WO2000014257A1 - Fusion receptors specific for prostate-specific membrane antigen and uses thereof - Google Patents

Fusion receptors specific for prostate-specific membrane antigen and uses thereof Download PDF

Info

Publication number
WO2000014257A1
WO2000014257A1 PCT/US1999/020349 US9920349W WO0014257A1 WO 2000014257 A1 WO2000014257 A1 WO 2000014257A1 US 9920349 W US9920349 W US 9920349W WO 0014257 A1 WO0014257 A1 WO 0014257A1
Authority
WO
WIPO (PCT)
Prior art keywords
psma
cells
cytoplasmic domain
fusion
transduced
Prior art date
Application number
PCT/US1999/020349
Other languages
French (fr)
Inventor
Michel Sadelain
Neil H. Bander
Michael Gong
Original Assignee
Sloan-Kettering Institute For Cancer Research
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sloan-Kettering Institute For Cancer Research filed Critical Sloan-Kettering Institute For Cancer Research
Priority to CA002343156A priority Critical patent/CA2343156A1/en
Priority to JP2000568998A priority patent/JP2002524081A/en
Priority to EP99945508A priority patent/EP1109921A4/en
Publication of WO2000014257A1 publication Critical patent/WO2000014257A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/7051T-cell receptor (TcR)-CD3 complex
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70517CD8
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70521CD28, CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/027Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a retrovirus

Definitions

  • This invention relates to fusion receptors for prostate-specific membrane antigen (PSMA), and to uses thereof in the treatment of prostate cancer, other cancers expressing PSMA and tumor neovasculature.
  • PSMA prostate-specific membrane antigen
  • the inventions provides fusion receptors, nucleic acids encoding these fusion receptors, and transduced cells expressing the fusion receptors, as well as methods of using the transduced cells.
  • T cell immunity Destruction of immunological targets requires T lymphocyte recognition via the T cell receptor (TCR) of antigenic peptides presented in the context of major histocompatibility complex (MHC) molecules on antigen- presenting cells (APC).
  • TCR T cell receptor
  • MHC major histocompatibility complex
  • APC antigen- presenting cells
  • T cell costimulation results from an interaction of the T cell surface receptor CD28 with the costimulatory hgand B7, which is p ⁇ maiily expressed on the surface of professional APCs and activated B cells leading to IL-2 secretion and clonal expansion of the activated T cells
  • CD28 costimulatory hgand B7
  • signals transduced by the CD28 receptor determine whether TCR occupancy results in a pioductive immune response or clonal anergy Therefore, one factor accounting for the pooi immunogenicity of MHC-expressmg tumors is that, despite presentation of potentially immunogemc peptides lithe context of MHC molecules, tumors lack the costimulatory molecule B7, and thus fail to elicit a full activation of T cells and therefore an effective anti-tumoi T cell response
  • the introduction of the B7 molecule (CD28 hgand) in tumor cells is one discussed therapy today (melanoma Townsend et al, 1993, Chen et al, 1992
  • T cells can lecogmze and lyse tumor cells provided that they bind to the tumor cells and are appiop ⁇ ately activated T cell activation operates according to the two signal model, which states that lymphocytes require for optimal activation both an antigen-specific signal delivered through the antigen receptor and a second antigen nonspecific or costimulatory signal T cell costimulatory pathways determine whether TCR complex engagement results in functional activation or clonal anergy of CD4 T cells
  • T lymphocytes One means of generating tumor-specific T lymphocytes is then modification by gene transfer of tumor-specific fusion molecules
  • the mtioduction of chime ⁇ c molecules m T cells combining tumor specific single chain variable fragment (scFv) with signal transduction domains of TCR related activation molecules is reported by a number of groups (Eshar et al, Springer Semin. Immunopathol. 18: 199-209 (1993)).
  • scFv tumor specific single chain variable fragment
  • These genetically modified T cells are able to target tumor cells and to destroy them in vitro, but based on the two signal model for T cell activation, the reinfusion of these transduced T lymphocytes is limited by the incomplete activation signal after antigen recognition and clonal expansion in vivo is not successful.
  • antigen dependent IL-2 secretion can be stimulated in vitro in Jurkat cells expressing a chimeric molecule formed from an antigen-specific scFv and the ⁇ -chain of CD3, when the cells are exposed to the antigen in the presence of anti-CD28 or ionomycin as a costimulatory signal.
  • Cells expressing both chimeric molecules displayed responses to either antigen in the presence of appropriate costimulatory molecules. Alvarez- Vallina et al. suggests that these results offer the possibility that addition of antigen-specific CD28 mediated signaling could improve adoptive immunotherapies.
  • T-cell receptors have not been shown to function in human peripheral blood lymphocytes (PBL) and in particular in the T cells of actual cancer patients.
  • PBL peripheral blood lymphocytes
  • the present invention provides a fusion receptor composition which is effective to promote a cellular immune response to a target antigen in vivo when the fusion receptor is expressed by T lymphocytes.
  • the target antigen is prostate-specific membrane antigen (PSMA)
  • PSMA prostate-specific membrane antigen
  • PSMA-scFv optional connector : cytoplasmic domain.
  • the PSMA-scFv in this structure is a single chain antibody cloned from the V region genes of a hybridoma specific for PSMA.
  • the optional connector region is provided to give a spacing between the PSMA-scFv and the cytoplasmic domain, such that both can retain substantial function.
  • a suitable connector is the CD8 hinge, although other connectors of greater or lesser length might be used.
  • the cytoplasmic domain is included to direct the function of the fusion receptor.
  • One exemplary cytoplasmic domain which can be used in the fusion receptor of the invention is a T cell receptor ⁇ -chain cytoplasmic domain.
  • an expression vector encoding the fusion receptor is transduced into primary T lymphocytes obtained from an individual to be treated, for example an expression vector encoding the PSMA-scFv containing fusion receptor is suitably transduced into cells from a human patient who has been diagnosed with prostate cancer.
  • the transduced lymphocytes are returned to the patient where cells expressing the fusion receptor secrete interleukin 2 and proliferate in response to PSMA- positive cells.
  • the resulting cytotoxic lymphocytes specifically lyse cells expressing PSMA and thus can be used to target PSMA-positive tumor cells.
  • NK cells natural killer cells or other immune effector cells allows these cells to target any tissue (including tumor tissue) expressing PSMA.
  • tissue including tumor tissue
  • NK cells can be used to treat prostate cancer, other cancers expressing PSMA and tumor-associated neovasculature.
  • Fig. 1 shows the structure of a retroviral vector Pz-1 including a gene for a PSMA-specific fusion receptor in accordance with the invention
  • Figs. 2 A-E show cytotoxicity of Pz-1 transduced PBL with respect to various target cells
  • Fig. 3 shows the time course for cocultivation of transduced T cells with fibroblasts
  • Fig. 4A shows T cells proliferation in coculture with various types of fibroblast cells
  • Fig. 4B shows cell lysis by T cells after prior exposure to coculture conditions
  • Fig. 5 shows IL-2 production by transduced T cells in coculture with various types of fibroblast cells.
  • the present invention provides fusion receptors which are useful in the generation of a cellular immune response to cells which express PSMA.
  • fusion receptors have the general structure:
  • PSMA-scFv optional connector : cytoplasmic domain. This structure is produced by expression in transduced cells of a DNA sequence encoding the ammo acid sequence of the fusion receptor
  • PSMA-scFv is a single chain antibody cloned from the V region genes of a hybridoma specific for PSMA
  • a suitable hybridoma for this purpose is J591, which is described m Liu et al , Cancer Res 57 3629-3635 (1997), although other hyb ⁇ domas which produce monoclonal antibodies specific to PSMA could also be employed The production of such hyb ⁇ domas has become routine, and the procedure will not be repeated here
  • V H variable region heavy chain
  • V L variable region light chain
  • the cytoplasmic domain portion of the general formula set forth above is selected to enhance the characteristics of the fusion leceptoi for purposes of promoting a cellular immune response to the antigen recognized by the scFv portion of the fusion receptor
  • the cytoplasmic domain is the cytoplasmic domain of a molecule which functions as a transducei of a mammalian immune response in the presence of an MHC- peptide complex or costimulatory factoi
  • Representative, non-limitmg examples of cytoplasmic domains which may be employed m the present invention include the ⁇ -cham cytoplasmic domain, the CD28 cytoplasmic domain (particularly a fragment spanning ammo acids 336 to 663 of CD28 cDNA), 41BB, CD40, ICOS and trance
  • the cytoplasmic domain is the ⁇ -chain derived the TCR complex
  • the fusion receptor of the invention closely mimics a native TCR In this case, it might be expected that binding of an antigen to the scFv
  • the fusion receptors of the invention may include othei cytoplasmic domains
  • CD28 can be used as the cytoplasmic domain to enhance T-cell activation, survival and prohfeiation
  • a preferred CD28 moiety is one which spans ammo acids 336 to 663 of CD28 cDNA, in which case no connector is needed to retain functon PSMA-fusion receptors incorporating 41BB as the cytoplasmic domain ha ⁇ e also been prepared
  • Both the PSMA-CD28 and the PSMA-41 BB fusion receptors have been made and tested m the same experimental model used with the PSMA- ⁇ chain fusion receptor In both cases, sustained proliferation was obseived in both human CD4 and CD8 primary T cells (PBL) m the presence of PSMA ⁇ cells, with moie sustained proliferation being provided by the PSMA-41BB fusion receptor High production of IFN- ⁇ and IL-2 was observed, for PSMA-41BB and PSMA-CD28 transduced, respectively In each of the experiments performed
  • the function of the connector is to act as a spacer so that both the scFv and the cytoplasmic domain can be functionally oriented within the membrane of the transduced cell.
  • One exemplary connector is the CD8 hinge, although other connectors of greater or lesser length could be used. In some cases, such as using the CD28 fragment described herein, no connector is required to permit the molecules to assume the desired orientation.
  • the chimeric fusion receptors are introduced into the individual to be treated (preferably a human) in one of two ways. Gene transfer can be carried out into bone marrow cells, either in vivo or ex vivo, or into immune effector/inflammatory cells such as T- lymphocytes or NK cells. Gene transfer may also be carried out into antigen presenting cells, particularly dendritic cells. In the case of dendritic cells, CD40 and trance are the preferred cytoplasmic domain.
  • a preferred approach to this gene transfer is using retroviral vectors encoding the fusion receptor.
  • a particularly preferred approach utilizes an SFG retroviral vector (Riviere et al., Proc. NatlAcad Sci. (USA) 92: 6733-6737 (1995)) transduced into patient PBL using gibbon ape leukemia virus (GALV) envelope-pseudotyped virions. (Gallardo et al., Blood 90: 952-957 (1997).
  • the PSMA-specific fusion receptor of the present invention is useful in the treatment of prostate cancer.
  • PSMA is also found in the neovasculature of renal cell, urothelial, colon, breast and lung carcinomas, melanomas and some sarcomas
  • the PSMA-specific fusion receptor of the invention has broader applicability.
  • the present application describes a method for treatment of cancers in which the cancer cells or neovasculature are characterized by expression of PSMA, comprising administering to a patient suffering from such a cancer patient-derived lymphocytes which express with a PSMA fusion receptor having the structure
  • PSMA-scFv optional connector : cytoplasmic domain.
  • administration is intended to encompass both in vivo methods, in which the fusion receptor is introduced into the lymphocytes without first removing them from the patient, and ex vivo methods where the patient-derived lymphocytes are obtained from the patient, transduced with the PSMA-specific fusion receptor and then reintroduced to the patient.
  • the transduced lymphocytes are introduced in an amount to provide therapeutic benefit. Where sufficient clonal expansion of the transduced lymphocytes occurs in vivo, a long-term immunity to the tumor cells may be induced after a single administration. If the transduced lymphocytes are less stable, multiple infusions may be required to obtain remission of a particular cancer, and long-term protection may not be achieved. In either case, the determination of the appropriate therapeutic regimen is a matter of routine developed in the course of clinical trials.
  • Example 1 PSMA-scFv was created by cloning the immunoglobulin genes from the J591 hybridoma encoding the variable region of the heavy chain (V H and the variable region of the light chain (V L ).
  • the V H and V L genes were cloned using the technique previously described by Orlandi et al., supra. Briefly, mRNA was isolated from the J591 hybridoma cell line and reverse transcribed into cDNA using a reverse transcriptase polymerase chain reaction (RT- PCR) kit obtained from Pharmacia, Pisacatway, NJ.
  • RT- PCR reverse transcriptase polymerase chain reaction
  • V H gene Sequence analysis of the V H gene confirmed an appropriate open reading frame. However, the V L gene sequence analysis revealed a stop codon in the anticipated reading frame. The sequences of the genes encoding the J591 monoclonal antibody heavy and light chain were compared to the sequences of the cloned products, and several discrepancies were noted between the V L sequences. The major difference was that the primer pair used deleted a nucleotide from the actual sequence, resulting in an open reading frame shift that produced a stop codon. Nucleotide sequence corrections in the V L product were made using corrective primers based on the actual sequences and using these primers in a second PCR amplification utilizing the obtained V, sequence as a template. The corrective primers were: V L backward: GAAGAAGATCJG CATTGTGATGACCAGTCTC CAAATTCATG Seq. ID. No. 5
  • an oligonucleotide encoding the human CD8 leader sequence was cloned to the 5'-end of the V H gene, and the 3'-end of the V,, gene was cloned to an oligonucleotide encoding a (gly-ser 2 ) 5 linker followed by the V L gene, creating the PSMA- specific scFv.
  • the scFv was then cloned to the CD8 hinge and transmembrane domains, followed by the T cell receptor ⁇ -chain cytoplasmic domain to create Pz-1, a PSMA-specific scFv/ ⁇ -chain chimeric T-cell receptor.
  • the Pz-1 fusion gene was then cloned into the SFG retroviral vector (Riviere et al, supra) as illustrated in Fig. 1.
  • Example 2 The SFG retroviral vector containing Pz-1 was transduced into PBL's harvested from five human patients suffering from prostate cancer with a variety of clinical stages using GALV envelope pseudo typed virions as previously described. Gallardo et al., supra. The clinical status of three representative patients is summarized in Table 1.
  • the age is the current age of the patient, the time since dx is the time elapsed between date of diagnosis of prostate cancer and PBL harvest; GG is the Gleason grade of the patient's most recent prostate cancer pathology; stage at dx is the clinical stage at the time of original diagnosis; RRP in the treatment column stands for radical retropubic prostatectomy; current stage is the current clinical or pathological state and current PSA is the most recent serum prostate specific antigen (PSA) level.
  • PSA serum prostate specific antigen
  • the PBL were expanded 4 to 14 days in the presence of interleukin-2 (IL-2).
  • IL-2 interleukin-2
  • Gene transfer efficiency was monitored by FACS analysis using a FITC-conjugated Pz-1 idiotype-specific antiserum. After incubation with the FITC labeled antiserum, the cells were washed incubated with 10% normal mouse serum, and stained with a PE-conjugated anti-CD8 mAb. The gene transfer efficiency observed varied between 20% and 50% in both CD8" and CD4 * cells for Pz-1 and controls.
  • Example 3 Cytotoxic T lymphocyte (CTL) assays were performed on the human prostate cancer cell line LNCaP which abundantly expresses PSMA. In order to confirm that the cytotoxicity was PSMA-specific, PSMA was expressed in PC-3, a PSMA-negative human prostate cancer cell line and EL-4, a murine thymoma cell line. PBL from the three patients of Table 1 were transduced with either Pz-1 or NTP, a mutated human low-affinity nerve growth factor receptor used as a control cell surface marker. (Gallardo et al., supra). Transduction efficiency (%TR) measured as described above, and the fraction of CD8 + and CD56 + cells on the day of the CTL assay are reported in the common legend of Fig. 2.
  • CTL Cytotoxic T lymphocyte
  • E:T effector to target
  • PBL transduced with Pz-1 but not NTP effectively lysed PSMA + targets.
  • Example 4 To further assess the response of Pz-1 transduced primary T cells to PSMA, we investigated whether Pz-1 + PBL could undergo proliferation upon engagement with cell- bound PSMA and sustain thereafter their cytolytic potential.
  • a cocultivation system was established in which transduced T cells were cultured with a layer of irradiated NIH3T3 fibroblasts expressing various combinations of PSMA and B7.1 for four days with periodic sampling to measure levels of IL-2 as illustrated in Fig. 3.
  • FACS cell counts using FITC- conjugated Pz-1 idiotype specific antibody and with either anti-CD4 or anti-CD8 were performed after four days of co-cultivation, and again 4 days later.
  • the transduced T cell count was derived by multiplying the percentage of CD4 + Pz-T or CD8 ⁇ Pz-l ⁇ double positive cells by the number of viable cells.
  • the results are shown in Fig. 4A, where /B7+PSMA, /PSMA and /B7 refer to the molecules expressed by the fibroblast layer.
  • PSMA induced proliferation of Pz-1 transduced T-cells increasing the number of cells 6-8 fold after 4 days, and these transduced cells destroyed PSMA + fibroblast layers within 48 hours, while the PSMA " layers remained intact during the entire 4-day cocultivation. By day 8, however, the absolute number of Pz-1 + cells dropped to 2-3 fold above initial levels.
  • B7.1 (CD80) was transduced into PSMA + and PSMA " fibroblasts.
  • Example 5 To test whether cytotoxic T cells retain their cytotoxic potential after restimulation with antigen, T cells were harvested 12-17 days after the start of coculture with fibroblast monolayers expressing PSMA and B7.1 and retested in the CTL assay. As shown in Fig. 5, expanded Pz-1 transduced T cells remained fully capable of lysing PSMA " target cells. Furthermore, the expanded Pz-1 + cells were capable of a second round of proliferation and IL-2 and IFN- ⁇ secretion when reexposed to PSMA + B7.1 + fibroblasts.
  • T cells bearing artificial TCRs the prospect of apoptotic cell death or anergy upon restimulation could be compounded by partial T cell activation if chimeric receptors fail to adequately recruit downstream signaling molecules. Faulty T cell activation could result in the induction of immune tolerance and the neutralization of the infused effector T cells. Such phenomenon could in part explain in vivo findings obtained with T cells expressing an ErbB- 2 specific- ⁇ chain fusion receptor, required repeated high dose intra-tumoral administration to effectively eliminate established tumors. Alternschmidt et al, J. Immunol. 159: 5509-5515 (1997).
  • Example 2 In order to prepare a fusion receptor in which the cytoplasmic domain is derived from CD-28, the procedure of Example 1 was repeated only using a CD28 fragment in place of the ⁇ -chain segment. The CD28 cDNA fragment was obtained as follow.
  • a segment of the human CD28 cDNA that encodes part of the extracellular, the transmembrane, and the cytoplasmic domains was amplified by PCR from the plasmid pbsCD28, using the upstream primer
  • primers contain Notl and BamHI sites respectively for the insertion of the PCR product in the retroviral Vector SFG.
  • the CD28 fragment was ligated into the Notl an BamHI sites of the retroviral vector SFG, containing the CD8 ⁇ leader sequence, followed by the single chain gene, encoding the V H and V L domains of the PSMA-specific antibody

