WO2007103009A2 - CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES - Google Patents

CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES Download PDF

Info

Publication number
WO2007103009A2
WO2007103009A2 PCT/US2007/004841 US2007004841W WO2007103009A2 WO 2007103009 A2 WO2007103009 A2 WO 2007103009A2 US 2007004841 W US2007004841 W US 2007004841W WO 2007103009 A2 WO2007103009 A2 WO 2007103009A2
Authority
WO
WIPO (PCT)
Prior art keywords
cells
cell
seq
peptide
melanoma
Prior art date
Application number
PCT/US2007/004841
Other languages
French (fr)
Other versions
WO2007103009A3 (en
Inventor
Zeling Cai
Ann Moriarty
Per A. Peterson
Jon M. Richards
Original Assignee
Janssen Pharmaceutica N.V.
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 Janssen Pharmaceutica N.V. filed Critical Janssen Pharmaceutica N.V.
Priority to AU2007222080A priority Critical patent/AU2007222080B2/en
Priority to MX2008011302A priority patent/MX2008011302A/en
Priority to US12/281,197 priority patent/US7993638B2/en
Priority to EP07751592.2A priority patent/EP1996232B1/en
Priority to JP2008557314A priority patent/JP2009531303A/en
Priority to CA2643337A priority patent/CA2643337C/en
Priority to BRPI0708446-3A priority patent/BRPI0708446A2/en
Priority to ES07751592.2T priority patent/ES2579762T3/en
Publication of WO2007103009A2 publication Critical patent/WO2007103009A2/en
Priority to IL193710A priority patent/IL193710A/en
Publication of WO2007103009A3 publication Critical patent/WO2007103009A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/20Interleukins [IL]
    • A61K38/2013IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4621Cellular immunotherapy characterized by the effect or the function of the cells immunosuppressive or immunotolerising
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/46433Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464454Enzymes
    • A61K39/464456Tyrosinase or tyrosinase related proteinases [TRP-1 or TRP-2]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/46449Melanoma antigens
    • A61K39/464491Melan-A/MART
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/46449Melanoma antigens
    • A61K39/464492Glycoprotein 100 [Gp100]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55522Cytokines; Lymphokines; Interferons
    • A61K2039/55527Interleukins
    • A61K2039/55533IL-2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/57Skin; melanoma

Definitions

  • the invention relates to methods for treating cancer in patients involving the administration of activated cytotoxic T lymphocytes, cytokines such as IL-2 and IFN- alpha-2b, and cladribine or denileukin diftitox as a lymphodepleting agent.
  • cytotoxic T lymphocytes such as IL-2 and IFN- alpha-2b
  • cladribine or denileukin diftitox as a lymphodepleting agent.
  • T-cell defined tumor antigens in melanoma has led to clinical trials that target cancer cells by attempting to augment the antigen-specific cellular immune response.
  • This approach has been pursued in numerous vaccination strategies in which the antigens are delivered in an immunogenic context in an attempt to induce potent T cell responses in vivo.
  • the magnitude of the induced T-cell response has generally been low, or undetectable and correlated poorly with clinical responses.
  • Immunization of melanoma patients with cancer antigens may increase the number of circulating CTL precursors; however it has not correlated with clinical tumor regression, suggesting a defect in function or activation in vivo.
  • Adoptive T cell therapy involves the removal of T cells from the host environment where tolerogenic mechanisms are active in vivo in cancer patients and contributes to the ineffective responses demonstrated in this patient population.
  • CD8 + T cells may be stimulated ex vivo to generate antigen-specific CTLs (see, e.g., U.S. Patent No. 6,225,042).
  • Early adoptive immunotherapy approaches used activated lymphocytes as a treatment for various cancers. 14 Initially, lymphokine-activated killer cells (LAK), and later tumor- infiltrating lymphocytes (TIL), activated ex vivo with EL-2, were used, but the demonstration of efficacy was equivocal.
  • LAK lymphokine-activated killer cells
  • TIL tumor- infiltrating lymphocytes
  • Cytotolytic CD8 + T cells are a major line of defense against viral infections.
  • CD8 + lymphocytes specifically recognize and lyse host cells that are infected with a virus.
  • it would be desirable to harness the cytotoxic activity of CTLs few in vitro/ex vivo procedures have been available to specifically activate CTLs.
  • the identification of key melanoma-associated antigens and a method for specific in vitro activation of CTLs allows for an efficient evaluation of adoptive immunotherapy for metastatic melanoma.
  • APCs antigen presenting cells
  • APCs naturally occurring antigen presenting cells
  • the efficiency of activation is low since the MHC Class I molecules of native APCs contain many other peptide epitopes, thus allowing minimal presentation of tumor-associated peptide epitopes.
  • Most of these presented peptides represent normal, innocuous endogenous proteins.
  • a more direct approach to this problem would be to activate CD8 + T cells specifically to those epitopes relevant to combating the disease, in this particular case, melanoma-associated antigens.
  • Figure 1 illustrates cytolytic activity of CTLs generated ex vivo directed against peptide-loaded target cells.
  • CDS + T cells isolated from a melanoma patient were immunized in vitro with Drosophila APCs loaded with five different melanoma-associated peptide epitopes.
  • the activated CD8 + T cells were cultured in vitro using IL-2 and IL-7 to selectively expand the melanoma-specific CTLs. Activity was assessed as specific lysis of 5!
  • Figure 2 provides a schematic illustration of two preferred embodiments of treatment regimens according to the invention along with a control regimen.
  • the terms "including”, “comprising”, and “containing” are used herein in their open, non-limiting sense.
  • the therapeutic regimens of the invention which comprise administering CTLs that have been activated by contacting xAPCs loaded with selected peptide in conjunction with cytokines and at least one lymphodepleting agent selected from cladribine and DAB-IL2 may be employed to treat cancer in a subject in need of such treatment.
  • the cancer is selected from malignant melanoma, multiple myeloma, prostate cancer, lymphoma, non-Hodgkin's lymphoma, leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, Burkitt's lymphoma, thyroid cancer, uterine cancer, kidney cancer, ovarian cancer, lung cancer, breast cancer, liver cancer, pancreatic cancer, prostate cancer, colon cancer, skin cancer, stomach cancer, and cervical cancer.
