108 to 3 x 109 viable units. Preferably, the dose for an administration is about 2 x 109 viable units (CFU). Preferably, the immunotherapeutic agent is administered several times, e.g., at least 3 times or at least 5 times up to 30 times, particularly about 15 times, at predetermined times during the treatment.
The immunotherapeutic agent is usually provided as a pharmaceutical preparation, which comprises the recombinant Mycobacterial cell in solid form, e.g., a lyophilized or cryoconserved preparation, which is reconstituted with a suitable liquid carrier before use. Alternatively, the preparation may be provided in liquid form, e.g., as suspension.
In one embodiment, the immunotherapeutic agent of the invention is administered for the treatment of carcinoma in situ. A standard schedule may comprise weekly administration of the agent for at least 4, e.g., 4, 5, 6, 7 or 8 weeks as an induction therapy. The induction therapy should not start until 2- 3 weeks after primary tumor surgery. After a treatment-free interval of, e.g., 4 weeks, administration may continue using maintenance therapy for at least 6 months or at least 1 year.
In a further embodiment, the immunotherapeutic agent is administered in an induction therapy in the prophylactic treatment of tumor recurrence. In this embodiment, therapy may start about 2-3 weeks after biopsy of the tumor site and be repeated, e.g., at weekly intervals for at least 4, e.g., 4, 5, 6, 7 or 8 weeks. In intermediate and high-risk tumors this may be followed by maintenance therapy.
Maintenance therapy may comprise long-term therapy, e.g., 6, 9 or 12 months therapy or even longer with treatments at monthly intervals. Alternatively, maintenance therapy may comprise 2, 3 or 4 administrations at weekly intervals, at month 3, 6, 12, 18, 24, 30 and 36.
In a particular embodiment, the immunotherapeutic agent, particularly recombinant BCG AUrec: : Hly+, is used for the treatment of non-muscle invasive bladder cancer in patients with recurrence after standard BCG therapy. The immunotherapeutic agent is administered into the bladder according to a schedule involving weekly instillations during an induction phase with e.g., 6 weekly instillations, a first maintenance phase after about 3 months with e.g., 3 weekly instillations, a second maintenance phase after about 6 months with e.g., 3 instillations and a third maintenance phase after about 12 months with e.g., 3 instillations.
The administration as immunotherapeutic agent of the recombinant Mycobacterium cell as described above may be combined with further anti tumor therapy, e.g., radiation and/or chemotherapy. Further, the immunotherapy as described above may be combined with a non-tumor site specific administration of the recombinant Mycobacterium cell in order to provide a general stimulation of the immune system. This non-site specific administration may be effected as described in WO 2012/085101 , e.g., before surgery of the primary tumor. In this case, the agent is preferably administered to a human subject in a dose of about 1-10 x 105, preferably about 2-8 x 105 cells. The agent is preferably administered as a single dose, e.g., by injection. Subcutaneous injection is preferred. Further, it is preferred to administer the agent without adjuvant.
Further, the invention is described in more detail by the following Figures and Examples 1 and 2. The immunotherapeutic agent “ rBCG ” used in these examples is recombinant M. bovis (BCG) Danish subtype Prague with an inactivated ureC sequence (AUrec) and without functional selection marker gene which expresses an Ag85B/Flly fusion protein as shown in SEQ ID No.2 (Hly+). Example 1 Phase I/ll open label clinical trial assessing safety and efficacy of intravesical instillation of recombinant BCG (rBCG) in human patients with recurrent non-muscle invasive bladder cancer after standard BCG therapy
1.1 Phase I Phase I was conducted to determine safety, tolerability, and the recommended phase II dose of intravesical rBCG instillations in patients with recurrence of non-muscle-invasive bladder cancer after TURB and standard BCG therapy.
1.2 Phase II
Phase II was conducted to investigate the efficacy, safety, tolerability and immunogenicity of intravesical rBCG instillations in patients with recurrence of non-muscle-invasive bladder cancer after TURB in standard BCG therapy. 1.3 Clinical protocol rBCG was administered into bladder in 15 weekly instillations (induction phase: instillations 1-6; maintenance 3 months: instillations 7-9; maintenance 6 months: instillations 10-12, maintenance 12 months: instillations 13-15).
The primary endpoint of the phase I was dose limiting toxicity (DLT) of intravesical rBCG instillations in patients with recurrence after standard BCG therapy in non-muscle invasive bladder cancer (NMIBC). The DLT period corresponds to 3 instillations plus 1 week and covers acute toxicities induced by treatment. Patients were treated in two cohorts of three, following the rules of a 3 + 3 design (dose de-escalation rules: if patients treated at dose level 1 show signs of DLT, dose of instilled rBCG will be reduced to level -1 , which is 10 times lower than level 1 ). The dose levels were as follows:
• Dose level 1: 1 - 19.2 x 108 CFUs of rBCG
• Dose level -1 : 1 - 19.2 x 107 CFUs of rBCG
1.4 Selection of patients
1.4.1 Inclusion criteria
- Histologically confirmed diagnosis of recurrent NMIBC including repeat TURB confirming tumor-free state of the bladder; for patients with pure CIS no repeat TURB is necessary.
- Negative cytology before start of treatment except for patients with concomitant CIS.
- Planned treatment start is 2-6 weeks after last TURB.
- One previous cycle of intravesical BCG (induction phase with at least 5 instillations ± maintenance) not more than 5 years ago for NMIBC.
- Recurrent high-grade NMIBC for progression (progression score 7-23) based on the European Organization for Research and Treatment of Cancer scoring system, failing BCG therapy (Babjuk M, Eur Urol 2008 PMID 18468779), for whom radical cystectomy or re-induction with standard BCG is indicated.