Abstract

A fusion receptor composition which is effective to promote a cellular immune response to prostate-specific membrane antigen (PSMA) in vivo when the fusion receptors is expressed by T lymphocytes has the structure: PSMA-scFv: connector: cytoplasmic domain. The PSMA-scFv in this structure is a single chain antibody cloned from the V region genes of a hybridoma specific for PSMA. The connector region is provided to give a spacing between the OSMA-scFv and the cytoplasmic domain, such that both can retain substantial function. A suitable connector is the CD8 hinge, although other connectors of greater or lesser length might be used. The cytoplasmic domain is included to direct the function of the fusion receptor. One exemplary cytoplasmic domain which can be used in the fusion receptor of the invention is a T cell receptor z-chain cytoplasmic domain. An expression vector encoding the fusion receptor is transduced into primary T lymphocytes obtained from an individual to be treated. The transduced lymphocytes are returned to the patient where cells expressing the fusion receptor secrete interleukin 2 and proliferate in response to PSMA-positive cells. The resulting cytotoxic lymphocytes specifically lyse cells expressing PSMA and thus can be used to target PSMA-positive tumor cells and neovasculature.

Description

FUSION RECEPTORS SPECIFIC FOR PROSTATE-SPECIFIC MEMBRANE ANTIGEN AND USES THEREOF
This application claims priority from US Provisional Application No. 60/099,138, filed September 4, 1998, which for purposes of the United States and countries allowing such incorporation, is incorporated herein by reference.
Field of the Invention
This invention relates to fusion receptors for prostate-specific membrane antigen (PSMA), and to uses thereof in the treatment of prostate cancer, other cancers expressing PSMA and tumor neovasculature. The inventions provides fusion receptors, nucleic acids encoding these fusion receptors, and transduced cells expressing the fusion receptors, as well as methods of using the transduced cells.
Background of the Invention
A long-standing goal of cancer research has been to stimulate the immunological rejection of tumors. This goal is based on the hypothesis that many tumors express foreign or mutated forms of antigens that can potentially serve as targets for their destruction by the immune system. Cellular immunity plays the key role in this rejection, with both T helper cells and cytolytic T lymphocytes (CTLs) being involved (Greenberg, Adv. Immunol. 49: 281-355 (1991).
There are several reasons why even those tumors that express rejection antigens can evade destruction by T cell immunity. Destruction of immunological targets requires T lymphocyte recognition via the T cell receptor (TCR) of antigenic peptides presented in the context of major histocompatibility complex (MHC) molecules on antigen- presenting cells (APC). Some tumors fail adequately to process and present antigens to T cells because of reduced expression of MHC class I molecules.
Many strategies have been devised to render tumor cells more immunogenic. One is based on the genetic engineering of tumor cells to stimulate the generation of tumor- specific effector T cells in vivo. This has been investigated by direct MHC class I gene transfection to enhance expression of MHC by introduction of the γ-interferon cDNA to upregulate endogenous class I antigens or by tranfectmg tumor cells with cytokmes with the hope that interleukin paracrine secretion of lymphokmes can substitute for T cell help, induce tumor specific cytotoxic T lymphocytes, and cause tumor rejection
The molecular basis of T cell costimulation results from an interaction of the T cell surface receptor CD28 with the costimulatory hgand B7, which is pπmaiily expressed on the surface of professional APCs and activated B cells leading to IL-2 secretion and clonal expansion of the activated T cells In vitro and in vivo studies showed that signals transduced by the CD28 receptor determine whether TCR occupancy results in a pioductive immune response or clonal anergy Therefore, one factor accounting for the pooi immunogenicity of MHC-expressmg tumors is that, despite presentation of potentially immunogemc peptides lithe context of MHC molecules, tumors lack the costimulatory molecule B7, and thus fail to elicit a full activation of T cells and therefore an effective anti-tumoi T cell response Thus, the introduction of the B7 molecule (CD28 hgand) in tumor cells is one discussed therapy today (melanoma Townsend et al, 1993, Chen et al, 1992 colon carcinoma Townsend et al, 1994) to provide protective immunity by autologous CD8 T cells which leads to a potent rejection of modified and unmodified tumor cells in vι\ o CD4 and CD8 immunity induced by immunization with class II+B7-1 -transfected sarcoma cells aic also widely discussed immunotherapy strategies
Another approach is based on the manipulation of the effector cells, I e , T lymphocytes, rather than the antigen-presentmg cells or tumoi cells T cells can lecogmze and lyse tumor cells provided that they bind to the tumor cells and are appiopπately activated T cell activation operates according to the two signal model, which states that lymphocytes require for optimal activation both an antigen-specific signal delivered through the antigen receptor and a second antigen nonspecific or costimulatory signal T cell costimulatory pathways determine whether TCR complex engagement results in functional activation or clonal anergy of CD4 T cells
One means of generating tumor-specific T lymphocytes is then modification by gene transfer of tumor-specific fusion molecules The mtioduction of chimeπc molecules m T cells combining tumor specific single chain variable fragment (scFv) with signal transduction domains of TCR related activation molecules is reported by a number of groups (Eshar et al, Springer Semin. Immunopathol. 18: 199-209 (1993)). These genetically modified T cells are able to target tumor cells and to destroy them in vitro, but based on the two signal model for T cell activation, the reinfusion of these transduced T lymphocytes is limited by the incomplete activation signal after antigen recognition and clonal expansion in vivo is not successful.
Alvarez-Vallina et al., Eur. J. Immunol. 26: 2304-2309 (1996) have shown that antigen dependent IL-2 secretion can be stimulated in vitro in Jurkat cells expressing a chimeric molecule formed from an antigen-specific single chain antibody variable fragment (scFv) and a truncated CD28 (amino acids 124-202; transmembrane and cytoplasmic domains), when the cells are exposed to the antigen in the presence of anti-CD3 or ionomycin as a costimulatory signal. Similarly, antigen dependent IL-2 secretion can be stimulated in vitro in Jurkat cells expressing a chimeric molecule formed from an antigen-specific scFv and the ζ-chain of CD3, when the cells are exposed to the antigen in the presence of anti-CD28 or ionomycin as a costimulatory signal. Cells expressing both chimeric molecules displayed responses to either antigen in the presence of appropriate costimulatory molecules. Alvarez- Vallina et al. suggests that these results offer the possibility that addition of antigen-specific CD28 mediated signaling could improve adoptive immunotherapies.
It is a substantial step, however, from such in vitro results to therapeutic efficacy in vivo. Importantly, while IL-2 secretion is suggestive of T cell activation, it can frequently be followed by T cell anergy or apoptosis (presumably as a result of inadequate costimulation) which results in the death of the T cells rather than the development of an immune response. There is no assurance that signaling species will be present in vivo to fulfill the role of the external costimulatory signals supplied by Alvarez-Vallina et al. in their in vitro experiments, or that these signals will result in the level of clonal expansion which is required to mount and maintain a therapeutically meaningful cytotoxic T cell response. Furthermore, artificial T-cell receptors have not been shown to function in human peripheral blood lymphocytes (PBL) and in particular in the T cells of actual cancer patients. The applicability of findings in normal T cells or carefully selected leukemic cells lines to T cells from cancer patients cannot be taken for granted given the signaling defects frequently observed in cancer patients and chronically tumor-bearing mice. Mizoguchi et al., Science 258: 1795-1798 (1992); Ochoa et al, in Important Advances in Oncology, J.B. Lippincott Co., Philadelphia (1995); Zier et al., Hum. Gene Ther. 6: 1259-1264 (1995). Some of these defects, which include ζ-chain, lck and ZAP-80 abnormalities, could limit the function of artificial TCRs in cancer patient T cells. Thus, there remains a need for a method for sustaining the formation of tumor-specific T lymphocytes which can be successfully reintroduced into a host organism, preferably a human being, for the in vivo generation of a cellular immune response to the tumor.
It is an object of the present invention to provide such a method, and to provide fusion proteins, and nucleic acid constructs encoding such fusion proteins which can be used in such a method.
It is a further object to provide such methods and fusion proteins specific for prostate-specific membrane antigen.
Summary of the Invention
The present invention provides a fusion receptor composition which is effective to promote a cellular immune response to a target antigen in vivo when the fusion receptor is expressed by T lymphocytes. By way of example, when the target antigen is prostate-specific membrane antigen (PSMA), the fusion receptor has the structure:
PSMA-scFv : optional connector : cytoplasmic domain. The PSMA-scFv in this structure is a single chain antibody cloned from the V region genes of a hybridoma specific for PSMA. The optional connector region is provided to give a spacing between the PSMA-scFv and the cytoplasmic domain, such that both can retain substantial function. In cases where a connector is needed, a suitable connector is the CD8 hinge, although other connectors of greater or lesser length might be used. The cytoplasmic domain is included to direct the function of the fusion receptor. One exemplary cytoplasmic domain which can be used in the fusion receptor of the invention is a T cell receptor ζ-chain cytoplasmic domain.
In accordance with the method of the invention, an expression vector encoding the fusion receptor is transduced into primary T lymphocytes obtained from an individual to be treated, for example an expression vector encoding the PSMA-scFv containing fusion receptor is suitably transduced into cells from a human patient who has been diagnosed with prostate cancer. The transduced lymphocytes are returned to the patient where cells expressing the fusion receptor secrete interleukin 2 and proliferate in response to PSMA- positive cells. The resulting cytotoxic lymphocytes specifically lyse cells expressing PSMA and thus can be used to target PSMA-positive tumor cells. Expression of the fusion receptor in CD4+ or CD8+ T-cells, natural killer (NK) cells or other immune effector cells allows these cells to target any tissue (including tumor tissue) expressing PSMA. Thus, such cells can be used to treat prostate cancer, other cancers expressing PSMA and tumor-associated neovasculature.
Brief Description of the Drawings
Fig. 1 shows the structure of a retroviral vector Pz-1 including a gene for a PSMA-specific fusion receptor in accordance with the invention;
Figs. 2 A-E show cytotoxicity of Pz-1 transduced PBL with respect to various target cells;
Fig. 3 shows the time course for cocultivation of transduced T cells with fibroblasts;
Fig. 4A shows T cells proliferation in coculture with various types of fibroblast cells;
Fig. 4B shows cell lysis by T cells after prior exposure to coculture conditions; and
Fig. 5 shows IL-2 production by transduced T cells in coculture with various types of fibroblast cells.
Detailed Description of the Invention
In a first aspect, the present invention provides fusion receptors which are useful in the generation of a cellular immune response to cells which express PSMA. Such fusion receptors have the general structure:
PSMA-scFv : optional connector : cytoplasmic domain. This structure is produced by expression in transduced cells of a DNA sequence encoding the ammo acid sequence of the fusion receptor
In the general formula set forth above, PSMA-scFv is a single chain antibody cloned from the V region genes of a hybridoma specific for PSMA A suitable hybridoma for this purpose is J591, which is described m Liu et al , Cancer Res 57 3629-3635 (1997), although other hybπdomas which produce monoclonal antibodies specific to PSMA could also be employed The production of such hybπdomas has become routine, and the procedure will not be repeated here
A technique which can be used for cloning the variable region heavy chain (VH) and variable region light chain (VL) has been described m Orlandi et al , Proc Natl Acad Sci (USA) 86 3833-3837 (1989) Briefly, mRNA is isolated from the hybridoma cell line, and reverse transcribed into complementary DNA (cDNA), for example using a reverse transcπptase polymerase chain reaction (RT-PCR) kit Sequence-specific primers corresponding to the sequence of the VH and VL genes weie used, for example Seq ID Nos 1-4 Sequence analysis of the cloned products and comparison to the known foi the V„ and VL genes showed that the cloned VH gene matched expectations For the VL gene, however, the cloned sequence did not match expectations, and contained a stop codon in the anticipated open reading frame To correct this, corrective primers (Seq ID Nos 5 and 6), containing inserted bases to correct the differences between the clone and the known sequence were used to amplify the cloned VL gene to produce polynucleotides encoding the V, gene
The cytoplasmic domain portion of the general formula set forth above is selected to enhance the characteristics of the fusion leceptoi for purposes of promoting a cellular immune response to the antigen recognized by the scFv portion of the fusion receptor In general, the cytoplasmic domain is the cytoplasmic domain of a molecule which functions as a transducei of a mammalian immune response in the presence of an MHC- peptide complex or costimulatory factoi Representative, non-limitmg examples of cytoplasmic domains which may be employed m the present invention include the ζ-cham cytoplasmic domain, the CD28 cytoplasmic domain (particularly a fragment spanning ammo acids 336 to 663 of CD28 cDNA), 41BB, CD40, ICOS and trance When the cytoplasmic domain is the ζ-chain derived the TCR complex, the fusion receptor of the invention closely mimics a native TCR In this case, it might be expected that binding of an antigen to the scFv portion of the fusion receptor would result in changes to the ζ-chain which would trigger an intracellular phosphorylation cascade comparable to that observed when an antigen bound to an APC interacts with a native TCR As noted above, however, this signal transduction is not sufficient to produce a complete cellular immune response, and a secondary signal must me provided to avoid anergy and premature cell death through apoptosis In the art, this secondary signal was provided by a signaling antibody unsuited for use in vivo The present invention provides foi the first time the understanding that cells expressing PSMA and a costimulatory signal such as B7 1 provide sufficient secondary signal to maintain a stable population of PSMA-targetmg T cells when the cells are transduced with the fusion protein of the invention