  • the invention provides an adoptive CTL therapy regimen for treating or ameliorating a metastatic melanoma comprising: obtaining na ⁇ ve CD8+ T cells from a subject; contacting the na ⁇ ve CD8+ T cells with xenogenic antigen presenting cells (xAPCs) that have been loaded with selected peptide antigen, thereby generating activated CTLs that target cells expressing the selected peptide antigen; administering the activated CTLs back to the subject; administering at least one lymphodepleting agent selected from cladribine and DAB-IL2; and administering at least two cytokines that effect CTL persistence.
  • xAPCs xenogenic antigen presenting cells
  • the adoptive CTL therapy regimen preferably employs autologous CD8 + T cells that are activated ex vivo, which when activated and administered to a subject in accordance with the invention are capable of in vivo destruction of tumor cells bearing cancer-associated antigenic epitopes,
  • a subject in this context refers to a mammalian patient in need of treatment for a cancer.
  • a subject may be a human diagnosed with a melanoma, such as an advanced malignant metastatic melanoma, e.g., a patient who has been diagnosed to be HLA- A2 positive and have Stage III/IV unresectable disease.
  • the CTL agent is prepared from xAPCs.
  • xAPCs xenogenic antigen presenting cells
  • xAPCs xenogenic antigen presenting cells
  • xAPCs may include the following components: an exogenous MHC I molecule; one or more exogenous assisting molecules to assist in the activation of na ⁇ ve T cells; and host cells capable of expressing the exogenous molecules their surface.
  • the exogenous molecules are encoded by xenogenic nucleic acid that has been introduced into host cells.
  • the xAPCs preferably also express exogenous co- stimulator and adhesion molecules, which potentiate T cell activation capability of the xAPCs.
  • the host cells are insect cells, more preferably Drosophila cells, such as Schneider 2 (S2) cells.
  • S2 Schneider 2
  • the xAPCs may be loaded with peptide antigen by a variety of methods known or available in the art.
  • Peptides are selected that are capable of binding to the empty MHC Class I molecules.
  • Selected peptides preferably correspond to epitopes comprising antigenic or immunogenic amino acid sequences derived from protein expressed on the surface of cells, which will serve as targets for the T cells employed in the adoptive CTL therapy.
  • one or more antigenic or immunogenic peptide species that binds to such empty MHC Class I molecules may be contacted with the xAPCs under suitable conditions for the binding to occur.
  • One or more antigenic or immunogenic peptide species may be selected. If more than one species is selected, they may be contacted with the xAPCs simultaneously or at distinct instances, resulting in multi-antigenic or multi-immunogenic MHC-peptide complexes produced on the xAPCs.
  • Loading of the selected peptide onto empty MHC molecules preferably occurs under conditions that approximate biological binding conditions, which may be approximated in vitro, ex vivo, or in vivo.
  • the artisan may consider one or more factors such as thermodynamic, electrostatic, energetic, and entropic considerations, as well as specific amino acids within selected peptides that are required for efficacious binding to MHC molecules.
  • Preferred peptides include, for example, the peptides corresponding to amino acid sequences selected from a tyrosinase protein, a gplOO protein, and a MART-I protein.
  • Other preferred peptides include YMNGTMSQV (SEQ ID NO:1), YMDGTMSQV (SEQ ID NO:2), AAGIGILTV (SEQ ID NO:3), ITDQVPFSV (SEQ ID NO:4), YLEPGPVTA (SEQ ID NO:5), and KTWGQYWQV (SEQ ID NO:6).
  • Additional exemplary peptides that maybe selected include, for example, the following amino acid sequences, where the protein from which each peptide is derived is noted parenthetically: SILSLKEAST (C- Lectin; SEQ ID NO:1), KMASRSMRL (C-Lectin; SEQ ID NO:2 ), ALALAALLW (Pec 60; SEQ ID NO: 3), ALLWDREV (Pec60; SEQ ID NO: 4), YMNGTMSQV (Tyrosinase; SEQ ID NO:5), YMDGTMSQV (Tyrosinase; SEQ ID NO:6), ITDQVPFSV (gplOO; SEQ ID NO:7), YLEPGPVTA (gplOO; SEQ ID NO:8), AAGIGILTV (MART-I; SEQ ID NO:9), ELAGIGILTV (MART-I; SEQ ID NO: 10), CLTSTVQLV (Her-2/neu; SEQ ID NO
  • Selected peptides may be presented to the cells via a variety of means and methods in the art. Selected peptides may be presented in a manner that allows them to enter an intracellular pool of peptides. For example, peptides may be presented via osmotic loading. Preferably, peptides are added to the xAPC system culture medium. The peptides may be added to the culture medium in the form of an intact polypeptide or protein that is subsequently degraded via cellular processes, such as, for example, enzymatic degradation.
  • the intact polypeptide or protein may be degraded via some other means such as chemical digestion (e.g., cyanogen bromide) or proteases (e.g., trypsin and chymotrypsin) prior to addition to the xAPC system culture medium.
  • an entire protein or polypeptide sequence may be cloned into an appropriate vector and inserted into a prokaryotic cell, whereby the cell generates significant amounts of the antigenic polypeptide that are then harvested, purified, and digested into peptides that are then added to the xAPC system culture medium.
  • a sufficient amount of each selected peptide may be added to the cell culture to allow the Class I MHC molecules to bind and subsequently present a large density of the peptide on the surface of human Class I MHC-expressing cells.
  • the xAPCs may be assayed for enhanced APC function compared to the APC function of nonxenogenic or endogenous antigen presenting cells.
  • Enhanced APC function may be determined by measuring any of a variety of parameters of CD8 + T cell activation, such as, for example, a degree of one or more cell surface proteins expression, which is indicative of CD8 + T cell activation, such as CD69 cell surface expression, a degree of differentiation, a degree of cyotoxic killing ability, a degree of specific cell lysis, and a degree cytokine production.
  • PBLs Peripheral blood leukocytes
  • Methods for purification of PBLs include, methods emplying Ficoll gradients may be utilized for this purpose.
  • PBLs are then mixed with xAPCs cells preincubated with the appropriate antigenic peptides.
  • PBLs are purified by magnetic bead purification systems that are in the art, such as Miltenyi beads (Myltenyi Biotec) and Dynabead systems (Dynal Biotech).
  • PBLs may also be purified via cell sorting procedures, such as with fluorescence assisted cell sorter (FACS)-based methods, or other appropriate cell sorting devices and methodology.
  • FACS fluorescence assisted cell sorter
  • GFP green fluorescent protein
  • Naive T cells are incubated in culture with the appropriate xAPCs and loaded with selected peptide for a time period sufficient to activate and further enrich for a population of CD8 + cells.