This particularly includes patients with high grade NMBIC tumor(s) present at both 3 and 6 months after start of the previous cycle of BCG therapy, and any worsening of the disease under BCG treatment such as higher number of recurrences, higher tumor category or higher grade, or appearances of CIS, including low grade tumors, in spite of an initial response.
1.4.2 Exclusion criteria
- Current or previous > T2 urothelial carcinoma (UC) of the urinary bladder.
- Concomitant UC of upper urinary tract, prostate or urethra, small cell carcinoma of the bladder, micropapillary urothelial carcinoma, pure squamous cell carcinoma of the bladder, pure adenocarcinoma, lympho-vascular invasion in the TURB specimen..
- Previous or current evidence of UC in a bladder diverticulum.
- Evidence of metastatic disease in the (thoraco-) abdominal CT scan at registration. - Bladder surgery or dramatic catheterization or TURB within 2 weeks prior to the start of treatment.
- Administration of systemic cytotoxic agents within the past 3 years or administration of repeated cytostatic/cytotoxic drugs by intravesical instillations after EUC recurrence after the first BCG therapy (during the past 5 years).
1.5 Current status
Clinical phase I is completed. Phase II was conducted with dose level 1: 1- 19.2 x 108CFU of rBCG.
The active agent was administered in 50 ml drug solution containing dextran, glucose, 0.9% sodium chloride, 0.025% Tween 80 and water for injection.
The follow-up data for recurrence in the bladder status of all evaluable phase II patients at 3 years are shown in Table 1. 20 patients experienced a confirmed event and 1 patient an unconfirmed event. Ten patients remained alive and on follow-up for this endpoint.
40 patients were included, all with high-grade tumors as specified above. The rate of CIS was high (68%). Recurrent-free state in the bladder (recurrence- free survival at 60 weeks was 49.3% and at three years 43.7 % with a 95% confidence interval [26,9%, 59,4% ]as shown in Fig. 1.
The treatment was safe and well tolerated, with only 5% of patients unable to tolerate adequate induction therapy. No dose-limiting toxicity occurred and no grade 3 or 4 adverse events were observed.
Overall survival after a median follow-up time of 2.9 years is shown in Fig. 2. Until this analysis, 7 patients died, 9 patients were lost to follow-up and 24 patients remained on follow-up. Only two patients (8%) were required to undergo cystectomy. 4 patients were required to switch to chemotherapy. Example 2 Phase I/ll open label clinical trial assessing safety and efficacy of intravesical instillation of recombinant BCG (rBCG) in human patients with recurrent non-muscle invasive bladder cancer after conventional BCG therapy
2.1 Patients and Methods
2.1.1 Study design and participants
The trial was designed as a multicentre, open-label, single-arm, phase I/ll study and conducted in compliance with the current version of the declaration of Helsinki, the ICH-GCP, and with national legal and regulatory requirements. Written informed consent was obtained from all patients prior to enrolment.
Eligible patients had recurrent NMIBC with intermediate and high-risk for progression (score 7-23 based on the European Organization for Research and Treatment of Cancer scoring system) after conventional BCG therapy.
Persistent T1 disease or high-grade non-muscle invasive tumour(s) present at both 3 and 6 months after start of the previous cycle of BCG therapy, or any worsening of the disease under BCG treatment, such as a higher number of recurrences, higher tumour category or higher grade, or appearance of carcinoma in situ (CIS), in spite of an initial response were required (EAU 2008 guideline definition). Prior therapy was defined as one previous cycle of intravesical BCG (induction phase >5 instillations ± BCG maintenance). For inclusion, histologically confirmed diagnosis of recurrent NMIBC and a repeat transurethral resection of the bladder (TURB) confirming a tumour-free state according to current pathology guidelines were mandatory. In patients presenting with CIS, selective upper tract cytology and biopsies of the prostatic urethra were recommended. At the start of the study treatment, bladder wash cytology had to be negative, except for patients with pure or concomitant CIS and imaging without evidence of metastatic disease.
Exclusion criteria were stage > T2 urothelial carcinoma of the bladder, concomitant urothelial carcinoma of the upper urinary tract, the non-prostatic urethra, or evidence of metastatic disease
(https://clinicaltrials.gov/ct2/show/NCT02371447). rBCG was provided as a formulated lyophilized powder of 1-19.2 c 108 CFU/vial of live Mycobacterium bovis BCG ureCwhly (SIIPL, Serum Institute of India Private Limited, Pune, India) and reconstituted in 50 ml of 0.9% saline water for intravesical application. Patients were scheduled for standard treatment of 6 weekly intravesical instillations followed by maintenance of 1 year (3 instillations at 3, 6 and 12 months after first instillation).
The primary endpoint of the phase II part was defined as the recurrence-free rate (RFR) in the bladder 60 weeks after registration. In case of suspected recurrence due to positive cytology or visual detection of a tumour in the bladder at cystoscopy, histological proof of the recurrence was required. A recurrence in the bladder associated with cancer in the prostatic urethra or evidence for cancer in the upper urinary tract were not considered as a recurrence in the bladder. Predefined secondary endpoints included time from trial registration to recurrence in the bladder, time to progression, overall survival (OS), AEs, tolerability, and QoL. Progression was defined as progression to muscle-invasive bladder cancer or progression to metastatic disease. OS was calculated from registration until death from any cause. Tolerability was defined as the proportion of patients finishing five instillations of induction within 12 weeks after treatment start. 2.1.2 Statistical analyses