Instead to the ζ-chain, the fusion receptors of the invention may include othei cytoplasmic domains For example, CD28 can be used as the cytoplasmic domain to enhance T-cell activation, survival and prohfeiation A preferred CD28 moiety is one which spans ammo acids 336 to 663 of CD28 cDNA, in which case no connector is needed to retain functon PSMA-fusion receptors incorporating 41BB as the cytoplasmic domain ha\ e also been prepared Both the PSMA-CD28 and the PSMA-41 BB fusion receptors have been made and tested m the same experimental model used with the PSMA-ζ chain fusion receptor In both cases, sustained proliferation was obseived in both human CD4 and CD8 primary T cells (PBL) m the presence of PSMA^ cells, with moie sustained proliferation being provided by the PSMA-41BB fusion receptor High production of IFN-γ and IL-2 was observed, for PSMA-41BB and PSMA-CD28 transduced, respectively In each of the experiments performed, an external signal to complement the signaling of the fusion receptoi was used However, transfection of the PBL with fusion receptors encoding both the ζ-cham and eithei CD28 or 41BB oi a comparable costimulatory molecule would eliminate this requirement Thus, foi example, PBL transduced with both the PSMA-ζ chain fusion recpetor and either a PSMA-fusion receptor with a secondary signaling moiety would provide therapeutic efficacy for in vivo use Between the scFv and the cytoplasmic domain may be a connector. The function of the connector is to act as a spacer so that both the scFv and the cytoplasmic domain can be functionally oriented within the membrane of the transduced cell. One exemplary connector is the CD8 hinge, although other connectors of greater or lesser length could be used. In some cases, such as using the CD28 fragment described herein, no connector is required to permit the molecules to assume the desired orientation.
The chimeric fusion receptors are introduced into the individual to be treated (preferably a human) in one of two ways. Gene transfer can be carried out into bone marrow cells, either in vivo or ex vivo, or into immune effector/inflammatory cells such as T- lymphocytes or NK cells. Gene transfer may also be carried out into antigen presenting cells, particularly dendritic cells. In the case of dendritic cells, CD40 and trance are the preferred cytoplasmic domain.
A preferred approach to this gene transfer is using retroviral vectors encoding the fusion receptor. A particularly preferred approach utilizes an SFG retroviral vector (Riviere et al., Proc. NatlAcad Sci. (USA) 92: 6733-6737 (1995)) transduced into patient PBL using gibbon ape leukemia virus (GALV) envelope-pseudotyped virions. (Gallardo et al., Blood 90: 952-957 (1997).
The PSMA-specific fusion receptor of the present invention is useful in the treatment of prostate cancer. In addition, because PSMA is also found in the neovasculature of renal cell, urothelial, colon, breast and lung carcinomas, melanomas and some sarcomas, the PSMA-specific fusion receptor of the invention has broader applicability. Thus, in one aspect of the invention, the present application describes a method for treatment of cancers in which the cancer cells or neovasculature are characterized by expression of PSMA, comprising administering to a patient suffering from such a cancer patient-derived lymphocytes which express with a PSMA fusion receptor having the structure
PSMA-scFv : optional connector : cytoplasmic domain. As used herein, the concept of "administration" is intended to encompass both in vivo methods, in which the fusion receptor is introduced into the lymphocytes without first removing them from the patient, and ex vivo methods where the patient-derived lymphocytes are obtained from the patient, transduced with the PSMA-specific fusion receptor and then reintroduced to the patient.
The transduced lymphocytes are introduced in an amount to provide therapeutic benefit. Where sufficient clonal expansion of the transduced lymphocytes occurs in vivo, a long-term immunity to the tumor cells may be induced after a single administration. If the transduced lymphocytes are less stable, multiple infusions may be required to obtain remission of a particular cancer, and long-term protection may not be achieved. In either case, the determination of the appropriate therapeutic regimen is a matter of routine developed in the course of clinical trials.
The invention will now be further described and illustrated with reference to the following non-limiting examples.
Example 1 PSMA-scFv was created by cloning the immunoglobulin genes from the J591 hybridoma encoding the variable region of the heavy chain (VH and the variable region of the light chain (VL). The VH and VL genes were cloned using the technique previously described by Orlandi et al., supra. Briefly, mRNA was isolated from the J591 hybridoma cell line and reverse transcribed into cDNA using a reverse transcriptase polymerase chain reaction (RT- PCR) kit obtained from Pharmacia, Pisacatway, NJ. The VH and VL genes were cloned from the cDNA using the following degenerate primers:
VH backward:
AGGTSMARCTGCAGSAGTCWGG Seq. ID. No. 1
VH forward:
TGAGGAGACGGTGACCGTGGTCCCTTGGCCCCAG Seq. ID. No. 2
VL backward:
GACATTGAGCTCACCCAGTCTCCA Seq. ID. No. 3 VL forward:
TGCGGCCGCCCGTTTGATCTCCAGCTTGGTCCC Seq. ID. No. 4
Sequence analysis of the VH gene confirmed an appropriate open reading frame. However, the VL gene sequence analysis revealed a stop codon in the anticipated reading frame. The sequences of the genes encoding the J591 monoclonal antibody heavy and light chain were compared to the sequences of the cloned products, and several discrepancies were noted between the VL sequences. The major difference was that the primer pair used deleted a nucleotide from the actual sequence, resulting in an open reading frame shift that produced a stop codon. Nucleotide sequence corrections in the VL product were made using corrective primers based on the actual sequences and using these primers in a second PCR amplification utilizing the obtained V, sequence as a template. The corrective primers were: VL backward: GAAGAAGATCJG CATTGTGATGACCAGTCTC CAAATTCATG Seq. ID. No. 5
VL forward:
TGCGGCCGCCCGTTTCAG(7TCG4GCATGGTCCCAGCACCG Seq. ID. No. 6
wherein the bold and italicized letters indicate additions/substitutions to correct the obtained VL sequence to the actual J591 VL sequence.
Next, an oligonucleotide encoding the human CD8 leader sequence was cloned to the 5'-end of the VH gene, and the 3'-end of the V,, gene was cloned to an oligonucleotide encoding a (gly-ser2)5 linker followed by the VL gene, creating the PSMA- specific scFv. The scFv was then cloned to the CD8 hinge and transmembrane domains, followed by the T cell receptor ζ-chain cytoplasmic domain to create Pz-1, a PSMA-specific scFv/ζ-chain chimeric T-cell receptor. The Pz-1 fusion gene was then cloned into the SFG retroviral vector (Riviere et al, supra) as illustrated in Fig. 1.
Example 2 The SFG retroviral vector containing Pz-1 was transduced into PBL's harvested from five human patients suffering from prostate cancer with a variety of clinical stages using GALV envelope pseudo typed virions as previously described. Gallardo et al., supra. The clinical status of three representative patients is summarized in Table 1.
Figure imgf000013_0001
In Table 1, the age is the current age of the patient, the time since dx is the time elapsed between date of diagnosis of prostate cancer and PBL harvest; GG is the Gleason grade of the patient's most recent prostate cancer pathology; stage at dx is the clinical stage at the time of original diagnosis; RRP in the treatment column stands for radical retropubic prostatectomy; current stage is the current clinical or pathological state and current PSA is the most recent serum prostate specific antigen (PSA) level. Normal PSA is in the range of 0-4 ng/ml. A rising PSA following RRP is biochemical evidence of residual prostate cancer, although the site of the residual disease is unknown.
After retroviral infection, the PBL were expanded 4 to 14 days in the presence of interleukin-2 (IL-2). Gene transfer efficiency was monitored by FACS analysis using a FITC-conjugated Pz-1 idiotype-specific antiserum. After incubation with the FITC labeled antiserum, the cells were washed incubated with 10% normal mouse serum, and stained with a PE-conjugated anti-CD8 mAb. The gene transfer efficiency observed varied between 20% and 50% in both CD8" and CD4* cells for Pz-1 and controls.
Example 3 Cytotoxic T lymphocyte (CTL) assays were performed on the human prostate cancer cell line LNCaP which abundantly expresses PSMA. In order to confirm that the cytotoxicity was PSMA-specific, PSMA was expressed in PC-3, a PSMA-negative human prostate cancer cell line and EL-4, a murine thymoma cell line. PBL from the three patients of Table 1 were transduced with either Pz-1 or NTP, a mutated human low-affinity nerve growth factor receptor used as a control cell surface marker. (Gallardo et al., supra). Transduction efficiency (%TR) measured as described above, and the fraction of CD8+ and CD56+ cells on the day of the CTL assay are reported in the common legend of Fig. 2. The cultured PBL, harvested 4 to 14 days after transduction, were incubated for 4 hours at different effector to target (E:T) rations with 104 51Cr-labeled target cells. Percent specific lysis was determined for PSMA+ LNCaP cells(Fig. 2A), PC3 cells transduced with PSMA (Fig. 2B), wild-type, PSMA"- PC3 cells (Fig. 2C); EL4 cells transduced with PSMA (Fig. 2D) and wild-type PSMA - EL4 cells (Fig. 2E). As shown, for all patients, PBL transduced with Pz-1 but not NTP effectively lysed PSMA+ targets. The same results were obtained using Pz-1 transduced PBL from two other patients with advanced, hormone refractory prostate cancer. The level of cytolytic activity did not closely correlate with the transduction efficiency achieve din each case. Background was variable on human target cells and uniformly low on murine cells. T cells transduced with an irrelevant scFv-ζ chain fusion receptor did not lyse target cells expressing PSMA above background levels. Thus, elevated PSMA-specific cytotoxicity was obtained in Pz-1 transduced T cells derived from five out of five prostate cancer patients tested, independent of their age or clinical stage of disease.
Example 4 To further assess the response of Pz-1 transduced primary T cells to PSMA, we investigated whether Pz-1+ PBL could undergo proliferation upon engagement with cell- bound PSMA and sustain thereafter their cytolytic potential. A cocultivation system was established in which transduced T cells were cultured with a layer of irradiated NIH3T3 fibroblasts expressing various combinations of PSMA and B7.1 for four days with periodic sampling to measure levels of IL-2 as illustrated in Fig. 3. FACS cell counts using FITC- conjugated Pz-1 idiotype specific antibody and with either anti-CD4 or anti-CD8 were performed after four days of co-cultivation, and again 4 days later. The transduced T cell count was derived by multiplying the percentage of CD4+Pz-T or CD8÷Pz-lΦ double positive cells by the number of viable cells. The results are shown in Fig. 4A, where /B7+PSMA, /PSMA and /B7 refer to the molecules expressed by the fibroblast layer.
PSMA induced proliferation of Pz-1 transduced T-cells, increasing the number of cells 6-8 fold after 4 days, and these transduced cells destroyed PSMA+ fibroblast layers within 48 hours, while the PSMA" layers remained intact during the entire 4-day cocultivation. By day 8, however, the absolute number of Pz-1 + cells dropped to 2-3 fold above initial levels. To address whether costimulation could amplify proliferation, B7.1 (CD80) was transduced into PSMA+ and PSMA" fibroblasts. After 8 days of cocultivation, Pz-1+ T cell counts were 5-8 fold higher in cultures with PSMA+ B7.1 fibroblasts than with PSMA+B7.1" fibroblasts, and 25-36 fold higher than in cultures with PSMA" B7.T fibroblasts, suggesting that Pz-1 signaling synergized with the B7.1 mediated costimulation. Fibroblasts expressing PSMA± B7.1 did no induce proliferation of T cells transduced with an irrelevant scFv-ζ chain fusion receptor.
Supernatants of the cocultured T cells were sampled after 24, 48 and 72 hours and assayed for IL-2 and interferon-γ (IFN-γ) secretion. Pz-1 transduced cells released significant amounts of IL2 (Fig. 4B) and IFN-γ 24 hours after exposure to PSMA+ fibroblasts. IL-2 was barely detectable in the supernatants of Pz-1 + cells cocultured with NIH3T3 fibroblasts expressing only B7.1, or in the absence of any feeder layer, nor was it detectable in any coculture condition with T cells transduced with a control irrelevant scFv-ζ chain fusion receptor. However, IL-2 and IFN-γ production was strongly potentiated in the presence of PSMA and B7.1, resulting in 10-fold higher IL-2 release after 24 hours (Fig. 4B). This confirms that B7.1 -mediated signaling provided functional costimulation in Pz-1 transduced cells.
Example 5 To test whether cytotoxic T cells retain their cytotoxic potential after restimulation with antigen, T cells were harvested 12-17 days after the start of coculture with fibroblast monolayers expressing PSMA and B7.1 and retested in the CTL assay. As shown in Fig. 5, expanded Pz-1 transduced T cells remained fully capable of lysing PSMA" target cells. Furthermore, the expanded Pz-1+ cells were capable of a second round of proliferation and IL-2 and IFN-γ secretion when reexposed to PSMA+B7.1+ fibroblasts. These findings indicate that at least a subset of the T cells transduced with the Pz-1 fusion receptor do no fatally undergo activation-induced cell death (AICD) or anergy, as they remain capable of proliferating and killing after contact with antigen. However, in the absence of costimulation, their proliferative potential appears limited.
These results are of critical importance to the utility of the invention in therapeutic applications. The acquisition of specific tumoricidal properties by autologous T cell is of only marginal importance if these cells are not able to carry out more than one cytotoxic hit. Indeed, physiological TCR engagement with peptide-MHC complexes result s in T cell activation which, in the absence of costimulation does not result in complete activation and can eventually lead to anergy or apoptosis. This is especially the case in previously activated cells, such as T cells, cultured for the purpose of clonal expansion or retroviral transduction. Restimulation of antigen-specific CD4+ clones following their adoptive transfer in tumor-bearing animals has been suggested to cause clonal anergy.. In the case of T cells bearing artificial TCRs, the prospect of apoptotic cell death or anergy upon restimulation could be compounded by partial T cell activation if chimeric receptors fail to adequately recruit downstream signaling molecules. Faulty T cell activation could result in the induction of immune tolerance and the neutralization of the infused effector T cells. Such phenomenon could in part explain in vivo findings obtained with T cells expressing an ErbB- 2 specific-ζ chain fusion receptor, required repeated high dose intra-tumoral administration to effectively eliminate established tumors. Alternschmidt et al, J. Immunol. 159: 5509-5515 (1997). The proliferation and cytokine release induced by PSMA" murine fibroblasts together with the cytotoxicity achieved against PSMA+ murine EL4 cells suggest that the Pz- 1 fusion receptor alone provides a sufficient signal to trigger T cell activation. However, although TCR signaling alone can induce lymphocyte proliferation, complete and more sustained activation generally requires that a costimulatory signal be provided in conjunction with TCR stimulation. The results of examples 1-5 show that Pz-1 receptor function is enhanced by costimulation and that costimulation can be provided by B7.1 expression on PSMA+ target cells. Example 6
In order to prepare a fusion receptor in which the cytoplasmic domain is derived from CD-28, the procedure of Example 1 was repeated only using a CD28 fragment in place of the ζ-chain segment. The CD28 cDNA fragment was obtained as follow.
A segment of the human CD28 cDNA that encodes part of the extracellular, the transmembrane, and the cytoplasmic domains (amino acids 336 to 663) was amplified by PCR from the plasmid pbsCD28, using the upstream primer
5'GCGGCCGCAATTGAAGTTATGTATCCT SEQ ID No. 7 and the downstream primer
5'TCGAGGATCTTGTCAGGAGCGATAGGCTGC SEQ ID No. 8.
These primers contain Notl and BamHI sites respectively for the insertion of the PCR product in the retroviral Vector SFG. Following digestion of the purified PCR product with Notl and BamHI, the CD28 fragment was ligated into the Notl an BamHI sites of the retroviral vector SFG, containing the CD8α leader sequence, followed by the single chain gene, encoding the VH and VL domains of the PSMA-specific antibody