  • U.S. Patent No. 4,690,915 describes a method of obtaining large numbers of lymphocytes via lymphocytopheresis.
  • the CD8 + cells are activated in an antigen-specific manner.
  • the ratio of resting or precursor CD8 (effector) cells to antigen-presenting cells may vary from individual to individual and may further depend upon variables such as the amenability of an individual's lymphocytes to culturing conditions and the nature and severity of the cancer.
  • the lymphocyte:antigen-presenting cell (e.g., Drosophila cell) ratio is preferably in the range of about 30:1 to 300:1.
  • the lymphocyte:antigen-presenting cell e.g., Drosophila cell
  • 3xlO 7 human PBL and IxIO 6 live Drosophila cells are admixed and maintained in 20 ml of RPMI 1640 culture medium.
  • the effector/antigen-presenting culture may be maintained for as long a time as is necessary to activate and enrich for a population of a therapeutically useable or effective number of CD8 + cells.
  • a maximum specific CD8 + activation level generally being observed after one to ten days of culture, e.g., after five days of culture, a preferred time is from about three to seven days.
  • in vitro activation of CD8 + T cells may be detected within a brief period of time after transfection of a cell line.
  • transient expression in a transfected cell line capable of activating CD8 + T cells is detectable within 48 hours of transfection.
  • Activated cytotoxic T lymphocytes may be effectively separated from the xAPCs (e.g., Drosophila cells) using a suitable method known or available in the art.
  • xAPCs e.g., Drosophila cells
  • monoclonal antibodies specific for the artificial APCs, for the peptides loaded onto the artificial APCs, or for the CTLs (or a segment thereof) may be utilized to bind their appropriate complementary ligand.
  • Antibody-tagged cells may then be extracted from the stimulator-effector cell admixture by any of a variety of methods in the art, such as, for example, immunoprecipitation and immunoassay methods.
  • a separation step may be omitted completely and the inactivated xAPCs may be left in culture with the activated CTLs.
  • Therapeutically effective, cytotoxic amounts of the activated CTLs may be selected as suitable for the described in vitro and in vivo use, e.g., in view of the amount and type of cells that are the ultimate target of these CTL cells. The amount will also be selected in view of the condition of the patient and maybe determined via consideration of all appropriate factors by the practitioner.
  • about IxIO 6 to about IxIO 12 , more preferably about 1x10 8 to about 1 x 10 ⁇ , and even more preferably, about IxIO 9 to about IxIO 10 , activated CD8 + cells are utilized for adult humans, compared to about 5xlO 6 -5x10 7 cells used in mice.
  • the activated CD8 + cells which are CTLs
  • the cell culture system is preferably not tumorigenic. Therefore, if complete separation of Drosophila cells and activated CD8 + cells is not achieved, there should be no inherent danger associated with the administration of a small number of Drosophila cells.
  • Na ⁇ ve CD4 + T cells or CD8 + T cells, or both CD4 + T cells and CD8 + T, cells may be preferably extracted from a subject prior to incubation with the xAPC cultures.
  • Subjects may undergo any of a variety of known or available blood cell separation procedures (e.g., leukopheresis) to collect white blood cells.
  • Na ⁇ ve T cells may be harvested from a subject prior to the initiation of other treatment or therapy that may interfere with, attenuate, or in any way limit the specific activation of na ⁇ ve T cells methods and uses as provided in the present invention.
  • a sample of na ⁇ ve T cells may be obtained prior to the initiation of chemotherapy or radiation treatment and kept in culture.
  • the na ⁇ ve T cells may be expanded and activated, and the activated CTLs may be introduced back into the subject.
  • naive T cells may be activated, and the activated CTLs may be introduced back into the subject from whom the na ⁇ ve T cells were obtained before, after, or in conjunction with other optional forms of treatment, such as chemotherapy or radiation.
  • the activated CTLs may also be suspended into an appropriate vehicle for delivery and infusion into a subject.
  • Techniques of reintroducing cellular components are known in the art and include procedures such as those exemplified in U.S. Pat. Nos. 4,844,893 and 4,690,915.
  • administration of activated CTLs cells via intravenous infusion may be employed. Multiple infusions may be required, and these infusions may occur over a period of several weeks or longer.
  • cytokines are also administered to attain CTL persistence and thereby enhance half-life, activity, potency, and/or selectivity properties of the CTLs that are administered to a subject. Such persistence may result from a direct effect on the CTLs or from an indirect effect that involves upregulation of antigen expression on target cells of the CTLs.
  • Preferred cytokines are IFN- ⁇ -2b and IL-2.
  • Cladribine (2-CdA, Leustatin®) and/or DAB IL-2 (ONTAK®) lymphodepleting agents are also administered in the inventive treatment regimens. These agents are non- myeloablative and elicit a transient immunosuppression in subjects receiving the CTL therapy.
  • each of the CTL, cytokine, and lymphodepleting agents may be selected by the artisan based on routine experimentation and the guidance herein, including the examples below.
  • Xenogenic APC (xAPC) lines are generated from Schneider S2 cells (S2 cells), which were established in 1969 from several hundred 20- to 24-hr old Oregon-R (wild type) Drosophila melanogaster (Oregon-R) embryos (ATCC CRL-1963), according to published procedures (Schneider, J. Embryol. Morph. 27:353-365,1972).
  • S2 cells Schneider S2 cells
  • the S2 cell line has been deposited with the American Type Culture Collection (CRLl 0974). The original supply of S2 cells used to derive the cell lines from which the xAPCs derived are obtained from this source.
  • S2 cells are transfected with vectors derived from plasmid vector pRMHa-3 (see, e.g., U.S. Patent No. 6,225,042).
  • One xAPC line designated clone A
  • a second xAPC line designated clone B
  • clone B is transfected with vectors encoding HLA-A2.1 Class I, B7.1, B7.2, ICAM-I and LFA-3.
  • a third xAPC cell line designated clone C, is transfected with vectors encoding HLA-A2.1 Class I 5 B7.1, ICAM-I, LFA-3, and CD70.
  • clone A expresses HLA-A2, B7.1, and ICAM-I
  • clone B expresses HLA-A2.1 Class I, B7.1, B7.2, ICAM-I and LFA-3
  • clone C expresses HLA- A2.1 Class I, B7.1, ICAM-I, LFA-3, and CD70.B7.2 and LFA-3.