For sample size calculation, we estimated the RFR from one of the few prospective randomized European trials comparing BCG re-exposure versus gemcitabine. In this trial, the group receiving BCG had an estimated RFR and progression-free rate 60 weeks after the last TURB of 15% and 62.5%, respectively. A single-stage design was used with HO: RFR at 60 weeks <15%; and H1 : RFR at 60 weeks >30%. For a significance level of 10% and a power of 80%, 37 evaluable patients for phase II were required based on the Kaplan- Meier estimator evaluated 60 weeks after registration (R package Optlnterim). The 6 patients from the phase I part were included for the phase II part. To account for non-evaluable patients, the sample size was increased to 39 patients.
2.2 Results
2.2.1 Patients
From September 2015 to April 2018, 42 patients were enrolled into the trial. A total of 16 sites were activated and 14 sites actively recruited patients. Two patients had to be excluded from efficacy analysis because of major inclusion criteria violation but were kept in for safety/ITT analysis. Forty patients defined the full analysis set (FAS) and the per protocol population (PP) consisted of 22 patients. Definitions of analysed populations and detailed reasons for exclusions are given in Fig. 3. The detailed tumour characteristics of the primary NMIBC occurrences of the patient cohort are given in Table 2 and the detailed patient and tumour characteristics of the FAS in Table 3. The median risk for progression of the recurrent NMIBC increased by 7 risk points as compared to the primary occurrence. A history of chemical exposure as a risk factor for bladder cancer was identified in 3 patients, while 21 patients (52.5%) had a smoking history with a median (range) of 38 (4.0-99.0) pack-years. Concomitant or pure CIS was present in 27 (67.5%) patients. Fourteen patients (35%) received BCG maintenance therapy. The median progression score was 16 (7-20) for those patients with and 16 (7-19) for those without previous BCG maintenance.
All patients enrolled in the trial started trial treatment. Thirty-two patients (80%) started maintenance therapy. Fifteen patients (37.5%) received all scheduled instillations of induction and maintenance therapy. Detailed reasons for stopping treatment during the trial are given in Fig. 3. Median follow-up at the time of analysis was 74.4 (12.3 - 140.9) weeks.
2.2.2 Primary endpoint
After rBCG induction and maintenance therapy, 49.3% [95% Cl 32.1, 64.4] of the patients were without evidence of recurrence in the bladder 60 weeks after trial registration. After a median follow-up of 2.9 years, the RFR remained at 47.4% [30.4%, 62.6%] and 43.7% [26.9%, 59.4%] 2 and 3 years after trial registration, respectively. The median time from trial registration to recurrence in the bladder was 54.1 weeks [95% Cl 38.4 weeks] (Fig. 4). The RFR in the bladder for the ITT and PP populations 60 weeks after registration was 48.0% [95% Cl 31.1, 63.0] and 40.8% [95% Cl 19.9, 60.9], respectively. If accounting for all potential recurrences including histologically unconfirmed but clinically evident extravesical recurrences in the FAS population, the 60-week RFR reached 47.6% [95% Cl 30.8, 62.6] Out of the patients with CIS (n=27), 15 (55.6%; 95% Cl [35.3%, 74.5%]) had a complete response (CR) at 12 weeks after treatment start. The median duration of response (DOR) for these 15 patients was 1.1 years (95% Cl [0.4 years - not reached]). Further subgroup evaluations related to the primary endpoint are given in Table 3. 2.2.3 Secondary endpoints
Seven patients from the FAS experienced progressive disease (median time to progression not reached). Three patients showed progression to muscle- invasive disease, and 4 patients progressed with metastatic disease (regional lymph nodes n=3, distant metastasis n=1). None of the patients with metastatic disease were found to have concomitant muscle invasive disease in the bladder.
Follow-up treatment for the patients with recurrence after trial treatment is listed in Table 5. Until the time of analysis, 5 patients had died (2 from bladder cancer, 2 from other cancers not known at inclusion, and one from acute respiratory distress syndrome unrelated to the study treatment).
In summary, 60 weeks after trial registration, progression-free rate and OS rate were 76.3% [95% Cl 56.4, 88.0] and 92.9% [95% Cl 74.3, 98.2], respectively.
2.2.4 Adverse events and tolerability
Treatment-related AEs are listed in Table 6. The major grade 2 AE term was genitourinary tract infection with common uropathogenic bacteria in one third of the patients. Two patients had to undergo in-hospital antibiotic treatment for urogenital tract infection.
No grade 4 or 5 AEs occurred during the treatment phase. Two of the 42 patients did receive less than five instillations, one because of a BCG-induced systemic inflammatory reaction (BCGitis) due to traumatic catheterisation and one because of patient refusal (Fig. 3).
2.2.5 PPD testing 77.5% of the patients were PPD negative (PPD-) at treatment start (Table 2).
Conversion of the PPD test after finishing the rBCG therapy was seen in 7 patients only (PPD- to PPD+: n=5; PPD+ to PPD-: n=2). Fifteen patients (37.5%) did not convert from PPD- to PPD+. In these patients, the RFR in the bladder was 62.9% (Table 4).
2.2.6 Quality of life
All 40 patients in the FAS completed the QoL questionnaire at baseline; 32 of 33 (97%) patients who started the maintenance therapy completed it at the beginning of maintenance therapy and 32 of 40 (77.5%) completed it at the end of treatment. Fourteen patients (45.2%) completed the QoL assessment after the 15 instillations as required by protocol. Patients reported high levels of QoL for the functional scales and a low symptom burden at baseline. Some impairment was reported with regard to urinary symptoms, future worries, and sexual issues. Most of the QoL scales showed stable scores from baseline to the beginning of maintenance therapy. Small improvements were observed for emotional functioning and urinary symptoms. Future worries improved (i.e. , patients expressed less worries) to a clinically relevant extent. Almost half (n=13, 49%) of the patients reported a clinically relevant improvement in emotional functioning, whereas approx one third reported a clinically relevant deterioration in physical well-being (n=10, 30%), global health status/QoL (n=11, 33%), or fatigue (n=10, 30%) during induction therapy. No major changes from baseline to the end of treatment were observed for any of the QoL scales in those patients who completed treatment according to protocol. 2.3 Discussion