Claims

1. A fusion receptor composition having the structure: PSMA-scFv : optional connector : cytoplasmic domain, wherein PSMA-scFv represents a single chain antibody cloned from the V region genes of a hybridoma specific for prostate-specific membrane antigen, the cytoplasmic domain is the cytoplasmic domain of a molecule which functions as a transducer of a mammalian immune response in the presence of a costimulatory factor, and the connector is a region of one or more amino acids disposed between the PSMA-scFv and the cytoplasmic domain, said connector to be of sufficient length to allow both the PSMA-scFv and the cytoplasmic domain to retain function, whereby the fusion receptor is effective when expressed in a T-cell to promote a cellular immune response to prostate-specific membrane antigen.
2. The fusion receptor of claim 1, wherein the cytoplasmic domain comprises a ╬╢-chain of CD3.
3. The fusion receptor of claim 1, wherein the cytoplasmic domain is derived from CD28.
4. The fusion receptor of claim 3, wherein the cytoplasmic domain is a portion of CD28 cDNA spanning amino acids 336-663.
5. The fusion receptor of claim 1, wherein the cytoplasmic domain is derived from 41-BB.
6. The fusion receptor of any of claims 1 to 5, wherein the connector is a CD8 hinge.
7. A method for treating a patient suffering from cancer, wherein the cells of the cancer or neovasculature associated with the cancer express prostate-specific membrane antigen, comprising the steps of:
(a) preparing an expression vector comprising an expressible polynucleotide molecule encoding a fusion protein in accordance with any of claims 1 to 5;
(b) transducing the expression vector into peripheral blood lymphocytes obtained from the patient to obtain transduced lymphocytes expressing the fusion protein; and
(c) reintroducing the transduced lymphocytes into the patient, whereby said transduced lymphocytes respond to antigen on the surface of the cells of the cancer to generate a cytolytic immune response to the cells of the cancer.
8. The method of claim 7, wherein the expression vector is transduced into the peripheral blood lymphocytes in an ex vivo process.
9. The method of claim 7, wherein the expression vector is an SFG vector.
10. The method of claim 9, wherein the expression vector is transduced into patient PBL using gibbon ape leukemia virus envelope-pseudotyped virions.
11. The method of claim 8, wherein the expression vector is transduced into patient PBL using gibbon ape leukemia virus envelope-pseudotyped virions.
12. Peripheral blood lymphocytes transduced with and expressing a fusion receptor in accordance with any of claims 1 to 5.
13. An expression vector comprising a polynucleotide sequence encoding a fusion receptor in accordance with any of claims 1 to 5 and control sequences effective to promote expression of the fusion receptor in mammalian lymphocytes.
14. The vector of claiml3, wherein the expression vector is an SFG vector.
15. The vector of claim 14, wherein the expression vector is packaged in gibbon ape leukemia virus envelope-pseudotyped virions.
16. The vector of claim 13, wherein the expression vector is packaged in gibbon ape leukemia virus envelope-pseudotyped virions.
PCT/US1999/020349 1998-09-04 1999-09-03 Fusion receptors specific for prostate-specific membrane antigen and uses thereof WO2000014257A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CA002343156A CA2343156A1 (en) 1998-09-04 1999-09-03 Fusion receptors specific for prostate-specific membrane antigen and uses thereof
JP2000568998A JP2002524081A (en) 1998-09-04 1999-09-03 Fusion receptor specific for prostate-specific membrane antigen and uses thereof
EP99945508A EP1109921A4 (en) 1998-09-04 1999-09-03 Fusion receptors specific for prostate-specific membrane antigen and uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US9913898P 1998-09-04 1998-09-04
US60/099,138 1998-09-04

Publications (1)

Publication Number Publication Date
WO2000014257A1 true WO2000014257A1 (en) 2000-03-16

Family

ID=22273019

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/020349 WO2000014257A1 (en) 1998-09-04 1999-09-03 Fusion receptors specific for prostate-specific membrane antigen and uses thereof

Country Status (4)

Country Link
EP (1) EP1109921A4 (en)
JP (1) JP2002524081A (en)
CA (1) CA2343156A1 (en)
WO (1) WO2000014257A1 (en)