  • Complete Drosophi Ia-SFM medium without G418 is added to bring the volume up to 15 ml/flask. Flasks are then incubated in a chamber at approximately 27°C. On approximately day -1 to —3, cells are then induced by addition of copper sulfate (CuSO 4 ) to a final concentration of 1.0 mM (1:200 dilution of 200 mM stock OfCuSO 4 ; 75 ⁇ l OfCuSO 4 for each T75 flask containing 15 ml of cell suspension) and returned to the 27°C chamber. The induction time lasts for approximately 24 to 72 hours.
  • CuSO 4 copper sulfate
  • flasks containing induced cell cultures are checked visually and microscopically for evidence of contamination. Uncontaminated flasks are pooled and viable cells counted. Samples of pooled cell cultures of approximately 6x10 6 cells are evaluated by flow cytometry using fluorescence assisted cell sorter (FACS) analysis to determine the level of expression of exogenous molecules. Cell cultures (approximately 1 xlO 7 cells/mL) are then tested to verify expression exogenous HLA-A2.1, B7.1 and ICAM-I (for clone A cells) or HLA-A2.1, B7.1, B7.2, ICAM-I and LFA-3 (for clone B cells) prior to peptide loading.
  • FACS fluorescence assisted cell sorter
  • each cell culture is washed by splitting each culture into two sterile 50 ml conical tubes. Each tube is then filled with HYQ SFX-Insect medium (Hyclone) and centrifuged at 1,700 rpm (600 x g) for approximately seven minutes to pellet the cells. Supernatants are discarded, and the tubes containing cell pellet are again centrifuged at 1,700 rpm (600 x g) for approximately one minute. Supematants are removed with a fine tipped pipette. Pellets from each split cell culture are then recombined and resuspended in 8 mL of SFX Insect medium to a cell density of approximately 1 x10 cell/mL.
  • HYQ SFX-Insect medium Hyclone
  • each cell culture suspension contains ⁇ 2 microglobulin at a final concentration of approximately 5 ⁇ g/mL and each selected peptide to be loaded onto xAPCs at a final concentration of approximately 0.1 ⁇ g/mL per peptide.
  • Cell cultures are incubated in the suspension containing ⁇ 2 microglobulin and peptides for at least four hours, and no more than eight hours, with swirling every 30 minutes at room temperature. After the peptide incubation period, approximately 1 mL aliquots of each cell culture are distributed separately into eight polypropylene tubes (Falcon 2006). Any residual cell culture suspension is discarded.
  • CD8 cells isolated from leukapheresis samples by positive selection with an anti- CD8 antibody are stimulated against human melanoma-associated peptides (tyrosinase-689.369-377. tyrosinase-792369-377 5 MART- 1-81927-35, gpl00-817209-2i7, gplOO- 8I8280-288 and gp 100-853154.162) presented by Drosophila cells expressing human Class I and co-stimulation/adhesion molecules HLA- A2.1 , B7.1 , CD70 , LFA-3 and ICAM- 1.
  • CD8 + cells are restimulated for two rounds with autologous monocytes (pulsed with the described epitopes) in the presence of IL-2 and IL-7.
  • the number of effector cells is then increased by non-specific stimulation with anti-CD3 monoclonal antibody in the presence of gamma-irradiated autologous feeder cells and IL-2.
  • Cytotoxic T lymphocyte activity is measured against peptide-loaded T2 cells and a panel of melanoma cells, while the purity of in v ⁇ ro-stimulated CD8 + T cells is assessed by flow cytometry. Materials.
  • Reagents rhIL-7 Recombinant human interleukin-7 (IL-7) is a lymphokine produced in E. coli and purified by the supplier (PeproTech) using high performance liquid chromatography (HPLC) but not antibody. IL-7, received as a powder, is diluted in sterile DPBS containing 1% human serum albumin. The bulk solution is then filtered through a 0.2- ⁇ m filter, aliquoted (30,000 U/mL, IOOOX concentration) into sterile vials, and stored at -80 0 C prior to use. rhIL-2. Recombinant human interleukin-2 (IL-2) is produced by recombinant DNA technology and supplied by Chiron Corporation (Proleukin ® ).
  • IL-2 received as a powder, is diluted in IL-2 diluent (0.5% human serum albumin in 50 mM acetic acid), filtered through a 0.2- ⁇ m filter, aliquoted into sterile vials (20,000 U/mL, 1000 X concentration), and then stored at -80 0 C prior to use.
  • Tyrosinase PeptideYMNGTMSQV A tyrosinase peptide (tyr 369-377), corresponding to amino acids 369-377 of human tyrosinase, is manufactured and purified using GLP compliance standards (Synpep Corporation). The peptide powder as received from the manufacturer (Synpep Corporation) is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0 C to —88 0 C prior to use.
  • DMSO dimethylsulfoxide
  • This stock peptide solution is mixed in equal parts with other peptide stock solutions (also at a concentration of 10 mg/ml) to generate combination peptide solutions for use in loading xAPCs.
  • the combination peptide solutions are aliquoted into sterile vials in a Class 10,000 clean room under aseptic conditions in a Class II biosafety cabinet.
  • Tyrosinase PeptideYMDGTMSQV A deamidated form of the tyr 369-377 peptide described above, which contains an aspartic acid residue in place of an asparagine residue at position three of the peptide, is manufactured and purified using GLP compliance standards (Synpep Corporation). This deamidation form is called tyr 369- 377d.
  • the peptide powder received from the manufacturer is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0 C to -88 0 C prior to use.
  • DMSO dimethylsulfoxide
  • a gplOO peptide (gpl00 20 ⁇ >-2i7), corresponding to amino acids 209-217 of human gp-100, is manufactured and purified using GLP compliance standards (Synpep Corporation).
  • the peptide powder is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0 C to —88 0 C prior to use.
  • DMSO dimethylsulfoxide
  • KTWGQYWQV A gplOO peptide (gp 100154- 1 62), corresponding to amino acids 154-162 of human gplOO, is manufactured and purified using GLP compliance standards.
  • the peptide powder as received from Synpep Corporation is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0 C to -88 °C prior to use.
  • DMSO dimethylsulfoxide
  • gplOO Peptide YLEPGVTA A gplOO peptide (gplOO 28 o-288), corresponding to amino acids 280-288 of human gplOO, is manufactured and purified using GLP compliance standards by Synpep Corporation.