2.3.1 Endpoint analysis
We have chosen a high-risk population for recurrence and progression with previous conventional BCG exposure to test rBCG. Given the poor outcome of BCG re-exposure in these patients with reported 12 and 24 months RFR of 15% and 3%, respectively, we considered a placebo-controlled trial or a trial comparator with conventional BCG as unethical. With 49.3% RFR in the bladder, this single-arm trial has clearly met the primary endpoint of the predefined RFR >30% 60 weeks after trial registration. This is also true when extending the definition of recurrence to extra-vesical recurrences including histologically unconfirmed clinically evident recurrences in the FAS population, the ITT and the PP population, as well as whether the patients received BCG maintenance or not (Table 3). With a RFR of 47.4% and 43.7% after 2 and 3 years, the treatment response remains stable indicating a potentially promising long-term effect.
After the initiation of this trial, the Food and Drug Administration (FDA) published a new definition for BCG failure and the International Bladder Cancer Group recommended a clinically meaningful initial complete response rate (for CIS) or recurrence-free rate (for papillary tumours) of at least 30% at 12 months for single arm trials in BCG unresponsive patients. According to the FDA definition of BCG failure, 16 patients (40.0%) qualified for BCG unresponsive disease. When applying these more stringent criteria, the 60- week RFR dropped to 24.0% (Table 3).
Further subgroup analysis related to the primary endpoint indicated that smoking status, presence of CIS, and previous BCG maintenance therapy were possibly associated with a worse outcome in this trial, which were, however, not statistically significant - a result again limited by the small sample size (Table 3).
Most of the patients with recurrence underwent cystectomy or other local treatments, and 3 patients underwent systemic chemotherapy (Table 4). While missed metastatic disease at inclusion is a potential explanation for such a fatal development, it cannot be excluded that metastatic disease evolved after TUR and/or under intravesical trial therapy. Notably, none of the patients with metastatic disease were found to have concomitant muscle invasive disease in the bladder.
2.3.2 Adverse events and tolerability
The most common AE was genitourinary (GU) infection with common bacteria, occurring in one third of the patients. Potential explanations are i) that lubricants containing antiseptics for instillations were not allowed, and ii) that asymptomatic patients with positive urine dipstick had to undergo antibiotic treatment when urine culture was positive. Importantly, the GU infections did not significantly impact on QoL of the patients (not shown). No grade 4 or 5 AE occurred, and tolerability (defined as patients receiving more than 4 instillations during induction) was 95.2%. The treatment can therefore be considered as safe and well tolerated.
2.3.3 PPD testing
Experimental models have suggested that either a PPD+ state prior to the start of treatment or a conversion from PPD- to PPD+ state during treatment might predict response to therapy. In clinical trials, the clinical correlation of the PPD test with the outcome has been conflicted by heterogeneous results, by previous mycobacterial exposure, and different handling in the execution and interpretation of the test itself. In the present study, most patients (77.5%) had a negative PPD test before the start of the treatment with rBCG, although they had been exposed to BCG previously. Unexpectedly, patients with negative PPD test at inclusion showed a 60-week RFR in the bladder of 52.8%, and even without conversion from PPD- to PPD+, the RFR in the bladder remained 62.9% (n=15; Table 3), indicating that a negative PPD test and non-conversion might not be able to predict treatment outcome in this setting.
2.3.4 QoL
Patients reported overall stable QoL during induction and maintenance treatment, with small improvements in emotional functioning and future worries. Around 30% of patients reported clinically relevant deteriorations for physical functioning, global QoL, or fatigue during induction therapy. Interestingly, for patients who completed the treatment according to protocol no major changes were observed for any of the QoL scales. Again, the limited sample size without a comparator arm stresses for a cautious interpretation of these QoL results.
If the favorable Qol and tolerability is confirmed in larger studies, rBCG has the potential to decrease the proportion of patients becoming intolerable to BCG treatment and may be combined with other agents such as check-point inhibitors in order to increase efficiency.
2.4 Conclusions
The study has met the primary endpoint and demonstrates freedom from recurrence in the bladder after intravesical rBCG treatment in almost half of the patients after one year. rBCG has a promising tolerability, safety, and QoL profile.
Tables and Figure Legends
Table 1. Follow-up data for recurrence in the bladder status of all evaluable phase II patients at 3 years.
Table 2. Baseline tumor characteristics of first occurrence of NMIBC (before first conventional BCG treatment).
Table 3. Baseline patient and tumor characteristics of the FAS (full analysis set). WHO: World Health Organisation. BCG: Bacillus Calmette Guerin. CIS: Carcinoma in situ. EORTC: European Organisation for Research and Treatment of Cancer.
Table 4. Subgroup efficacy analysis of the FAS showing recurrence-free rate in the bladder at 60 weeks together with 95% Cl. CIS: Carcinoma in situ. BCG: Bacillus Calmette-Guerin. FDA: Food and Drug Administration. PPD: Purified protein derivative.