Cited By (220)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002040059A2 (en) * 2000-11-01 2002-05-23 American Foundation For Biological Research, Inc. Methods and compositions for inducing cell-mediated immune responses
KR100388942B1 (en) * 2000-07-08 2003-06-25 한화석유화학 주식회사 A Single Chain Variable Fragment of an Antibody for Protein Expressed on Cell Surface of Human Cortical Thymocyte and Leukemic Cell
JP2004505117A (en) * 2000-08-07 2004-02-19 ペプリン リサーチ プロプライエタリー リミティッド Prostate cancer treatment
EP1427377A2 (en) * 2001-09-20 2004-06-16 Cornell Research Foundation, Inc. Methods and compositions for treating or preventing skin disorders using binding agents specific for prostate specific membrane antigen
US8114965B2 (en) 2001-10-23 2012-02-14 Psma Development Company, Llc Compositions of PSMA antibodies
US8470330B2 (en) 2001-10-23 2013-06-25 Psma Development Company, Llc PSMA antibodies and uses thereof
US8859229B2 (en) 2007-02-02 2014-10-14 Yale University Transient transfection with RNA
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
WO2015164675A1 (en) 2014-04-23 2015-10-29 Juno Therapeutics, Inc. Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
US9181527B2 (en) 2009-10-29 2015-11-10 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US9242012B2 (en) 2008-09-08 2016-01-26 Psma Development Company, Llc Methods for killing PSMA-expressing, taxane-resistant cancer cells
US9273283B2 (en) 2009-10-29 2016-03-01 The Trustees Of Dartmouth College Method of producing T cell receptor-deficient T cells expressing a chimeric receptor
WO2016033570A1 (en) 2014-08-28 2016-03-03 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for cd19
WO2016064929A1 (en) 2014-10-20 2016-04-28 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
WO2016073602A2 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
WO2016090190A1 (en) 2014-12-03 2016-06-09 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
WO2016115177A1 (en) 2015-01-12 2016-07-21 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
WO2016115559A1 (en) 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
WO2016196388A1 (en) 2015-05-29 2016-12-08 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
US9573988B2 (en) 2013-02-20 2017-02-21 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
US9605049B2 (en) 2003-11-05 2017-03-28 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
WO2017053902A1 (en) 2015-09-25 2017-03-30 Abvitro Llc High throughput process for t cell receptor target identification of natively-paired t cell receptor sequences
WO2017053906A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Hiv antibody compositions and methods of use
WO2017068425A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068419A2 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
WO2017068421A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017079703A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
WO2017079705A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Chimeric receptors containing traf-inducing domains and related compositions and methods
WO2017096327A2 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Compositions and methods for reducing immune responses against cell therapies
WO2017096329A1 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
US9745368B2 (en) 2013-03-15 2017-08-29 The Trustees Of The University Of Pennsylvania Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2017161212A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for adaptive design of a treatment regimen and related treatments
WO2017161208A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
WO2017165571A1 (en) 2016-03-22 2017-09-28 Seattle Children's Hospital (dba Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US9790278B2 (en) 2012-05-07 2017-10-17 The Trustees Of Dartmouth College Anti-B7-H6 antibody, fusion proteins, and methods of using the same
WO2017193107A2 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
US9815901B2 (en) 2014-08-19 2017-11-14 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
WO2017205846A1 (en) 2016-05-27 2017-11-30 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
US9833476B2 (en) 2011-08-31 2017-12-05 The Trustees Of Dartmouth College NKP30 receptor targeted therapeutics
WO2017214207A2 (en) 2016-06-06 2017-12-14 Juno Therapeutics, Inc. Methods for the treatment of b cell malignancies using adoptive cell therapy
US9856322B2 (en) 2003-11-05 2018-01-02 St Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
WO2018005556A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Mhc-e restricted epitopes, binding molecules and related methods and uses
WO2018005559A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018023094A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
WO2018023093A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Immunomodulatory polypeptides and related compositions and methods
WO2018049420A1 (en) 2016-09-12 2018-03-15 Juno Therapeutics, Inc. Perfusion bioreactor bag assemblies
WO2018063985A1 (en) 2016-09-28 2018-04-05 Atossa Genetics Inc. Methods of adoptive cell therapy
WO2018067618A1 (en) 2016-10-03 2018-04-12 Juno Therapeutics, Inc. Hpv-specific binding molecules
WO2018071873A2 (en) 2016-10-13 2018-04-19 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
WO2018093591A1 (en) 2016-11-03 2018-05-24 Juno Therapeutics, Inc. Combination therapy of a cell based therapy and a microglia inhibitor
WO2018102787A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
WO2018102786A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for modulation of car-t cells
WO2018102785A2 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods and compositions for use of therapeutic t cells in combination with kinase inhibitors
WO2018106732A1 (en) 2016-12-05 2018-06-14 Juno Therapeutics, Inc. Production of engineered cells for adoptive cell therapy
US10017782B2 (en) 2007-02-02 2018-07-10 Yale University Immune cells modified by transient transfection of RNA
WO2018132518A1 (en) 2017-01-10 2018-07-19 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
WO2018134691A2 (en) 2017-01-20 2018-07-26 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
WO2018157171A2 (en) 2017-02-27 2018-08-30 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
WO2018170188A2 (en) 2017-03-14 2018-09-20 Juno Therapeutics, Inc. Methods for cryogenic storage
WO2018187791A1 (en) 2017-04-07 2018-10-11 Juno Therapeutics, Inc Engineered cells expressing prostate-specific membrane antigen (psma) or a modified form thereof and related methods
WO2018191723A1 (en) 2017-04-14 2018-10-18 Juno Therapeutics, Inc. Methods for assessing cell surface glycosylation
WO2018195175A1 (en) 2017-04-18 2018-10-25 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
WO2018197949A1 (en) 2017-04-27 2018-11-01 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
WO2018204427A1 (en) 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
WO2018223098A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
WO2018223101A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
US10155038B2 (en) 2007-02-02 2018-12-18 Yale University Cells prepared by transient transfection and methods of use thereof
WO2018234370A1 (en) 2017-06-20 2018-12-27 Institut Curie Immune cells defective for suv39h1
WO2019006427A1 (en) 2017-06-29 2019-01-03 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
WO2019027850A1 (en) 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors
WO2019032929A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods and compositions for preparing genetically engineered cells
WO2019032927A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
US10221245B2 (en) 2013-03-16 2019-03-05 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
WO2019046832A1 (en) 2017-09-01 2019-03-07 Juno Therapeutics, Inc. Gene expression and assessment of risk of developing toxicity following cell therapy
WO2019051335A1 (en) 2017-09-07 2019-03-14 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
WO2019057102A1 (en) 2017-09-20 2019-03-28 Tsinghua University A gRNA TARGETING HPK1 AND A METHOD FOR EDITING HPK1 GENE
US10253086B2 (en) 2015-04-08 2019-04-09 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
WO2019070541A1 (en) 2017-10-03 2019-04-11 Juno Therapeutics, Inc. Hpv-specific binding molecules
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
WO2019090004A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for producing a t cell composition
WO2019090202A1 (en) 2017-11-06 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of cblb in t cells for immunotherapy
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019089969A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
WO2019090003A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Chimeric antigen receptors specific for b-cell maturation antigen (bcma)
WO2019089855A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for generating therapeutic compositions of engineered cells
WO2019089884A2 (en) 2017-11-01 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
WO2019094835A1 (en) 2017-11-10 2019-05-16 Juno Therapeutics, Inc. Closed-system cryogenic vessels
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
WO2019113559A2 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
US10336804B2 (en) 2004-09-24 2019-07-02 Trustees Of Dartmouth College Chimeric NK receptor and methods for treating cancer
US10357514B2 (en) 2014-04-07 2019-07-23 The Trustees Of The University Of Pennsylvania Treatment of cancer using anti-CD19 Chimeric Antigen Receptor
WO2019152743A1 (en) 2018-01-31 2019-08-08 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
WO2019152747A1 (en) 2018-01-31 2019-08-08 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
WO2019170845A1 (en) 2018-03-09 2019-09-12 Ospedale San Raffaele S.R.L. Il-1 antagonist and toxicity induced by cell therapy
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
WO2019195492A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
WO2019195486A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
WO2019213184A1 (en) 2018-05-03 2019-11-07 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020033916A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
WO2020047099A1 (en) 2018-08-28 2020-03-05 Fred Hutchinson Cancer Research Center Methods and compositions for adoptive t cell therapy incorporating induced notch signaling
WO2020056047A1 (en) 2018-09-11 2020-03-19 Juno Therapeutics, Inc. Methods for mass spectrometry analysis of engineered cell compositions
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
WO2020089343A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020092848A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020097132A1 (en) 2018-11-06 2020-05-14 Juno Therapeutics, Inc. Process for producing genetically engineered t cells
WO2020097403A1 (en) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Methods and combinations for treatment and t cell modulation
WO2020102770A1 (en) 2018-11-16 2020-05-22 Juno Therapeutics, Inc. Methods of dosing engineered t cells for the treatment of b cell malignancies
WO2020106621A1 (en) 2018-11-19 2020-05-28 Board Of Regents, The University Of Texas System A modular, polycistronic vector for car and tcr transduction
WO2020113029A2 (en) 2018-11-28 2020-06-04 Board Of Regents, The University Of Texas System Multiplex genome editing of immune cells to enhance functionality and resistance to suppressive environment
WO2020112493A1 (en) 2018-11-29 2020-06-04 Board Of Regents, The University Of Texas System Methods for ex vivo expansion of natural killer cells and use thereof
WO2020113188A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for dosing and treatment of b cell malignancies in adoptive cell therapy
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020160050A1 (en) 2019-01-29 2020-08-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
US10738278B2 (en) 2014-07-15 2020-08-11 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
US10774388B2 (en) 2014-10-08 2020-09-15 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US10780120B2 (en) 2018-03-06 2020-09-22 The Trustees Of The University Of Pennsylvania Prostate-specific membrane antigen cars and methods of use thereof
US10786533B2 (en) 2015-07-15 2020-09-29 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
WO2020223535A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
US10829735B2 (en) 2015-07-21 2020-11-10 The Trustees Of The University Of Pennsylvania Methods for improving the efficacy and expansion of immune cells
WO2020247832A1 (en) 2019-06-07 2020-12-10 Juno Therapeutics, Inc. Automated t cell culture
WO2020252218A1 (en) 2019-06-12 2020-12-17 Juno Therapeutics, Inc. Combination therapy of a cell-mediated cytotoxic therapy and an inhibitor of a prosurvival bcl2 family protein
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2021041994A2 (en) 2019-08-30 2021-03-04 Juno Therapeutics, Inc. Machine learning methods for classifying cells
WO2021043804A1 (en) 2019-09-02 2021-03-11 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021078910A1 (en) 2019-10-22 2021-04-29 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021084050A1 (en) 2019-10-30 2021-05-06 Juno Therapeutics Gmbh Cell selection and/or stimulation devices and methods of use
WO2021092498A1 (en) 2019-11-07 2021-05-14 Juno Therapeutics, Inc. Combination of a t cell therapy and (s)-3-[4-(4-morpholin-4 ylmethyl-benzyloxy)-l-oxo-l,3-dihydro-isoindol-2-yl]- piperidine-2,6-dione
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
WO2021113776A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
WO2021113770A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Methods related to toxicity and response associated with cell therapy for treating b cell malignancies
WO2021151008A1 (en) 2020-01-24 2021-07-29 Juno Therapuetics, Inc. Methods for dosing and treatment of follicular lymphoma and marginal zone lymphoma in adoptive cell therapy
WO2021154887A1 (en) 2020-01-28 2021-08-05 Juno Therapeutics, Inc. Methods for t cell transduction
WO2021163389A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021163391A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Cd19-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021167908A1 (en) 2020-02-17 2021-08-26 Board Of Regents, The University Of Texas System Methods for expansion of tumor infiltrating lymphocytes and use thereof
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
WO2021231657A1 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2021228999A1 (en) 2020-05-12 2021-11-18 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
WO2021231661A2 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Process for producing donor-batched cells expressing a recombinant receptor
WO2021237068A2 (en) 2020-05-21 2021-11-25 Board Of Regents, The University Of Texas System T cell receptors with vgll1 specificity and uses thereof
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
WO2022023576A1 (en) 2020-07-30 2022-02-03 Institut Curie Immune cells defective for socs1
WO2022029660A1 (en) 2020-08-05 2022-02-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
WO2022104109A1 (en) 2020-11-13 2022-05-19 Catamaran Bio, Inc. Genetically modified natural killer cells and methods of use thereof
EP4011381A1 (en) 2016-06-03 2022-06-15 Memorial Sloan-Kettering Cancer Center Adoptive cell therapies as early treatment options
EP4012415A2 (en) 2015-12-04 2022-06-15 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
WO2022133030A1 (en) 2020-12-16 2022-06-23 Juno Therapeutics, Inc. Combination therapy of a cell therapy and a bcl2 inhibitor
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
WO2022187406A1 (en) 2021-03-03 2022-09-09 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
WO2022189620A1 (en) 2021-03-11 2022-09-15 Institut Curie Transmembrane neoantigenic peptides
WO2022189639A1 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides and uses thereof
WO2022189626A2 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides
WO2022204070A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Methods of determining potency of a therapeutic cell composition
WO2022204071A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
US11459390B2 (en) 2015-01-16 2022-10-04 Novartis Ag Phosphoglycerate kinase 1 (PGK) promoters and methods of use for expressing chimeric antigen receptor
WO2022212400A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Methods for dosing and treatment with a combination of a checkpoint inhibitor therapy and a car t cell therapy
WO2022212384A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Combination of a car t cell therapy and an immunomodulatory compound for treatment of lymphoma
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023014922A1 (en) 2021-08-04 2023-02-09 The Regents Of The University Of Colorado, A Body Corporate Lat activating chimeric antigen receptor t cells and methods of use thereof
US11608382B2 (en) 2018-06-13 2023-03-21 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
WO2023105000A1 (en) 2021-12-09 2023-06-15 Zygosity Limited Vector
WO2023115041A1 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae attachment glycoproteins
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
WO2023178348A1 (en) 2022-03-18 2023-09-21 The Regents Of The University Of Colorado, A Body Corporate Genetically engineered t-cell co-receptors and methods of use thereof
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
WO2023196921A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Granzyme expressing t cells and methods of use
WO2023196933A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Chimeric antigen receptor t cells and methods of use thereof
WO2023211972A1 (en) 2022-04-28 2023-11-02 Medical University Of South Carolina Chimeric antigen receptor modified regulatory t cells for treating cancer
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
US11845803B2 (en) 2017-02-17 2023-12-19 Fred Hutchinson Cancer Center Combination therapies for treatment of BCMA-related cancers and autoimmune disorders
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
EP4302768A2 (en) 2017-06-22 2024-01-10 Board Of Regents, The University Of Texas System Methods for producing regulatory immune cells and uses thereof
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
WO2024081820A1 (en) 2022-10-13 2024-04-18 Sana Biotechnology, Inc. Viral particles targeting hematopoietic stem cells

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996024676A1 (en) * 1995-02-08 1996-08-15 Whitehead Institute For Biomedical Research Bioactive fusion proteins and pre-existing tumor therapy

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996024676A1 (en) * 1995-02-08 1996-08-15 Whitehead Institute For Biomedical Research Bioactive fusion proteins and pre-existing tumor therapy

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
LINDEMANN ET AL.: "Versatile retrovirus vector systems for regulated gene expression in vitro and in vivo", MOL. MED., vol. 3, no. 7, July 1997 (1997-07-01), pages 466 - 476, XP002925323 *
LIU ET AL.: "Identification of rat prostate steroid-binding protein as a target antigen of experimental autoimmune prostatitis. Implications for prostate cancer therapy", J. IMMUNOL., vol. 159, no. 1, 1997, pages 472 - 480, XP002925325 *
MURPHY ET AL.: "Phase I clinical trial: T-cell therapy for prostate cancer using autologous dendritic cells pulsed with HLA-A0201-specific peptides from prostate-specific membrane antigen", THE PROSTATE, vol. 29, no. 6, 1996, pages 371 - 380, XP002925322 *
SALGALLER ET AL.: "Report of immune monitoring of prostate cancer patients undergoing T-cell therapy using dendritic cells pulsed with HLA-A2-specific peptides from prostate-specific membrane antigen (PSMA)", PROSTATE, vol. 35, no. 2, May 1998 (1998-05-01), pages 144 - 151, XP002925321 *
See also references of EP1109921A4 *
SIMMONS ET AL.: "GM-CSF as a systemic adjuvant in a phase II prostate cancer vaccine trial", PROSTATE, vol. 39, no. 4, June 1999 (1999-06-01), pages 291 - 297, XP002925324 *