  • the peptide powder is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0 C to -88 0 C prior to use.
  • MART-I Peptide AAGIGILTV A MART-I peptide (MART-127-35), corresponding to amino acids 27- 35 of human MART-I, is manufactured and purified using GLP compliance (Synpep Corporation). The peptide powder is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0 C to -88 0 C prior to use.
  • DMSO dimethylsulfoxide
  • DYNABEADS ® M-450 are sterile paramagnetic beads coated with polyclonal sheep anti-mouse IgG that bind the primary mouse IgG.
  • DYNABEADS available from Baxter Oncology Inc., are stored at 4 0 C prior to use in T cell isolation using the Isolex 300i Magnetic Cell Selector.
  • Human Serum Albumin 25 % HSA, USP (Baxter Fenwal Laboratories; the plasma source for each lot tested to be negative for HIV-I HIV-2, HCV, and HBV), is stored at RT prior to use as a source of buffered protein during the following T cell preparation and activation steps steps: purification of CD8 + T cells and CD8 ' T cells; peptide-loading of adherent cells; and final formulation of activated T cells.
  • Anti-CD8 monoclonal antibody (37B1A) is a murine monoclonal antibody directed against the CD8 antigen of T cells, which is used to select CD8 + T cells with the Isolex 300i Magnetic Cell Selector System.
  • the concentrate solution is diluted in sterile DPBS for use in CDS + T cell isolation or activation processes.
  • the bulk solution is filtered through a 0.2- ⁇ m filter and then aliquoted into single-use vials in a Class 10,000 clean room under aseptic conditions in a Class II biosafety cabinet. Aliquots (10.0 mg/mL) are stored at -80 0 C prior to use.
  • CD8 Alpha Chain Peptide - AAEGLDTQRFSG CD8 alpha light chain peptide (AAEGLDTQRFSG) is purified and manufactured under GLP compliance standards.
  • the CD8 alpha chain peptide is used in CD8 + T cell isolation processes to release CD8 + T cells captured using CD8 (37B1A) antibody and the Isolex 30Oi Magnetic Cell Selector.
  • Each lot of peptide is manufactured by Synpep Corporation to meet pharmaceutical grade standards, and is tested for peptide sequence, purity, molecular weight, and appearance.
  • the CD8 alpha chain peptide, received as a powder, is further processed to create a stock solution of 10 mg/ml.
  • This stock solution is diluted in DPBS, filtered through a 0.2- ⁇ m filter, aliquoted into sterile vials, and stored at -72 0 C to -88 0 C prior to use.
  • Viali ⁇ g of the peptide reagent is performed in a Class 10,000 clean room under aseptic conditions in a Class II biosafety cabinet.
  • Human ⁇ 2M Microglobulin A concentrate of human beta— 2 microglobulin ( ⁇ 2M) produced by recombinant DNA technology is diluted in sterile DPBS to achieve a concentration of 1.0 mg/mL. The bulk solution is then filtered through a 0.2- ⁇ m filter, aliquoted into sterile vials and stored at -8O 0 C prior to use during the preparation and peptide-loading of xAPCs and peptide loading of adherent cells.
  • ⁇ 2M beta— 2 microglobulin
  • Sodium Citrate Solution A sterile, nonpyrogenic anticoagulant sodium citrate solution, USP (Baxter Fenwal), is stored at room temperature (RT) prior to use as a buffer additive for running the Isolex 30Oi Magnetic Cell Selector for selection of CD8 + T cells and CD8 " T cells.
  • RT room temperature
  • Schneider's Drosophila medium is a culture medium used for culturing Drosophila cells. Each lot of medium is tested by the supplier (Invitrogen Corporation) for osmolality, pH, sterility, and the ability to sustain the growth of Drosophila cells.
  • Schneider's Drosophila medium (IX concentration) is stored at 2 0 C to 6°C prior to use.
  • Fetal Bovine Serum Fetal bovine serum (FBS), which is used as a protein source for the growth of host cells or xAPCs cells, is stored at — 80 0 C.
  • the FBS available from Gemini Bioproducts, is processed from bovine fetal blood from animals of United States origin. The maternal animals from which the blood is derived are free of infectious and contagious diseases and injurious parasites.
  • Hyclone's SFX Insect Medium (Hyclone Corporation) is a serum- free culture medium (IX concentration) used during the peptide loading of xAPCs, and is stored at 2 0 C to 6 0 C prior to use. This medium does not contain products of bovine origin.
  • Copper Sulfate Copper (II) sulfate pentahydrate (Aldrich) is used for induction of modified host cells to express human HLA, co-stimulatory, and adhesion molecules.
  • the stock solution is formulated by dissolving the crystalline CuSO 4 in endo toxin-free sterile water to achieve a concentration of 200 mM and aseptically filtering the solution through a 0.2- ⁇ m filter into a sterile container in a Class II biosafety cabinet.
  • the filtered stock solution is stored at 2 0 C to 6 0 C prior to use.
  • RPMI RPMI culture medium
  • IX concentration serum- and antibiotic-free
  • Dulbecco 's Phosphate Buffered Saline DPBS
  • Dulbecco's Phosphate Buffered Saline DPBS
  • DPBS Dulbecco's Phosphate Buffered Saline
  • IX concentration available from Invitrogen Corporation or Gibco, IX concentration
  • DPBS is used for the following procedures: running the Isolex 300i Magnetic Cell Selector instrument during the selection of CDS + T cells and CD8 " T cells; washing non-adherent cells during restimulation steps and washing unbound OKT3 monoclonal antibody during non-specific expansion; and diluting human ⁇ 2 microglobulin, IL-7, CD8 peptide, and OKT3.
  • Leibovitz's L-15 medium (without L-glutamine; IX concentration), available from Sigma- Aldrich, is stored at 2 0 C to 6 0 C prior to use during peptide-loading of the T cell activation process.
  • Orthoclone OKT ® 3 (l.Omg/mL), a murine monoclonal antibody specific for the CD3 antigen of T cells supplied in ampoules as a sterile solution approved for clinical use (available from Ortho), is aliquoted into single-use vials under sterile conditions and stored frozen at— 80°C prior to use in the activation of T cells.
  • Geneticin (G418). Geneticin (Invitrogen Corporation) is a selective antibiotic used in the culture of Drosophila cells for maintaining expression of exogenous molecules encoded by xenogenic nucleic acid. Geneticin is supplied as a sterile stock solution (50 mg/mL), and is stored at 2 to 6 °C prior to use.