Table 5. Treatment of patients with recurrence and/or progression after trial treatment. BCG: Bacillus Calmette Guerin. Some patients received more than one treatment. One of the 13 patients undergoing cystectomy received surgery due to chronic bladder infection and not because of cancer recurrence.
* one of these treatments was performed based on a recurrence later than 60 weeks after registration.
Table 6. Treatment associated adverse events (AE) in the treatment phase. BCG: Bacillus Calmette-Guerin. GU: Genito-urinary.
Figure 1. Kaplan Meier plot for time to recurrence in the bladder after a follow- up time up to 2.9 years.
Figure 2. Overall survival after a follow-up time up to 2.9 years.
Figure 3. CONSORT 2010 diagram of SAKK06/14. BCG: Bacillus Calmette- Guerin. ITT: Intention to treat. FAS: Full analysis set (population). PP: Per- protocol (population).
1 includes also the 2 exclusions from FAS.
Figure 4: Kaplan Meier plot for time to recurrence in the bladder for the FAS (follow up to 4 years).
Table 1
Figure imgf000031_0001
Table 2.
Figure imgf000032_0001
Table 3.
Figure imgf000033_0001
Table 4
Figure imgf000034_0001
Table 5.
Figure imgf000035_0002
_
Table 6.
Figure imgf000035_0001
The sequences listed as SEQ ID N0.1 and SEQ ID NO.2 are as follows:
SEQ ID NO. 1: Nucleotide sequence encoding a phagolysosomal domain (nt 211-1722) and a stop codon (nt 1879-1881) atgacagacg tgagccgaaa gattcgagct tggggacgcc gattgatgat cggcacggca gcggctgtag tccttccggg cctggtgggg cttgccggcg gagcggcaac cgcgggcgcg ttctcccggc cggggctgcc ggtcgagtac ctgcagtctg caaagcaatc cgctgcaaat aaattgcact cagcaggaca aagcacgaaa gatgcatctg cattcaataa agaaaattca atttcatcca tggcaccacc agcatctccg cctgcaagtc ctaagacgcc aatcgaaaag aaacacgcgg atgaaatcga taagtatata caaggattgg attacaataa aaacaatgta ttagtatacc acggagatgc agtgacaaat gtgccgccaa gaaaaggtta caaagatgga aatgaatata ttgttgtgga gaaaaagaag aaatccatca atcaaaataa tgcagacatt caagttgtga atgcaatttc gagcctaacc tatccaggtg ctctcgtaaa agcgaattcg gaattagtag aaaatcaacc agatgttctc cctgtaaaac gtgattcatt aacactcagc attgatttgc caggtatgac taatcaagac aataaaatcg ttgtaaaaaa tgccactaaa tcaaacgtta acaacgcagt aaatacatta gtggaaagat ggaatgaaaa atatgctcaa gcttatccaa atgtaagtgc aaaaattgat tatgatgacg aaatggctta cagtgaatca caattaattg cgaaatttgg tacagcattt aaagctgtaa ataatagctt gaatgtaaac ttcggcgcaa tcagtgaagg gaaaatgcaa gaagaagtca ttagttttaa acaaatttac tataacgtga atgttaatga acctacaaga ccttccagat ttttcggcaa agctgttact aaagagcagt tgcaagcgct tggagtgaat gcagaaaatc ctcctgcata tatctcaagt gtggcgtatg gccgtcaagt ttatttgaaa ttatcaacta attcccatag tactaaagta aaagctgctt ttgatgctgc cgtaagcgga aaatctgtct caggtgatgt agaactaaca aatatcatca aaaattcttc cttcaaagcc gtaatttacg gaggttccgc aaaagatgaa gttcaaatca tcgacggcaa cctcggagac ttacgcgata ttttgaaaaa aggcgctact tttaatcgag aaacaccagg agttcccatt gcttatacaa caaacttcct aaaagacaat gaattagctg ttattaaaaa caactcagaa tatattgaaa caacttcaaa agcttataca gatggaaaaa ttaacatcga tcactctgga ggatacgttg ctcaattcaa catttcttgg gatgaagtaa attatgatcc tgaaggtaac gaaattgttc aacataaaaa ctggagcgaa aacaataaaa gcaagctagc tcatttcaca tcgtccatct atttgccagg taacgcgaga aatattaatg tttacgctaa agaatgcact ggtttagctt gggaatggtg gagaacggta attgatgacc ggaacttacc acttgtgaaa aatagaaata tctccatctg gggcaccacg ctttatccga aatatagtaa taaagtagat aatccaatcg aatatgcatt agcctatgga agtcagggtg atcttaatcc attaattaat gaaatcagca aaatcatttc agctgcagtt ctttcctctt taacatcgaa gctacctgca gagttcgtta ggcgcggatc cggaattcga agcttatcga tgtcgacgta g SEQ ID NO. 2:
Amino acid sequence encoded by the nucleotide sequence of SEQ ID NO: 1
Met Thr Asp Val Ser Arg Lys lie Arg Ala Trp Gly Arg Arg Leu Met lie Gly Thr Ala Ala Ala Val Val Leu Pro Gly Leu Val Gly Leu Ala Gly Gly Ala Ala Thr Ala Gly Ala Phe Ser Arg Pro Gly Leu Pro Val Glu Tyr Leu Gin Ser Ala Lys Gin Ser Ala Ala Asn Lys Leu His Ser Ala Gly Gin Ser Thr Lys Asp Ala Ser Ala Phe Asn Lys Glu Asn Ser lie Ser Ser Met Ala Pro Pro Ala Ser Pro Pro Ala Ser Pro Lys Thr Pro lie Glu Lys Lys His Ala Asp Glu lie Asp Lys Tyr lie Gin Gly Leu Asp Tyr Asn Lys Asn Asn Val Leu Val Tyr His Gly Asp Ala Val Thr Asn Val Pro Pro Arg Lys Gly Tyr Lys Asp Gly Asn Glu Tyr lie Val Val Glu Lys Lys Lys Lys Ser lie Asn Gin Asn Asn Ala Asp lie Gin Val Val Asn Ala lie Ser Ser Leu Thr Tyr Pro Gly Ala Leu Val Lys Ala Asn Ser Glu Leu Val Glu Asn Gin Pro Asp Val Leu Pro Val Lys Arg Asp Ser Leu Thr Leu Ser lie Asp Leu Pro Gly Met Thr Asn Gin Asp Asn Lys lie Val Val Lys Asn Ala Thr Lys Ser Asn Val Asn Asn Ala Val Asn Thr Leu Val Glu Arg Trp Asn Glu Lys Tyr Ala Gin Ala Tyr Pro Asn Val Ser Ala Lys lie Asp Tyr Asp Asp Glu Met Ala Tyr Ser Glu Ser Gin Leu lie Ala Lys Phe Gly Thr Ala Phe Lys Ala Val Asn Asn Ser Leu Asn Val Asn Phe Gly Ala lie Ser Glu Gly Lys Met Gin Glu Glu Val lie Ser Phe Lys Gin lie Tyr Tyr Asn Val Asn Val Asn Glu Pro Thr Arg Pro Ser Arg Phe Phe Gly Lys Ala Val Thr Lys Glu Gin Leu Gin Ala Leu Gly Val Asn Ala Glu Asn Pro Pro Ala Tyr lie Ser Ser Val Ala Tyr Gly Arg Gin Val Tyr Leu Lys Leu Ser Thr Asn Ser His Ser Thr Lys Val Lys Ala Ala Phe Asp Ala Ala Val Ser Gly Lys Ser Val Ser Gly Asp Val Glu Leu Thr Asn lie lie Lys Asn Ser Ser Phe Lys Ala Val lie Tyr Gly Gly Ser Ala Lys Asp Glu Val Gin lie lie Asp Gly Asn Leu Gly Asp Leu Arg Asp lie Leu Lys Lys Gly Ala Thr Phe Asn Arg Glu Thr Pro Gly Val Pro lie Ala Tyr Thr Thr Asn Phe Leu Lys Asp Asn Glu Leu Ala Val lie Lys Asn Asn Ser Glu Tyr lie Glu Thr Thr Ser Lys Ala Tyr Thr Asp Gly Lys lie Asn lie Asp His Ser Gly Gly Tyr Val Ala Gin Phe Asn lie Ser Trp Asp Glu Val Asn Tyr Asp Pro Glu Gly Asn Glu lie Val Gin His Lys Asn Trp Ser Glu Asn Asn Lys Ser Lys Leu Ala His Phe Thr Ser Ser lie Tyr Leu Pro Gly Asn Ala Arg Asn lie Asn Val Tyr Ala Lys Glu Cys Thr Gly Leu Ala Trp Glu Trp Trp Arg Thr Val lie Asp Asp Arg Asn Leu Pro Leu Val Lys Asn Arg Asn lie Ser lie Trp Gly Thr Thr Leu Tyr Pro Lys Tyr Ser Asn Lys Val Asp Asn Pro lie Glu Tyr Ala Leu Ala Tyr Gly Ser Gin Gly Asp Leu Asn Pro Leu lie Asn Glu lie Ser Lys lie lie Ser Ala Ala Val Leu Ser Ser Leu Thr Ser Lys Leu Pro Ala Glu Phe Val Arg Arg Gly Ser Gly lie Arg Ser Leu Ser Met Ser Thr