Cited By (335)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100388942B1 (en) * 2000-07-08 2003-06-25 한화석유화학 주식회사 A Single Chain Variable Fragment of an Antibody for Protein Expressed on Cell Surface of Human Cortical Thymocyte and Leukemic Cell
JP2004505117A (en) * 2000-08-07 2004-02-19 ペプリン リサーチ プロプライエタリー リミティッド Prostate cancer treatment
WO2002040059A2 (en) * 2000-11-01 2002-05-23 American Foundation For Biological Research, Inc. Methods and compositions for inducing cell-mediated immune responses
WO2002040059A3 (en) * 2000-11-01 2003-09-12 American Foundation For Biolog Methods and compositions for inducing cell-mediated immune responses
US7192586B2 (en) 2001-09-20 2007-03-20 Cornell Research Foundation, Inc. Methods and compositions for treating or preventing skin disorders using binding agents specific for prostate specific membrane antigen
EP1427377A4 (en) * 2001-09-20 2006-04-12 Cornell Res Foundation Inc Methods and compositions for treating or preventing skin disorders using binding agents specific for prostate specific membrane antigen
EP1427377A2 (en) * 2001-09-20 2004-06-16 Cornell Research Foundation, Inc. Methods and compositions for treating or preventing skin disorders using binding agents specific for prostate specific membrane antigen
US8114965B2 (en) 2001-10-23 2012-02-14 Psma Development Company, Llc Compositions of PSMA antibodies
US8470330B2 (en) 2001-10-23 2013-06-25 Psma Development Company, Llc PSMA antibodies and uses thereof
US9695248B2 (en) 2001-10-23 2017-07-04 Psma Development Company, Llc PSMA antibodies and uses thereof
US9856322B2 (en) 2003-11-05 2018-01-02 St Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
US9605049B2 (en) 2003-11-05 2017-03-28 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
US11858976B2 (en) 2004-09-24 2024-01-02 The Trustees Of Dartmouth College Nucleic acid constructs encoding chimeric NK receptor, cells containing, and therapeutic use thereof
US10336804B2 (en) 2004-09-24 2019-07-02 Trustees Of Dartmouth College Chimeric NK receptor and methods for treating cancer
US11208454B2 (en) 2004-09-24 2021-12-28 Trustees Of Dartmouth College Chimeric NK receptor and methods for treating cancer
US9834590B2 (en) 2004-11-04 2017-12-05 St. Jude Children's Research Hospital, Inc. Chimeric receptors with 4-1BB stimulatory signaling domain
US11673937B2 (en) 2004-11-04 2023-06-13 St. Jude Children's Research Hospital, Inc. Methods for expanding immune cells
US10017782B2 (en) 2007-02-02 2018-07-10 Yale University Immune cells modified by transient transfection of RNA
US10155038B2 (en) 2007-02-02 2018-12-18 Yale University Cells prepared by transient transfection and methods of use thereof
US8859229B2 (en) 2007-02-02 2014-10-14 Yale University Transient transfection with RNA
US9242012B2 (en) 2008-09-08 2016-01-26 Psma Development Company, Llc Methods for killing PSMA-expressing, taxane-resistant cancer cells
US9957480B2 (en) 2009-10-29 2018-05-01 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US9938497B2 (en) 2009-10-29 2018-04-10 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US9821011B1 (en) 2009-10-29 2017-11-21 The Trustees Of Dartmouth College T-cell receptor-deficient T cell compositions
US9273283B2 (en) 2009-10-29 2016-03-01 The Trustees Of Dartmouth College Method of producing T cell receptor-deficient T cells expressing a chimeric receptor
US9181527B2 (en) 2009-10-29 2015-11-10 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US9663763B2 (en) 2009-10-29 2017-05-30 The Trustees Of Dartmouth College T-cell receptor-deficient T cell compositions
US9822340B1 (en) 2009-10-29 2017-11-21 The Trustees Of Dartmouth College T-cell receptor-deficient T cell compositions
US11834676B2 (en) 2009-10-29 2023-12-05 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US10689616B1 (en) 2009-10-29 2020-06-23 The Trustees Of Dartmouth College T-cell receptor-deficient t cell compositions
US11136549B2 (en) 2009-10-29 2021-10-05 The Trustees Of Dartmouth College T-cell receptor-deficient T cell compositions
US10689618B2 (en) 2009-10-29 2020-06-23 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US10689619B2 (en) 2009-10-29 2020-06-23 The Trustees Of Dartmouth College T cell receptor-deficient T cell compositions
US10689617B1 (en) 2009-10-29 2020-06-23 The Trustees Of Dartmouth College T-cell receptor-deficient T cell compositions
US9101584B2 (en) 2010-12-09 2015-08-11 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
US9540445B2 (en) 2010-12-09 2017-01-10 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US9518123B2 (en) 2010-12-09 2016-12-13 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US8916381B1 (en) 2010-12-09 2014-12-23 The Trustees Of The University Of Pennyslvania Methods for treatment of cancer
US8975071B1 (en) 2010-12-09 2015-03-10 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US9499629B2 (en) 2010-12-09 2016-11-22 The Trustees Of The University Of Pennsylvania Use of chimeric antigen receptor-modified T-cells to treat cancer
US9481728B2 (en) 2010-12-09 2016-11-01 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US9464140B2 (en) 2010-12-09 2016-10-11 The Trustees Of The University Of Pennsylvania Compositions and methods for treatment of cancer
US8911993B2 (en) 2010-12-09 2014-12-16 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US8906682B2 (en) 2010-12-09 2014-12-09 The Trustees Of The University Of Pennsylvania Methods for treatment of cancer
US9102761B2 (en) 2010-12-09 2015-08-11 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US9102760B2 (en) 2010-12-09 2015-08-11 The Trustees Of The University Of Pennsylvania Compositions for treatment of cancer
US9328156B2 (en) 2010-12-09 2016-05-03 The Trustees Of The University Of Pennsylvania Use of chimeric antigen receptor-modified T cells to treat cancer
US9833476B2 (en) 2011-08-31 2017-12-05 The Trustees Of Dartmouth College NKP30 receptor targeted therapeutics
US11872248B2 (en) 2011-08-31 2024-01-16 The Trustees Of Dartmouth College Nucleic acids encoding chimeric receptor comprising NKP30 receptor and CD28 and CD3 zeta domains and human T cell containing
US10682378B2 (en) 2011-08-31 2020-06-16 The Trustees Of Dartmouth College NKP30 receptor targeted therapeutics
US11034766B2 (en) 2012-05-07 2021-06-15 Trustees Of Dartmouth College Anti-B7-H6 antibody, fusion proteins, and methods of using the same
US9790278B2 (en) 2012-05-07 2017-10-17 The Trustees Of Dartmouth College Anti-B7-H6 antibody, fusion proteins, and methods of using the same
US9394368B2 (en) 2013-02-20 2016-07-19 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US9573988B2 (en) 2013-02-20 2017-02-21 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
US11865167B2 (en) 2013-02-20 2024-01-09 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US10308717B2 (en) 2013-02-20 2019-06-04 Novartis Ag Treatment of cancer using humanized anti-EGFRvIII chimeric antigen receptor
US11028177B2 (en) 2013-02-20 2021-06-08 Novartis Ag Effective targeting of primary human leukemia using anti-CD123 chimeric antigen receptor engineered T cells
US9745368B2 (en) 2013-03-15 2017-08-29 The Trustees Of The University Of Pennsylvania Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US11919946B2 (en) 2013-03-15 2024-03-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10640553B2 (en) 2013-03-15 2020-05-05 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10927184B2 (en) 2013-03-16 2021-02-23 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
US10221245B2 (en) 2013-03-16 2019-03-05 Novartis Ag Treatment of cancer using humanized anti-CD19 chimeric antigen receptor
US10640569B2 (en) 2013-12-19 2020-05-05 Novartis Ag Human mesothelin chimeric antigen receptors and uses thereof
US10287354B2 (en) 2013-12-20 2019-05-14 Novartis Ag Regulatable chimeric antigen receptor
US11578130B2 (en) 2013-12-20 2023-02-14 Novartis Ag Regulatable chimeric antigen receptor
US11028143B2 (en) 2014-01-21 2021-06-08 Novartis Ag Enhanced antigen presenting ability of RNA CAR T cells by co-introduction of costimulatory molecules
US10357514B2 (en) 2014-04-07 2019-07-23 The Trustees Of The University Of Pennsylvania Treatment of cancer using anti-CD19 Chimeric Antigen Receptor
EP3647412A1 (en) 2014-04-23 2020-05-06 Juno Therapeutics, Inc. Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
WO2015164675A1 (en) 2014-04-23 2015-10-29 Juno Therapeutics, Inc. Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
US11400115B2 (en) 2014-04-23 2022-08-02 Juno Therapeutics, Inc. Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
EP4219687A1 (en) 2014-04-23 2023-08-02 Juno Therapeutics, Inc. Methods for isolating, culturing, and genetically engineering immune cell populations for adoptive therapy
US10428305B2 (en) 2014-05-15 2019-10-01 National University Of Singapore Modified natural killer cells that express IL15 and uses thereof
US10774311B2 (en) 2014-05-15 2020-09-15 National University Of Singapore Natural killer cells modified to express membrane-bound interleukin 15 and uses thereof
US11560548B2 (en) 2014-05-15 2023-01-24 National University Of Singapore Immune cells expressing membrane-bound interleukin 15 (mbIL15) and uses thereof
US10738278B2 (en) 2014-07-15 2020-08-11 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
US10851166B2 (en) 2014-07-21 2020-12-01 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US10568947B2 (en) 2014-07-21 2020-02-25 Novartis Ag Treatment of cancer using a CLL-1 chimeric antigen receptor
US9777061B2 (en) 2014-07-21 2017-10-03 Novartis Ag Treatment of cancer using a CD33 chimeric antigen receptor
US10174095B2 (en) 2014-07-21 2019-01-08 Novartis Ag Nucleic acid encoding a humanized anti-BCMA chimeric antigen receptor
US11084880B2 (en) 2014-07-21 2021-08-10 Novartis Ag Anti-BCMA chimeric antigen receptor
US11542488B2 (en) 2014-07-21 2023-01-03 Novartis Ag Sortase synthesized chimeric antigen receptors
US10703819B2 (en) 2014-08-09 2020-07-07 The Trustees Of The University Of Pennsylvania Treatment of cancer using a CD123 chimeric antigen receptor
US9815901B2 (en) 2014-08-19 2017-11-14 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
US11591404B2 (en) 2014-08-19 2023-02-28 Novartis Ag Treatment of cancer using a CD123 chimeric antigen receptor
US11827714B2 (en) 2014-08-28 2023-11-28 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for CD19
US10533055B2 (en) 2014-08-28 2020-01-14 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for CD19
WO2016033570A1 (en) 2014-08-28 2016-03-03 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for cd19
EP3805267A1 (en) 2014-08-28 2021-04-14 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for cd19
US10577417B2 (en) 2014-09-17 2020-03-03 Novartis Ag Targeting cytotoxic cells with chimeric receptors for adoptive immunotherapy
US10774388B2 (en) 2014-10-08 2020-09-15 Novartis Ag Biomarkers predictive of therapeutic responsiveness to chimeric antigen receptor therapy and uses thereof
US10507219B2 (en) 2014-10-20 2019-12-17 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
WO2016064929A1 (en) 2014-10-20 2016-04-28 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
US11633426B2 (en) 2014-10-20 2023-04-25 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
EP3932950A1 (en) 2014-10-20 2022-01-05 Juno Therapeutics, Inc. Methods and compositions for dosing in adoptive cell therapy
US11802295B2 (en) 2014-11-05 2023-10-31 Juno Therapeutics, Inc. Methods for transduction and cell processing
WO2016073602A2 (en) 2014-11-05 2016-05-12 Juno Therapeutics, Inc. Methods for transduction and cell processing
EP3757206A1 (en) 2014-11-05 2020-12-30 Juno Therapeutics, Inc. Methods for transduction and cell processing
US10428351B2 (en) 2014-11-05 2019-10-01 Juno Therapeutics, Inc. Methods for transduction and cell processing
US11266739B2 (en) 2014-12-03 2022-03-08 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
WO2016090190A1 (en) 2014-12-03 2016-06-09 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
EP3766895A1 (en) 2014-12-03 2021-01-20 Juno Therapeutics, Inc. Methods and compositions for adoptive cell therapy
US10273300B2 (en) 2014-12-29 2019-04-30 The Trustees Of The University Of Pennsylvania Methods of making chimeric antigen receptor-expressing cells
US10363269B2 (en) 2015-01-12 2019-07-30 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
WO2016115177A1 (en) 2015-01-12 2016-07-21 Juno Therapeutics, Inc. Modified hepatitis post-transcriptional regulatory elements
US10889652B2 (en) 2015-01-16 2021-01-12 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ROR1
US11919970B2 (en) 2015-01-16 2024-03-05 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ROR1
WO2016115559A1 (en) 2015-01-16 2016-07-21 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
US11459390B2 (en) 2015-01-16 2022-10-04 Novartis Ag Phosphoglycerate kinase 1 (PGK) promoters and methods of use for expressing chimeric antigen receptor
EP3760644A1 (en) 2015-01-16 2021-01-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for ror1
US11161907B2 (en) 2015-02-02 2021-11-02 Novartis Ag Car-expressing cells against multiple tumor antigens and uses thereof
US10253086B2 (en) 2015-04-08 2019-04-09 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
US11149076B2 (en) 2015-04-08 2021-10-19 Novartis Ag CD20 therapies, CD22 therapies, and combination therapies with a CD19 chimeric antigen receptor (CAR)-expressing cell
WO2016166568A1 (en) 2015-04-16 2016-10-20 Juno Therapeutics Gmbh Methods, kits and apparatus for expanding a population of cells
US11896614B2 (en) 2015-04-17 2024-02-13 Novartis Ag Methods for improving the efficacy and expansion of chimeric antigen receptor-expressing cells
WO2016196388A1 (en) 2015-05-29 2016-12-08 Juno Therapeutics, Inc. Composition and methods for regulating inhibitory interactions in genetically engineered cells
US10786533B2 (en) 2015-07-15 2020-09-29 Juno Therapeutics, Inc. Engineered cells for adoptive cell therapy
US10829735B2 (en) 2015-07-21 2020-11-10 The Trustees Of The University Of Pennsylvania Methods for improving the efficacy and expansion of immune cells
US11667691B2 (en) 2015-08-07 2023-06-06 Novartis Ag Treatment of cancer using chimeric CD3 receptor proteins
US11747346B2 (en) 2015-09-03 2023-09-05 Novartis Ag Biomarkers predictive of cytokine release syndrome
EP3662930A1 (en) 2015-09-24 2020-06-10 AbVitro LLC Hiv antibody compositions and methods of use
WO2017053906A1 (en) 2015-09-24 2017-03-30 Abvitro Llc Hiv antibody compositions and methods of use
WO2017053902A1 (en) 2015-09-25 2017-03-30 Abvitro Llc High throughput process for t cell receptor target identification of natively-paired t cell receptor sequences
US11913024B2 (en) 2015-10-22 2024-02-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068419A2 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
US11466253B2 (en) 2015-10-22 2022-10-11 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
WO2017068425A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
US11248238B2 (en) 2015-10-22 2022-02-15 Juno Therapeutics Gmbh Methods, kits, agents and apparatuses for transduction
WO2017068421A1 (en) 2015-10-22 2017-04-27 Juno Therapeutics Gmbh Methods for culturing cells and kits and apparatus for same
US11020429B2 (en) 2015-11-05 2021-06-01 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
WO2017079705A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Chimeric receptors containing traf-inducing domains and related compositions and methods
WO2017079703A1 (en) 2015-11-05 2017-05-11 Juno Therapeutics, Inc. Vectors and genetically engineered immune cells expressing metabolic pathway modulators and uses in adoptive cell therapy
EP4212547A1 (en) 2015-12-03 2023-07-19 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
EP4212166A1 (en) 2015-12-03 2023-07-19 Juno Therapeutics, Inc. Compositions and methods for reducing immune responses against cell therapies
WO2017096327A2 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Compositions and methods for reducing immune responses against cell therapies
WO2017096329A1 (en) 2015-12-03 2017-06-08 Juno Therapeutics, Inc. Modified chimeric receptors and related compositions and methods
US11815514B2 (en) 2015-12-04 2023-11-14 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
EP4012415A2 (en) 2015-12-04 2022-06-15 Juno Therapeutics, Inc. Methods and compositions related to toxicity associated with cell therapy
US11413340B2 (en) 2015-12-22 2022-08-16 Novartis Ag Mesothelin chimeric antigen receptor (CAR) and antibody against PD-L1 inhibitor for combined use in anticancer therapy
WO2017161212A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for adaptive design of a treatment regimen and related treatments
WO2017161208A1 (en) 2016-03-16 2017-09-21 Juno Therapeutics, Inc. Methods for determining dosing of a therapeutic agent and related treatments
US11518814B2 (en) 2016-03-22 2022-12-06 Seattle Children's Hospital Early intervention methods to prevent or ameliorate toxicity
US11760804B2 (en) 2016-03-22 2023-09-19 Seattle Children's Hospital Early intervention methods to prevent or ameliorate toxicity
EP4015536A1 (en) 2016-03-22 2022-06-22 Seattle Children's Hospital (DBA Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
WO2017165571A1 (en) 2016-03-22 2017-09-28 Seattle Children's Hospital (dba Seattle Children's Research Institute) Early intervention methods to prevent or ameliorate toxicity
US11549099B2 (en) 2016-03-23 2023-01-10 Novartis Ag Cell secreted minibodies and uses thereof
WO2017193107A2 (en) 2016-05-06 2017-11-09 Juno Therapeutics, Inc. Genetically engineered cells and methods of making the same
EP3910059A1 (en) 2016-05-27 2021-11-17 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
WO2017205846A1 (en) 2016-05-27 2017-11-30 Aadigen, Llc Peptides and nanoparticles for intracellular delivery of genome-editing molecules
EP4011381A1 (en) 2016-06-03 2022-06-15 Memorial Sloan-Kettering Cancer Center Adoptive cell therapies as early treatment options
WO2017214207A2 (en) 2016-06-06 2017-12-14 Juno Therapeutics, Inc. Methods for the treatment of b cell malignancies using adoptive cell therapy
WO2018005556A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Mhc-e restricted epitopes, binding molecules and related methods and uses
EP3992632A1 (en) 2016-06-27 2022-05-04 Juno Therapeutics, Inc. Mhc-e restricted epitopes, binding molecules and related methods and uses
WO2018005559A1 (en) 2016-06-27 2018-01-04 Juno Therapeutics, Inc. Method of identifying peptide epitopes, molecules that bind such epitopes and related uses
US11421287B2 (en) 2016-07-29 2022-08-23 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018023094A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Methods for assessing the presence or absence of replication competent virus
WO2018023093A1 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Immunomodulatory polypeptides and related compositions and methods
WO2018049420A1 (en) 2016-09-12 2018-03-15 Juno Therapeutics, Inc. Perfusion bioreactor bag assemblies
EP4353319A2 (en) 2016-09-28 2024-04-17 Atossa Therapeutics, Inc. Methods of adoptive cell therapy
WO2018063985A1 (en) 2016-09-28 2018-04-05 Atossa Genetics Inc. Methods of adoptive cell therapy
WO2018067618A1 (en) 2016-10-03 2018-04-12 Juno Therapeutics, Inc. Hpv-specific binding molecules
US11072660B2 (en) 2016-10-03 2021-07-27 Juno Therapeutics, Inc. HPV-specific binding molecules
US11026976B2 (en) 2016-10-07 2021-06-08 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
USRE49847E1 (en) 2016-10-07 2024-02-27 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
US11872249B2 (en) 2016-10-07 2024-01-16 Novartis Ag Method of treating cancer by administering immune effector cells expressing a chimeric antigen receptor comprising a CD20 binding domain
US10525083B2 (en) 2016-10-07 2020-01-07 Novartis Ag Nucleic acid molecules encoding chimeric antigen receptors comprising a CD20 binding domain
WO2018071873A2 (en) 2016-10-13 2018-04-19 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
US11896615B2 (en) 2016-10-13 2024-02-13 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
EP4190335A1 (en) 2016-10-13 2023-06-07 Juno Therapeutics, Inc. Immunotherapy methods and compositions involving tryptophan metabolic pathway modulators
WO2018093591A1 (en) 2016-11-03 2018-05-24 Juno Therapeutics, Inc. Combination therapy of a cell based therapy and a microglia inhibitor
WO2018085731A2 (en) 2016-11-03 2018-05-11 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and a btk inhibitor
WO2018102787A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
WO2018102786A1 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods for modulation of car-t cells