  • Calcium Chloride Calcium chloride hydrate is used for clotting of autologous plasma obtained from the lymphopheresis product to generate autologous serum used in CD8 + T cell isolation or activation processes. Calcium chloride hydrate is received as a crystalline powder, is compounded into a stock solution (IM), and stored at 2 °C to 6 0 C prior to use. The stock solution is formulated by dissolving calcium chloride in endotoxin-free sterile water and aseptically filtering through a 0.2- ⁇ m filter into a sterile container in a Class II biosafety cabinet.
  • Acetic Acid Acetic acid (17.4M) used for the preparation of stock solutions of IL-2 is obtained from Sigma Corporation and stored at RT prior to use.
  • FICOLL-PAQUE ® Plus Following isolation of CD8 + T cells and CD8 " T cells with the Isolex 300i Cell Selector System, mononuclear cells from the non- CD8 + fraction are further fractionated using FICOLL-PAQUE ® Plus (IX concentration), a Ficoll gradient reagent without any animal components available from Amersham Pharmacia Biotech used to remove dead cells, neutrophils, and red blood cells. The reagent is stored at RT prior to use.
  • PENTASPAN® is a sterile solution of 10% pentastarch in 0.9% sodium chloride for clinical use (NDC 0264-1972-10), and is stored at RT prior to use. It is used (IX concentration) as a cryoprotectant in the cryopreservation of isolated CD8 " T cells and CD8 T cells.
  • DMSO Dimethyl Sulfoxide
  • IX concentration available from Sigma- Aldrich, is stored at RT prior to use.
  • L-Glutamine L-Glutamine (USP), 20OmM (10OX concentration), available from Invitrogen Corporation, is used as an RPMI culture medium supplement, and is stored at -8O 0 C prior to use.
  • MEM Sodium Pyruvate Solution MEM sodium pyruvate solution (10OmM, IOOX concentration), available from Invitrogen Corporation, is used to supplement RPMI medium, and is stored at 2 to 6 0 C prior to use.
  • Non-Essential Amino-Acids Non-Essential Amino-Acids.
  • Non-essential amino-acids l OmM; IOOX concentration
  • X-vivo 10-Cell Medium X-vivo 10-cell culture medium, supplied by BioWhittaker, is stored at 2 0 C to 6 0 C prior to use. This medium (IX concentration), which is serum-, phenol red-, and antibiotic-free, is used during the phase of non-specific expansion of T cells activated by exposure to peptide-loaded xAPCs.
  • Sodium Chloride Injection A 0.9% sodium chloride solution, USP, available from Baxter Fenwal Laboratories, is used for cell washing procedures during harvesting of T cells. The solution, which is sterile, non-pyrogenic, and free of animal components, is stored at RT prior to use.
  • Dextrose + Sodium Chloride Solution An injectable solution of 5% dextrose and 0.9% sodium chloride, USP (Baxter Fenwal Laboratories), is obtained as a sterile, non-pyrogenic solution free of animal components. The solution, which is used as a storage buffer for activated T cells, is stored at RT prior to use.
  • Lactated Ringer's Solution A 0.9 % Lactated Ringer's solution, USP (Baxter Healthcare Laboratories), which is a sterile, low-endotoxin solution of calcium chloride, potassium chloride, sodium chloride, and sodium lactate in water for injection (free of animal components), is stored at RT prior to use in harvesting and suspending T cells.
  • USP Boxter Healthcare Laboratories
  • Cell culture grade distilled water which is obtained by membrane- filtering and endotoxin-screening (Invitrogen Corporation), is used as a solvent for the preparation of stock solutions of copper sulfate, calcium chloride, and interleukin— 2 (IL-2) and is stored at RT prior to use.
  • IL-2 interleukin— 2
  • Lymphopheresis products are collected from human subjects diagnosed with melanoma and are stored at RT prior to use for the generation of an autologous, patient- specific cell product.
  • Autologous human serum is used as a protein source for culturing of isolated T cells.
  • Autologous human plasma is prepared from lymphopheresis product by adding calcium chloride to achieve fibrin clotting and then collecting the liquid serum phase.
  • the collected liquid serum phase is stored at 4 0 C for short-term storage and at -8O 0 C for long- term storage.
  • a Drosophila xAPC line derived from the xenogenic Drosoph ⁇ la clone B, which is used as a seed stock to create a continuous Drosophila xAPC culture, is obtained as described below. Procedures.
  • CD8 + cells are isolated from leukapheresis samples using an Isolex 30Oi machine (Baxter) by positive selection with an anti-CD8 monoclonal antibody (antibody 1) followed by a DynabeadsTM (Dynal) isolation procedure using sheep anti-mouse IgG (antibody2) coated onto magnetic beads (SAM beads).
  • the anti-human CD8 mouse monoclonal antibody is added to washed cells resuspended in Dulbecco's PBS supplemented with 1% HSA (Baxter-Hyland) and 0.2% sodium citrate.
  • Dynal magnetic beads (DynabeadsTM) are added at a bead to cell ratio of 1:1 to 1 :2 depending on the number of PBMCs.
  • the isolated CD8 + cells are removed by magnetic separation.
  • the remaining non-CD8 fraction is collected and cryopreserved for future use during the restimulation and non-specific expansion steps.
  • Dissociation of the CD8 cell-antibody l-antibody2 -bead complex is achieved by incubation at 37°C for 45 minutes in the presence of CD8 peptides9 -7 o (AAEGLDTQRFSG), the peptide to which antibodyl was generated. Released beads are removed magnetically and the CD8 + cells are counted and analyzed by flow cytometry to evaluate purity.
  • Heat- inactivated serum is prepared by collecting the autologous plasma at the time of the initial cell washing step. Fibrin is clotted using CaCl 2 and the fibrin clot removed. Serum is heat-inactivated, filtered, aliquoted and frozen at -80 0 C. Non-CD8 + cells are retained from the positive selection procedure and purified using a Ficoll gradient. These cells are cryopreserved in DMSO, Pentaspan, and heat-inactivated autologous serum and stored in liquid nitrogen (LN 2 ). These cells are used as the source of adherent cells at the time of the restimulations and are peptide-pulsed prior to use as antigen presenting cells.
  • LN 2 liquid nitrogen
  • Transfected Drosophila S2 cells are incubated in Schneider's medium (10 6 cells/mL) supplemented with 10% fetal calf serum and copper sulfate at 27°C for 24 to 72 hours.