Claims

Claims
1. A recombinant Mycobacterium cell, which is a urease-deficient, recombinant M. bovis BCG cell from strain Danish subtype Prague, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and
(b) a phagolysosomal escape domain for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a subject suffering from bladder carcinoma, as a second-line therapy.
2. The recombinant Mycobacterium cell of claim 1 for the use of claim 1 , which is a urease-deficient, recombinant M. bovis BCG cell from strain
Danish subtype Prague, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response comprising the amino acid sequence from aa.41 to aa.51 in SEQ ID No.2, and (b) a Listeria phagolysosomal escape domain encoded by a nucleic acid molecule selected from
(i) a nucleotide sequence comprising nucleotides 211-1722 as shown in SEQ ID No.1 ,
(ii) a nucleotide sequence which encodes the same amino acid sequence as the sequence from (i), and
(iii) a nucleotide sequence hybridizing under stringent conditions with the sequence from (i) or (ii), for use in a method, wherein the recombinant Mycobacterium cell is administered as an immunotherapeutic agent to a human subject suffering from bladder carcinoma as a second-line therapy.
3. The cell of claim 1 or 2 for the use of claim 1 or 2, wherein the recombinant nucleic acid molecule does not comprise any functional selection marker.
4. The cell of claim 1 , 2 or 3 for the use of claim 1 , 2 or 3, wherein the human subject is suffering from non-muscle-invasive bladder carcinoma (NMIBC).
5. The cell of claim 1 , 2 or 3 for the use of claim 1 , 2 or 4, wherein the human subject is suffering from recurrent bladder carcinoma.
6. The cell of claim 1 , 2 or 3 for the use of any one of claims 1 , 2 or 4-5, wherein the human subject has relapsed and/or has progressed after a first treatment of bladder carcinoma.
7. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-6, wherein the administration comprises vesicular instillation into the urinary bladder.
8. The cell of claim 1 , 2 or 3 for the use of any one of claims 1 or 4-7 in the treatment of a subject having previously undergone at least one unsuccessful first-line therapy of bladder carcinoma.
9. The cell of claim 1 , 2 or 3 for the use any of one of claims 1 or 4-8 in the treatment of a subject having previously undergone at least one unsuccessful first-line therapy of bladder carcinoma comprising an immunotherapy, particularly an immunotherapy with standard BCG.
10. The cell of claim 1 , 2 or 3 for the use of any one of claims 1 or 4-9 in the treatment of a subject having previously undergone at least one unsuccessful first-line therapy of bladder carcinoma comprising cystectomy or another local treatment and/or systemic chemotherapy.
11. The cell of claim 1 , 2 or 3 for the use of any one of any one of claims 1 or 4-10 in the treatment of a subject having previously undergone at least one unsuccessful first-line therapy of bladder carcinoma selected from the group of cisplatin-based chemotherapy, in particular cisplatin- based chemotherapy followed by bladder surgery and/or radiation therapy, concomitant chemotherapy and standard BCG.
12. The cell of claim 1 , 2 or 3 for the use of any one of any one of claims 1 or 4-11 , wherein the subject is a smoker.
13. The cell of claim 1 , 2 or 3 for the use of any one of any one of claims 1 or 4-12, wherein the immunotherapeutic agent is administered into the bladder according to a schedule involving weekly instillations
14. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-13, wherein the immunotherapeutic agent is administered into the bladder according to a schedule involving weekly instillations during (i) an induction phase with, in particular 6 weekly instillations, (ii) a maintenance phase of at least one year, in particular a first maintenance phase after about 3 months with e.g. 3 weekly instillations, a second maintenance phase after about 6 months with e.g. 3 instillations and a third maintenance phase after about 12 months with e.g. 3 instillations.
15. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-14, wherein the immunotherapeutic agent is administered at a dose of from about 107 to 5 x 109 CFU, particularly about 2 x 109 CFU per administration.
16. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-15, wherein disease recurrence is inhibited for a time period of at least 1 year, particularly for a time period of at least 2 years, of at least 3 years or of at least 4 years.
17. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-16, wherein the immunotherapy provides a disease-free time period of at least 1 year, particularly of at least 2 years, of at least 3 years or of at least 4 years for at least 30% of the patients and particularly for at least 45% of the patients.
18. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-17, wherein disease recurrence is blocked for at least one further year in a subject without disease recurrence for a time period of 2 years after treatment start.
19. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-18, wherein overall survival is increased compared to a subject without treatment in a time period of at least 1 year, particularly in a time period of at least 2 years, of at least 3 years or of at least 4 years.
20. The cell of claim 1, 2 or 3 for the use of any one of claims 1 or 4-19, wherein the need of cystectomy is reduced or avoided, e.g., for a time period of at least a 1 year, particularly for a time period of at least 2 years, of at least 3 years or of at least 4 years in at least 30% of the patients and particularly for at least 45% of the patients.
21. A method for the immunotherapeutic treatment of bladder carcinoma, particularly of non-muscle-invasive bladder carcinoma in a subject in need thereof, comprising administering to said subject as a second-line therapy a recombinant Mycobacterium cell, which is a urease-deficient, recombinant M. bovis BCG cell, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising:
(a) a domain capable of eliciting an immune response, and (b) a Listeria phagolysosomal escape domain.
22. A method for the immunotherapeutic treatment of bladder carcinoma, particularly of non-muscle-invasive bladder carcinoma in a human subject in need thereof, comprising administering to said subject as a second-line therapy a recombinant Mycobacterium cell, which is a urease-deficient, recombinant M. bovis BCG cell from strain Danish subtype Prague, which comprises a recombinant nucleic acid molecule encoding a fusion polypeptide comprising: (a) a domain capable of eliciting an immune response comprising the amino acid sequence from aa.41 to aa.51 in SEQ ID No.2, and
(b) a Listeria phagolysosomal escape domain encoded by a nucleic acid molecule selected from (i) a nucleotide sequence comprising nucleotides 211 -1722 as shown in
SEQ ID No.1,
(ii) a nucleotide sequence which encodes the same amino acid sequence as the sequence from (i), and
(iii) a nucleotide sequence hybridizing under stringent conditions with the sequence from (i) or (ii).
PCT/EP2022/070373 2021-07-22 2022-07-20 Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma WO2023001895A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
IL310225A IL310225A (en) 2021-07-22 2022-07-20 Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma
AU2022313491A AU2022313491A1 (en) 2021-07-22 2022-07-20 Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma
EP22751731.5A EP4373519A1 (en) 2021-07-22 2022-07-20 Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma
CA3224671A CA3224671A1 (en) 2021-07-22 2022-07-20 Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US202163224575P 2021-07-22 2021-07-22
US63/224,575 2021-07-22
EP21187253.6A EP4122491A1 (en) 2021-07-22 2021-07-22 Recombinant microbacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma
EP21187253.6 2021-07-22
US17/667,784 2022-02-09
US17/667,784 US20220160858A1 (en) 2015-05-04 2022-02-09 Recombinant Mycobacterium as an Immunotherapeutic Agent for the Treatment of Cancer

Publications (1)

Publication Number Publication Date
WO2023001895A1 true WO2023001895A1 (en) 2023-01-26

Family

ID=82846547

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2022/070373 WO2023001895A1 (en) 2021-07-22 2022-07-20 Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma

Country Status (5)

Country Link
EP (1) EP4373519A1 (en)
AU (1) AU2022313491A1 (en)
CA (1) CA3224671A1 (en)
IL (1) IL310225A (en)
WO (1) WO2023001895A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5733151A (en) 1996-08-23 1998-03-31 Edsall; David Electrical clamping connection device
WO1999001496A1 (en) 1997-06-30 1999-01-14 Cardiac Crc Nominees Pty. Ltd. Process for the purification of polyethers
WO2004094469A1 (en) 2003-04-23 2004-11-04 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Tuberculosis vaccine with improved efficacy
WO2012085101A1 (en) 2010-12-21 2012-06-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Recombinant mycobacterium as a vaccine
EP3090757A1 (en) * 2015-05-04 2016-11-09 Vakzine Projekt Management GmbH Recombinant mycobacterium as an immunotherapeutic agent for the treatment of cancer

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5733151A (en) 1996-08-23 1998-03-31 Edsall; David Electrical clamping connection device
WO1999001496A1 (en) 1997-06-30 1999-01-14 Cardiac Crc Nominees Pty. Ltd. Process for the purification of polyethers
WO2004094469A1 (en) 2003-04-23 2004-11-04 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Tuberculosis vaccine with improved efficacy
WO2012085101A1 (en) 2010-12-21 2012-06-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Recombinant mycobacterium as a vaccine
EP3090757A1 (en) * 2015-05-04 2016-11-09 Vakzine Projekt Management GmbH Recombinant mycobacterium as an immunotherapeutic agent for the treatment of cancer
WO2016177717A1 (en) 2015-05-04 2016-11-10 Vakzine Projekt Management Gmbh Recombinant mycobacterium as an immunotherapeutic agent for the treatment of cancer