WO2018102785A2 (en) 2016-12-03 2018-06-07 Juno Therapeutics, Inc. Methods and compositions for use of therapeutic t cells in combination with kinase inhibitors
EP4279136A2 (en) 2016-12-03 2023-11-22 Juno Therapeutics, Inc. Methods for determining car-t cells dosing
US11590167B2 (en) 2016-12-03 2023-02-28 Juno Therapeutic, Inc. Methods and compositions for use of therapeutic T cells in combination with kinase inhibitors
WO2018106732A1 (en) 2016-12-05 2018-06-14 Juno Therapeutics, Inc. Production of engineered cells for adoptive cell therapy
WO2018132518A1 (en) 2017-01-10 2018-07-19 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
US11821027B2 (en) 2017-01-10 2023-11-21 Juno Therapeutics, Inc. Epigenetic analysis of cell therapy and related methods
WO2018134691A2 (en) 2017-01-20 2018-07-26 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
US11517627B2 (en) 2017-01-20 2022-12-06 Juno Therapeutics Gmbh Cell surface conjugates and related cell compositions and methods
US11535662B2 (en) 2017-01-26 2022-12-27 Novartis Ag CD28 compositions and methods for chimeric antigen receptor therapy
US11845803B2 (en) 2017-02-17 2023-12-19 Fred Hutchinson Cancer Center Combination therapies for treatment of BCMA-related cancers and autoimmune disorders
EP4353818A2 (en) 2017-02-27 2024-04-17 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
WO2018157171A2 (en) 2017-02-27 2018-08-30 Juno Therapeutics, Inc. Compositions, articles of manufacture and methods related to dosing in cell therapy
US11850262B2 (en) 2017-02-28 2023-12-26 Purdue Research Foundation Compositions and methods for CAR T cell therapy
US11759480B2 (en) 2017-02-28 2023-09-19 Endocyte, Inc. Compositions and methods for CAR T cell therapy
WO2018170188A2 (en) 2017-03-14 2018-09-20 Juno Therapeutics, Inc. Methods for cryogenic storage
US11851659B2 (en) 2017-03-22 2023-12-26 Novartis Ag Compositions and methods for immunooncology
US11365236B2 (en) 2017-03-27 2022-06-21 Nkarta, Inc. Truncated NKG2D chimeric receptors and uses thereof in natural killer cell immunotherapy
WO2018187791A1 (en) 2017-04-07 2018-10-11 Juno Therapeutics, Inc Engineered cells expressing prostate-specific membrane antigen (psma) or a modified form thereof and related methods
US11796534B2 (en) 2017-04-14 2023-10-24 Juno Therapeutics, Inc. Methods for assessing cell surface glycosylation
WO2018191723A1 (en) 2017-04-14 2018-10-18 Juno Therapeutics, Inc. Methods for assessing cell surface glycosylation
EP4083063A2 (en) 2017-04-18 2022-11-02 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
WO2018195175A1 (en) 2017-04-18 2018-10-25 FUJIFILM Cellular Dynamics, Inc. Antigen-specific immune effector cells
WO2018197949A1 (en) 2017-04-27 2018-11-01 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
US11866465B2 (en) 2017-04-27 2024-01-09 Juno Therapeutics Gmbh Oligomeric particle reagents and methods of use thereof
EP4327878A2 (en) 2017-05-01 2024-02-28 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
WO2018204427A1 (en) 2017-05-01 2018-11-08 Juno Therapeutics, Inc. Combination of a cell therapy and an immunomodulatory compound
WO2018223101A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
US11944647B2 (en) 2017-06-02 2024-04-02 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
US11413310B2 (en) 2017-06-02 2022-08-16 Juno Therapeutics, Inc. Articles of manufacture and methods for treatment using adoptive cell therapy
WO2018223098A1 (en) 2017-06-02 2018-12-06 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
US11740231B2 (en) 2017-06-02 2023-08-29 Juno Therapeutics, Inc. Articles of manufacture and methods related to toxicity associated with cell therapy
EP3828264A1 (en) 2017-06-20 2021-06-02 Institut Curie Immune cells defective for suv39h1
WO2018234370A1 (en) 2017-06-20 2018-12-27 Institut Curie Immune cells defective for suv39h1
EP4302768A2 (en) 2017-06-22 2024-01-10 Board Of Regents, The University Of Texas System Methods for producing regulatory immune cells and uses thereof
WO2019006427A1 (en) 2017-06-29 2019-01-03 Juno Therapeutics, Inc. Mouse model for assessing toxicities associated with immunotherapies
WO2019027850A1 (en) 2017-07-29 2019-02-07 Juno Therapeutics, Inc. Reagents for expanding cells expressing recombinant receptors
US11851678B2 (en) 2017-08-09 2023-12-26 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
WO2019032927A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods for producing genetically engineered cell compositions and related compositions
WO2019032929A1 (en) 2017-08-09 2019-02-14 Juno Therapeutics, Inc. Methods and compositions for preparing genetically engineered cells
WO2019046832A1 (en) 2017-09-01 2019-03-07 Juno Therapeutics, Inc. Gene expression and assessment of risk of developing toxicity following cell therapy
WO2019051335A1 (en) 2017-09-07 2019-03-14 Juno Therapeutics, Inc. Methods of identifying cellular attributes related to outcomes associated with cell therapy
WO2019057102A1 (en) 2017-09-20 2019-03-28 Tsinghua University A gRNA TARGETING HPK1 AND A METHOD FOR EDITING HPK1 GENE
WO2019070541A1 (en) 2017-10-03 2019-04-11 Juno Therapeutics, Inc. Hpv-specific binding molecules
US11952408B2 (en) 2017-10-03 2024-04-09 Juno Therapeutics, Inc. HPV-specific binding molecules
EP4215543A2 (en) 2017-10-03 2023-07-26 Juno Therapeutics, Inc. Hpv-specific binding molecules
US11851679B2 (en) 2017-11-01 2023-12-26 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019089982A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Method of assessing activity of recombinant antigen receptors
WO2019090004A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for producing a t cell composition
US11066475B2 (en) 2017-11-01 2021-07-20 Juno Therapeutics, Inc. Chimeric antigen receptors specific for B-cell maturation antigen and encoding polynucleotides
US11564946B2 (en) 2017-11-01 2023-01-31 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019089969A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for b-cell maturation antigen
WO2019089848A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods associated with tumor burden for assessing response to a cell therapy
WO2019089858A2 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Methods of assessing or monitoring a response to a cell therapy
WO2019089884A2 (en) 2017-11-01 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of tgfbr2 in t cells for immunotherapy
US11623961B2 (en) 2017-11-01 2023-04-11 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for B-cell maturation antigen
WO2019090003A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Chimeric antigen receptors specific for b-cell maturation antigen (bcma)
WO2019089855A1 (en) 2017-11-01 2019-05-09 Juno Therapeutics, Inc. Process for generating therapeutic compositions of engineered cells
WO2019090364A1 (en) 2017-11-06 2019-05-09 Juno Therapeutics, Inc. Combination of a cell therapy and a gamma secretase inhibitor
WO2019090202A1 (en) 2017-11-06 2019-05-09 Editas Medicine, Inc. Methods, compositions and components for crispr-cas9 editing of cblb in t cells for immunotherapy
WO2019094835A1 (en) 2017-11-10 2019-05-16 Juno Therapeutics, Inc. Closed-system cryogenic vessels
WO2019109053A1 (en) 2017-12-01 2019-06-06 Juno Therapeutics, Inc. Methods for dosing and for modulation of genetically engineered cells
WO2019113557A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Process for producing a composition of engineered t cells
WO2019113556A1 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Serum-free media formulation for culturing cells and methods of use thereof
WO2019113559A2 (en) 2017-12-08 2019-06-13 Juno Therapeutics, Inc. Phenotypic markers for cell therapy and related methods
WO2019118937A1 (en) 2017-12-15 2019-06-20 Juno Therapeutics, Inc. Anti-cct5 binding molecules and methods of use thereof
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
US11779602B2 (en) 2018-01-22 2023-10-10 Endocyte, Inc. Methods of use for CAR T cells
WO2019152747A1 (en) 2018-01-31 2019-08-08 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
WO2019152743A1 (en) 2018-01-31 2019-08-08 Celgene Corporation Combination therapy using adoptive cell therapy and checkpoint inhibitor
US11535903B2 (en) 2018-01-31 2022-12-27 Juno Therapeutics, Inc. Methods and reagents for assessing the presence or absence of replication competent virus
US10780120B2 (en) 2018-03-06 2020-09-22 The Trustees Of The University Of Pennsylvania Prostate-specific membrane antigen cars and methods of use thereof
WO2019170845A1 (en) 2018-03-09 2019-09-12 Ospedale San Raffaele S.R.L. Il-1 antagonist and toxicity induced by cell therapy
WO2019195492A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. Methods of producing cells expressing a recombinant receptor and related compositions
WO2019195486A1 (en) 2018-04-05 2019-10-10 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
US11471489B2 (en) 2018-04-05 2022-10-18 Juno Therapeutics, Inc. T cell receptors and engineered cells expressing same
WO2019213184A1 (en) 2018-05-03 2019-11-07 Juno Therapeutics, Inc. Combination therapy of a chimeric antigen receptor (car) t cell therapy and a kinase inhibitor
US11939389B2 (en) 2018-06-13 2024-03-26 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11952428B2 (en) 2018-06-13 2024-04-09 Novartis Ag BCMA chimeric antigen receptors and uses thereof
US11608382B2 (en) 2018-06-13 2023-03-21 Novartis Ag BCMA chimeric antigen receptors and uses thereof
WO2020033927A2 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Processes for generating engineered cells and compositions thereof
WO2020033916A1 (en) 2018-08-09 2020-02-13 Juno Therapeutics, Inc. Methods for assessing integrated nucleic acids
WO2020047099A1 (en) 2018-08-28 2020-03-05 Fred Hutchinson Cancer Research Center Methods and compositions for adoptive t cell therapy incorporating induced notch signaling
WO2020056047A1 (en) 2018-09-11 2020-03-19 Juno Therapeutics, Inc. Methods for mass spectrometry analysis of engineered cell compositions
WO2020089343A1 (en) 2018-10-31 2020-05-07 Juno Therapeutics Gmbh Methods for selection and stimulation of cells and apparatus for same
WO2020092848A2 (en) 2018-11-01 2020-05-07 Juno Therapeutics, Inc. Methods for treatment using chimeric antigen receptors specific for b-cell maturation antigen
WO2020097132A1 (en) 2018-11-06 2020-05-14 Juno Therapeutics, Inc. Process for producing genetically engineered t cells
WO2020097403A1 (en) 2018-11-08 2020-05-14 Juno Therapeutics, Inc. Methods and combinations for treatment and t cell modulation
WO2020102770A1 (en) 2018-11-16 2020-05-22 Juno Therapeutics, Inc. Methods of dosing engineered t cells for the treatment of b cell malignancies
WO2020106621A1 (en) 2018-11-19 2020-05-28 Board Of Regents, The University Of Texas System A modular, polycistronic vector for car and tcr transduction
WO2020113029A2 (en) 2018-11-28 2020-06-04 Board Of Regents, The University Of Texas System Multiplex genome editing of immune cells to enhance functionality and resistance to suppressive environment
WO2020112493A1 (en) 2018-11-29 2020-06-04 Board Of Regents, The University Of Texas System Methods for ex vivo expansion of natural killer cells and use thereof
WO2020113194A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for treatment using adoptive cell therapy
WO2020113188A2 (en) 2018-11-30 2020-06-04 Juno Therapeutics, Inc. Methods for dosing and treatment of b cell malignancies in adoptive cell therapy
WO2020160050A1 (en) 2019-01-29 2020-08-06 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for receptor tyrosine kinase like orphan receptor 1 (ror1)
US11154575B2 (en) 2019-03-05 2021-10-26 Nkarta, Inc. Cancer immunotherapy using CD19-directed chimeric antigen receptors
US11253547B2 (en) 2019-03-05 2022-02-22 Nkarta, Inc. CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
US11141436B2 (en) 2019-03-05 2021-10-12 Nkarta, Inc. Immune cells engineered to express CD19-directed chimeric antigen receptors and uses thereof in immunotherapy
WO2020223571A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a chimeric receptor from a modified cd247 locus, related polynucleotides and methods
WO2020223535A1 (en) 2019-05-01 2020-11-05 Juno Therapeutics, Inc. Cells expressing a recombinant receptor from a modified tgfbr2 locus, related polynucleotides and methods
WO2020247832A1 (en) 2019-06-07 2020-12-10 Juno Therapeutics, Inc. Automated t cell culture
WO2020252218A1 (en) 2019-06-12 2020-12-17 Juno Therapeutics, Inc. Combination therapy of a cell-mediated cytotoxic therapy and an inhibitor of a prosurvival bcl2 family protein
WO2021013950A1 (en) 2019-07-23 2021-01-28 Mnemo Therapeutics Immune cells defective for suv39h1
WO2021035194A1 (en) 2019-08-22 2021-02-25 Juno Therapeutics, Inc. Combination therapy of a t cell therapy and an enhancer of zeste homolog 2 (ezh2) inhibitor and related methods
WO2021041994A2 (en) 2019-08-30 2021-03-04 Juno Therapeutics, Inc. Machine learning methods for classifying cells
WO2021043804A1 (en) 2019-09-02 2021-03-11 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021078910A1 (en) 2019-10-22 2021-04-29 Institut Curie Immunotherapy targeting tumor neoantigenic peptides
WO2021084050A1 (en) 2019-10-30 2021-05-06 Juno Therapeutics Gmbh Cell selection and/or stimulation devices and methods of use
WO2021092498A1 (en) 2019-11-07 2021-05-14 Juno Therapeutics, Inc. Combination of a t cell therapy and (s)-3-[4-(4-morpholin-4 ylmethyl-benzyloxy)-l-oxo-l,3-dihydro-isoindol-2-yl]- piperidine-2,6-dione
WO2021113780A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to gprc5d-targeted binding domains and related compositions and methods
WO2021113776A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to bcma-targeted binding domains and related compositions and methods
WO2021113770A1 (en) 2019-12-06 2021-06-10 Juno Therapeutics, Inc. Methods related to toxicity and response associated with cell therapy for treating b cell malignancies
WO2021151008A1 (en) 2020-01-24 2021-07-29 Juno Therapuetics, Inc. Methods for dosing and treatment of follicular lymphoma and marginal zone lymphoma in adoptive cell therapy
WO2021154887A1 (en) 2020-01-28 2021-08-05 Juno Therapeutics, Inc. Methods for t cell transduction
WO2021163389A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Bcma-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021163391A1 (en) 2020-02-12 2021-08-19 Juno Therapeutics, Inc. Cd19-directed chimeric antigen receptor t cell compositions and methods and uses thereof
WO2021167908A1 (en) 2020-02-17 2021-08-26 Board Of Regents, The University Of Texas System Methods for expansion of tumor infiltrating lymphocytes and use thereof
WO2021207689A2 (en) 2020-04-10 2021-10-14 Juno Therapeutics, Inc. Methods and uses related to cell therapy engineered with a chimeric antigen receptor targeting b-cell maturation antigen
WO2021228999A1 (en) 2020-05-12 2021-11-18 Institut Curie Neoantigenic epitopes associated with sf3b1 mutations
WO2021231661A2 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Process for producing donor-batched cells expressing a recombinant receptor
WO2021231657A1 (en) 2020-05-13 2021-11-18 Juno Therapeutics, Inc. Methods of identifying features associated with clinical response and uses thereof
WO2021237068A2 (en) 2020-05-21 2021-11-25 Board Of Regents, The University Of Texas System T cell receptors with vgll1 specificity and uses thereof
WO2021260186A1 (en) 2020-06-26 2021-12-30 Juno Therapeutics Gmbh Engineered t cells conditionally expressing a recombinant receptor, related polynucleotides and methods
WO2022023576A1 (en) 2020-07-30 2022-02-03 Institut Curie Immune cells defective for socs1
WO2022029660A1 (en) 2020-08-05 2022-02-10 Juno Therapeutics, Inc. Anti-idiotypic antibodies to ror1-targeted binding domains and related compositions and methods
WO2022104109A1 (en) 2020-11-13 2022-05-19 Catamaran Bio, Inc. Genetically modified natural killer cells and methods of use thereof
WO2022133030A1 (en) 2020-12-16 2022-06-23 Juno Therapeutics, Inc. Combination therapy of a cell therapy and a bcl2 inhibitor
WO2022150731A1 (en) 2021-01-11 2022-07-14 Sana Biotechnology, Inc. Use of cd8-targeted viral vectors
WO2022187406A1 (en) 2021-03-03 2022-09-09 Juno Therapeutics, Inc. Combination of a t cell therapy and a dgk inhibitor
WO2022189620A1 (en) 2021-03-11 2022-09-15 Institut Curie Transmembrane neoantigenic peptides
WO2022189639A1 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides and uses thereof
WO2022189626A2 (en) 2021-03-11 2022-09-15 Mnemo Therapeutics Tumor neoantigenic peptides
WO2022204070A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Methods of determining potency of a therapeutic cell composition
WO2022204071A1 (en) 2021-03-22 2022-09-29 Juno Therapeutics, Inc. Method to assess potency of viral vector particles
WO2022212400A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Methods for dosing and treatment with a combination of a checkpoint inhibitor therapy and a car t cell therapy
WO2022212384A1 (en) 2021-03-29 2022-10-06 Juno Therapeutics, Inc. Combination of a car t cell therapy and an immunomodulatory compound for treatment of lymphoma
WO2022234009A2 (en) 2021-05-06 2022-11-10 Juno Therapeutics Gmbh Methods for stimulating and transducing t cells
WO2023014922A1 (en) 2021-08-04 2023-02-09 The Regents Of The University Of Colorado, A Body Corporate Lat activating chimeric antigen receptor t cells and methods of use thereof
WO2023015217A1 (en) 2021-08-04 2023-02-09 Sana Biotechnology, Inc. Use of cd4-targeted viral vectors
WO2023105000A1 (en) 2021-12-09 2023-06-15 Zygosity Limited Vector
WO2023115039A2 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae fusion glycoproteins
WO2023115041A1 (en) 2021-12-17 2023-06-22 Sana Biotechnology, Inc. Modified paramyxoviridae attachment glycoproteins
WO2023126458A1 (en) 2021-12-28 2023-07-06 Mnemo Therapeutics Immune cells with inactivated suv39h1 and modified tcr
WO2023139269A1 (en) 2022-01-21 2023-07-27 Mnemo Therapeutics Modulation of suv39h1 expression by rnas
WO2023147515A1 (en) 2022-01-28 2023-08-03 Juno Therapeutics, Inc. Methods of manufacturing cellular compositions
WO2023150518A1 (en) 2022-02-01 2023-08-10 Sana Biotechnology, Inc. Cd3-targeted lentiviral vectors and uses thereof
WO2023178348A1 (en) 2022-03-18 2023-09-21 The Regents Of The University Of Colorado, A Body Corporate Genetically engineered t-cell co-receptors and methods of use thereof
WO2023180552A1 (en) 2022-03-24 2023-09-28 Institut Curie Immunotherapy targeting tumor transposable element derived neoantigenic peptides in glioblastoma
WO2023193015A1 (en) 2022-04-01 2023-10-05 Sana Biotechnology, Inc. Cytokine receptor agonist and viral vector combination therapies
WO2023196933A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Chimeric antigen receptor t cells and methods of use thereof
WO2023196921A1 (en) 2022-04-06 2023-10-12 The Regents Of The University Of Colorado, A Body Corporate Granzyme expressing t cells and methods of use
WO2023211972A1 (en) 2022-04-28 2023-11-02 Medical University Of South Carolina Chimeric antigen receptor modified regulatory t cells for treating cancer
WO2023213969A1 (en) 2022-05-05 2023-11-09 Juno Therapeutics Gmbh Viral-binding protein and related reagents, articles, and methods of use
WO2023220655A1 (en) 2022-05-11 2023-11-16 Celgene Corporation Methods to overcome drug resistance by re-sensitizing cancer cells to treatment with a prior therapy via treatment with a t cell therapy
WO2023222928A2 (en) 2022-05-20 2023-11-23 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
EP4279085A1 (en) 2022-05-20 2023-11-22 Mnemo Therapeutics Compositions and methods for treating a refractory or relapsed cancer or a chronic infectious disease
WO2023230581A1 (en) 2022-05-25 2023-11-30 Celgene Corporation Methods of manufacturing t cell therapies
WO2023250400A1 (en) 2022-06-22 2023-12-28 Juno Therapeutics, Inc. Treatment methods for second line therapy of cd19-targeted car t cells
WO2024006960A1 (en) 2022-06-29 2024-01-04 Juno Therapeutics, Inc. Lipid nanoparticles for delivery of nucleic acids
WO2024044779A2 (en) 2022-08-26 2024-02-29 Juno Therapeutics, Inc. Antibodies and chimeric antigen receptors specific for delta-like ligand 3 (dll3)
WO2024054944A1 (en) 2022-09-08 2024-03-14 Juno Therapeutics, Inc. Combination of a t cell therapy and continuous or intermittent dgk inhibitor dosing
WO2024062138A1 (en) 2022-09-23 2024-03-28 Mnemo Therapeutics Immune cells comprising a modified suv39h1 gene
WO2024081820A1 (en) 2022-10-13 2024-04-18 Sana Biotechnology, Inc. Viral particles targeting hematopoietic stem cells