  • S2 cells (clone 1120-3-9 ) are harvested, washed and resuspended in HYQ SFX-Insect medium (Hyclone) containing 0.1 ⁇ g/mL each of human gpl00i54-i62, gpl00209-2i7, gpl0028 ⁇ -288, MART-I27-35, tyrosinase-N369-377 and tyrosinase- D3 69 - 377 peptides and 5 ⁇ g/mL human ⁇ 2 microglobulin recombinant protein purified from E.
  • the S2 cells are mixed with CD8 + cells at a ratio of 1:10 ⁇ Drosophila cells:T cells) in Roswell Park Memorial Institute (RPMI) medium (Gibco) supplemented with 5-10% autologous serum.
  • RPMI Roswell Park Memorial Institute
  • the cell mixture is incubated at 37°C during which time the Drosophila cells die off (by 48 hours).
  • IL-2 (20 U/mL) and IL-7 (30 U/mL) are added to selectively expand the melanoma-specific CTL population.
  • PBMCs obtained at the time of lymphapheresis and frozen for future use, are thawed, washed and resuspended at 10 7 cells/mL in RPMI medium containing 10% autologous serum, 5 ⁇ g/mL recombinant human ⁇ 2 microglobulin and 5 ⁇ g/mL of gpl00i54 -1 62, gpl00209-2i7, gpl0 ⁇ 28o-288, MART-l27-3S, tyrosinase-N369- 377 and tyrosinase-D 36 9. 3 7 7 peptides.
  • the cells are incubated at 37°C for two hours in the flasks used for the restimulation. Nonadherent cells are removed by washing with Dulbecco's PBS.
  • Adherent monocytes are loaded with the peptide epitopes and incubated for 90 minutes in Leibovitz medium containing 5 ⁇ g/mL human ⁇ 2 microglobulin in 1% HSA and 5 ⁇ g/mL of each peptide.
  • the supernatant is removed and the Drosopfa/ ⁇ -activated CD8 + cell suspension (2.5 x 10 6 cells/mL in RPMI medium with 10% autologous serum) is added at a ratio of about 10 CD8 + cells to one adherent monocyte.
  • IL-2 (20 U/mL
  • IL-7 (30 U/mL) are added with a medium change to selectively expand the melanoma-specific CTL population.
  • Non-specific Expansion The CD8 + effector cells that have undergone two rounds of restimulation are expanded in cell culture bags along with feeder cells (irradiated autologous non-CD8 + selected cells) after being stimulated with OKT3 antibody. Frozen non-CD8 + selected cells are thawed, washed and then gamma-irradiated (3,500 rads). A ratio of 4: 1 (feeder: effector) is placed in T-225 flasks that have been coated with OKT3 antibody. OKT3 stimulation is performed in complete RPMI medium containing 10% autologous serum supplemented with 20 U/mL of IL-2. Two days later, the stimulated T cells are diluted with fresh media (X-vivo 10 medium) and transferred to cell culture bags for expansion. Fresh media and IL-2 are supplemented approximately every two to three days to feed the rapidly expanding T cells.
  • CTLs Cell Harvest and Final Product Formulation.
  • Harvest of the final cell product is performed by centrifugation to remove culture media and to concentrate the cells. After centrifugation, cells are washed in saline containing 1% human serum albumin (HSA), filtered through a 70- ⁇ m filter, and then diluted in infusion medium.
  • HSA human serum albumin
  • the cell product for infusion contains autologous CTLs in 30OmL Lactated Ringer's Injection, USP (76% v/v), 5% Dextrose in 0.9% Sodium chloride (4% v/v) and 25% HSA (20% v/v).
  • Final product cells are packaged in a 1000 mL transfer bag in a chilled insulated shipping container with a temperature data logger.
  • Phenotype A portion of the cell harvest is used to test the phenotype of the CD8 product prior to shipment.
  • the final product specification is CD3, CD8 and TCR expression. Additional phenotypic evaluation for markers consistent with memory, activation, etc. are measured and recorded.
  • the HLA-A2, HLA-C and HLA-DR status of the sample is determined by analysis of a DNA preparation isolated from a PBMC sample prepared at the time of receipt of the initial leukapheresis and from the CD8+ T cells collected at the end of the culture, taken from the harvested cell product. PCR analysis of DNA samples is performed with HLA-specific primer oligos provided by Genovision. The final product specification is identity with day 0 sample.
  • the desired cell dose for infusion is 10 10 CD8+ T cells.
  • the final product infusion limit is 10 10 cells.
  • the total number of cells generated is patient-dependent.
  • Cell doses averaged 9 X 10 9 in a previous study where the cells were generated under the same ex vivo protocol.
  • the final cell count is made by determining the number of viable cells.
  • Viability The viability of the final cell dose is >70%. The number of viable cells is established by microscopically counting cells that exclude trypan blue dye.
  • Mycoplasma Testing Testing of the cells for mycoplasma is performed before the cells are administered to the patient to confirm negative mycoplasma detection.
  • the Roche PCR kit is used which also includes an ELISA assay.
  • samples of the T cell cultures are collected at the first cytokine feeding (day 6 ) and sent to BioReliance to fulfill the 28-day culture procedure. The results of the culture are received post T cell infusion.
  • Endotoxin testing The endotoxin test on the final cellular product is performed using a Limulus Amebocyte Lysate assay (BioWhittaker, Walkersville, MD). The endotoxin level limit is less than 5 EU/kg infused into the patients. The final cell product specification is ⁇ lEU/mL.
  • Process Product Sterility Tests In process testing for sterility is performed using the BacT/ Alert system. Samples are taken during times of media changes. A sample from the final dose is also tested by the BacT/Alert system, and the final product specification is absence of growth.
  • Drosophila DNA and Insect Viral RNA Detection DNA is isolated from the final CD8+ T cell product and used as a template for a Drosophila DNA PCR assay, which uses primers specific for the insect vector used to prepare the recombinant Drosophila APCs. This sample is compared to the na ⁇ ve CD8+ sample that acts as the negative control and Drosophila cell DNA is used as the positive control.
  • Total RNA is also isolated from the same CD8+ T cell samples and a quantitative real time RT-PCR, which can detect 20 copies/ ⁇ g of cDNA of three different, known insect viral RNA viruses, is performed.
  • the product specification for both assays is negative detection of insect genomic and viral nucleic acid.