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
BROSCH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 104, 2007, pages 5396 - 5601
CLINICALTRIALS GOV: "VPM1002BC in Recurrent Non-muscle Invasive Bladder Cancer", INTERNET CITATION, 10 February 2015 (2015-02-10), pages 1 - 4, XP002746390, Retrieved from the Internet <URL:https://clinicaltrials.gov/ct2/show/NCT02371447> [retrieved on 20151012] *
DI LORENZO ET AL., CANCER, 2010
GALLAGHER ET AL., UROLOGY, vol. 71, 2008, pages 297 - 301
MARK DOHERTY T ET AL: "Comparative analysis of different vaccine constructs expressing defined antigens from mycobacterium tuberculosis", JOURNAL OF INFECTIOUS DISEASES, UNIVERSITY OF CHICAGO PRESS, US, vol. 190, no. 12, 15 December 2004 (2004-12-15), pages 2146 - 2153, XP008120938, ISSN: 0022-1899, [retrieved on 20041108], DOI: 10.1086/425931 *
ODDENS ET AL., EUR UROL, vol. 63, 2013, pages 462 - 472
RENTSCH C A ET AL: "VPM1002- a recombinant BCG with favourable preclinical toxicity and immunogenicity for potential improvement of BCG immunotherapy for non-muscle invasive bladder cancer", EUROPEAN UROLOGY SUPPLEMENTS; 29TH ANNUAL CONGRESS OF THE EUROPEAN ASSOCIATION OF UROLOGY ABSTRACTS, ELSEVIER BV, NL, vol. 13, no. 1, 11 April 2014 (2014-04-11), pages e521 - 1, XP002746389, ISSN: 1569-9056, DOI: 10.1016/S1569-9056(14)60513-3 *
RENTSCH CYRILL ET AL: "MP39-08 A LISTERIOLYSIN EXPRESSING BCG WITH FAVOURABLE IMMUNOGENICITY AND PRECLINICAL TOXICITY AS A NOVEL TREATMENT FOR NON-MUSCLE INVASIVE BLADDER CANCER", JOURNAL OF UROLOGY, vol. 191, no. 4, 1 April 2014 (2014-04-01) - 1 April 2014 (2014-04-01), XP028602675, ISSN: 0022-5347, DOI: 10.1016/J.JURO.2014.02.1323 *
ROBINS DJ ET AL: "Long-term survival outcomes with intravesical nanoparticle albumin-bound paclitaxel for recurrent non-muscle-invasive bladder cancer after previous bacillus Calmette-Guérin therapy", UROLOGY, BELLE MEAD, NJ, US, vol. 103, 1 May 2017 (2017-05-01), pages 149 - 153, XP009532482, ISSN: 0090-4295, DOI: 10.1016/J.UROLOGY.2017.01.018 *
SAMBROOK ET AL.: "Molecular Cloning. A laboratory manual", 1989, COLD SPRING HARBOR LABORATORY PRESS, pages: 101 - 104
YATES ET AL., EUR UROL, vol. 62, 2012, pages 1088 - 1096

Also Published As

Publication number Publication date
EP4373519A1 (en) 2024-05-29
IL310225A (en) 2024-03-01
AU2022313491A1 (en) 2024-01-18
CA3224671A1 (en) 2023-01-26

Similar Documents

Publication Publication Date Title
JP2021088568A (en) Immunogenic anti-inflammatory compositions
EP2938627B1 (en) Signal peptide fusion partners facilitating listerial expression of antigenic sequences and methods of preparation and use thereof
JP5757863B2 (en) Dual delivery system for xenoantigens
JP2017522027A (en) Recombinant Listeria strain expressing heterologous antigen fusion protein and method of use thereof
TW201726171A (en) Listeria-based immunotherapy and methods of use thereof
US11426453B2 (en) Recombinant mycobacterium as an immunotherapeutic agent for the treatment of cancer
US20210024940A1 (en) Recombinant therapeutic interventions for cancer
CN105816866B (en) Immunogenic anti-inflammatory compositions
WO2023001895A1 (en) Recombinant mycobacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma
US9976149B2 (en) Modified listeria bacteria and uses thereof
EP1882741A1 (en) Recombinant Mycobacterium strain expressing a mycobacterial FAP protein under the control of a promoter active under hypoxia and its application for cancer therapy
US20220160858A1 (en) Recombinant Mycobacterium as an Immunotherapeutic Agent for the Treatment of Cancer
EP4122491A1 (en) Recombinant microbacterium as an immunotherapeutic agent for the second-line therapy of bladder carcinoma
KR20240082332A (en) Recombinant mycobacterium as an immunotherapy agent for secondary treatment of bladder carcinoma.
KR102667859B1 (en) Recombinant mycobacterium as an immunotherapy agent for cancer treatment
US10925945B2 (en) Bacterial vaccines deficient in the 2-C-methyl-D-erythritol-4-phosphate pathway and methods of preparation and use thereof
AU2011284737B8 (en) Immunogenic anti-inflammatory compositions
Xu Modulating Local Immune Environment to Deter Tuberculosis Progression
WO2021163602A1 (en) Recombinant therapeutic interventions for cancer
Qi et al. COVID-19 Vaccine and Bladder Cancer: Friend or Foe?

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22751731

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2022313491

Country of ref document: AU

Ref document number: AU2022313491

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 3224671

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 807088

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 310225

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2022313491

Country of ref document: AU

Date of ref document: 20220720

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2024/000980

Country of ref document: MX

Ref document number: 2401000424

Country of ref document: TH

WWE Wipo information: entry into national phase

Ref document number: P6000154/2024

Country of ref document: AE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112024001159

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 202490292

Country of ref document: EA

Ref document number: 1020247005936

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2022751731

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022751731

Country of ref document: EP

Effective date: 20240222

ENP Entry into the national phase

Ref document number: 112024001159

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20240119