Also Published As

Publication number Publication date
EP1109921A4 (en) 2002-08-28
EP1109921A1 (en) 2001-06-27
CA2343156A1 (en) 2000-03-16
JP2002524081A (en) 2002-08-06

Similar Documents

Publication Publication Date Title
WO2000014257A1 (en) Fusion receptors specific for prostate-specific membrane antigen and uses thereof
US20210277114A1 (en) Car based immunotherapy
US20210077532A1 (en) Modified Cell Expansion and Uses Thereof
EP3586852B1 (en) Modified cell expansion and uses thereof
US20210137980A1 (en) Prostate-specific membrane antigen cars and methods of use thereof
US10561686B2 (en) Modified cell expansion and uses thereof
US20240041928A1 (en) Genetically Modified Immune Cells Targeting NY-ESO-1 and Methods of Use Thereof
US7446190B2 (en) Nucleic acids encoding chimeric T cell receptors
Wang et al. AT cell-independent antitumor response in mice with bone marrow cells retrovirally transduced with an antibody/Fc-γ chain chimeric receptor gene recognizing a human ovarian cancer antigen
US20160235787A1 (en) Epitope Spreading Associated with CAR T-Cells
US20130071414A1 (en) Engineered cd19-specific t lymphocytes that coexpress il-15 and an inducible caspase-9 based suicide gene for the treatment of b-cell malignancies
US20230265154A1 (en) Universal chimeric antigen receptor t-cell preparation technique
CA2978171A1 (en) T-cell receptors directed against the preferentially expressed antigen of melanoma and uses thereof
US20240024476A1 (en) CAR Cells and Polyspecific Binding Molecules for Treating Solid Tumor
US20200332004A1 (en) Method for improving production of car t cells
US11273178B2 (en) Affinity maturated T cell receptors and use thereof
AU687271B2 (en) Targeted T lymphocytes
CN110819596A (en) Modified cells with enhanced migratory capacity
US20040260061A1 (en) Continuous, normal human t-lymphocyte cell lines comprising a recombinant immune receptor with defined antigen specificity
TW201927812A (en) Pharmaceutical chimeric receptor composition and method thereof
EP4305054A1 (en) Selective stimulation of t cells in solid tumors using oncolytic viral delivery of orthogonal il-2
Tong et al. Chimeric antigen receptor (CAR) T cells therapies for hematological malignancy
GB2569692A (en) T cell antigen receptor chimera

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2343156

Country of ref document: CA

Ref country code: CA

Ref document number: 2343156

Kind code of ref document: A

Format of ref document f/p: F

ENP Entry into the national phase

Ref country code: JP

Ref document number: 2000 568998

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: 1999945508

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09786502

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1999945508

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1999945508

Country of ref document: EP