  • Lytic activity is evaluated for the final CD8+ T cell product by a chromium-release assay in both peptide-loaded T2 cell targets, to measure anti- peptide activity, and established or autologous melanoma cell HLA- A2 targets and donor-matched tumor and non-tumor lines (Malme 3 and Malme 3M) to asses melanoma-specific killing.
  • the product specification is specific killing of target cells.
  • Tetrameric Staining CD8+ T cells for the na ⁇ ve leukapheresis-derived sample is evaluated for the presence of antigen-specific T cells with tetrameric molecules which have been designed to enumerate T cells with specificity for the peptide epitopes used to generate the CTLs ex vivo. This allows the monitoring of antigen-specific T cells after treatment.
  • Sensitive, quantitative real time PCR assays allow for the monitoring of circulating melanoma cells in patient blood samples, before, during and after drug treatment. While not all stage III/IV melanoma patients demonstrate circulating tumor cells, the detection of these cells can be used as a surrogate marker for response to therapy.
  • CTL products prepared as described above may be used in the combination therapy treatment regimens of the invention as illustrated by the preferred embodiments described in the following examples.
  • CTLs autologous cytotoxic T lymphocytes
  • a lymphodepletion agent selected from cladribine and DAB IL2 as well as cytokines IL-2 and IFN- ⁇ — 2b, as well as a control treatment.
  • the cell therapy agent which comprises autologous CTLs that have been activated such that they specifically target melanoma cells expressing MART-I, gplOO, and tyrosinase by contacting Drosophila xAPCs loaded with MART-I-, gplOO-, and tyrosinase-derived antigenic peptides, is designed to enhance and maintain a patient's immune targeting of melanoma cells expressing MART-I, gpl 00, and tyrosinase.
  • the addition of immunomodulators IFN- ⁇ -2b and IL-2 is included to augment the response of the CTLs.
  • a non-myeloablative, but lymphocyte-depleting, preparative regimen (cladribine) or a specific T cell subset depletion denileukin diflitox (DAB IL-2) is administered prior to the CTL infusion to enhance the engraftment of the CTLs.
  • the treatment is designed to cause tumor regression and clinical benefit through persistence of melanoma-specific CTLs.
  • a non-myeloablative, but lymphocyte-depleting, preparative regimen consisting of either cladribine (0.12 mg/kg/day x 5 days) or DAB IL-2 (a single injection of 18 ⁇ g/kg).
  • a concomitant regimen of interferon- ⁇ -2b IFN- ⁇ -2b; 10MIU/m 2 ) is administered daily, for 5 consecutive days, by subcutaneous injection prior to CTL infusion.
  • the patients receive a single dose of ex vivo- generated, Drosophila xAPC-stimulated, autologous CD8 + T cells, which display the CTL phenotype.
  • the cell infusion is immediately followed by subcutaneous daily administration of low-dose (e.g., 3MIU) IL-2, for twenty-eight (28) days, followed by tumor assessment.
  • subcutaneous, daily administration of low-dose (e.g., 3MIU) IL-2 is continued in patients without evidence of disease progression.
  • the CTLs are generated by a method in which purified na ⁇ ve CD8 + T cells are stimulated with Drosophila xAPCs presenting six different melanoma-associated T cell peptide epitopes in the context of human MHC class I restriction, resulting in CTLs that display multiple specificities for MART-I, gplOO, and tyrosinase, after approximately 34 days in culture.
  • Blood is collected at screening to qualify patients for the study and the preparative leukapheresis procedure takes place on day 0 or day 1.
  • Patients undergo a standard 2.0- 3.0X the blood volume leukapheresis to obtain enough white blood cells required to prepare the cell therapy agent.
  • additional leukapheresis procedures ( ⁇ of third the amount used to obtain the initial CD8 sample) is be performed to obtain peripheral blood cells for analysis. These samples are used to monitor the presences of antigen-specific T cells and possible circulating melanoma tumor cells. Plasma and blood cells are also collected to meet the xenotransplantation requirement for archiving pre- and post-treatment samples.
  • Leukapheresis products are transported to a facility where CD8 + T cells are isolated, stimulated and cultured for approximately 34 days. An evaluation of measurable lesions is conducted at both 4 and 8 weeks following the CTL infusion. An objective of the study is to determine whether administration of a full non-myeloablative, lymphocyte-depleting (cladribine) or a selective T cell subset depletion (DAB IL-2), prior to the T-cell infusion and in conjunction with an established immune-enhancing cytokine regimen, will result in antigen-specific T cell persistence with accompanying objective tumor regression.
  • cladribine lymphocyte-depleting
  • DAB IL-2 selective T cell subset depletion
  • CDS + cells isolated from leuJcapheresis samples by positive selection with an anti- CD8 antibody are stimulated with human melanoma-associated antigenic peptides
  • a non-specific expansion step with anti-CD3 mAb is also included to increase the total number of cells: generally 25- fold greater than that obtained at the end of the second restimulation step.
  • Cytolytic T cell activity is measured against peptide-loaded T2 cells and a panel of A2 + melanoma cells, while the purity of the in vz7ro-stimulated CD8 + T cells is assessed by flow cytometry. Additionally, interferon-gamma production, in response to antigen-specific stimulation and peptide-specif ⁇ c tetrameric analysis, confirms the effector function and specificity of the generated CTLs, respectively.
  • TILs tumor-infiltrating lymphocytes
  • CTLs generated ex vivo with Drosophila-xAVCs are potent, high- avidity, antigen-specific T cells capable of peptide-specific lysis (see, e.g., Figure 1) and strong melanoma tumor cell killing.
  • the reproducible generation of the CTLs, with strong cytolytic activity for tumor cells, is believed to be attributable to several contributing factors.
  • CD8 + purification with a peptide-specific, anti-CD8 monoclonal antibody selected for cells with a high density of CDS while the antibody release in the presence of specific peptide, avoided non-specific stimulation of the T cells.
  • the highly- purified CD8 + T cells (Table I) are stimulated with xAPCs presenting specific T-cell epitopes in the context of MHC molecules, where a high density of antigen presentation results in a potent primary T-cell immune response.
  • the inclusion of multiple costimulation molecules on the APCs optimizes the T cell stimulation.
  • the restimulation steps incorporate autologous, mononuclear cells that have been loaded with the same melanoma- associated T cell epitopes used in the primary stimulation and provide a boost to the proliferating T cells.
  • a single non-specific expansion step with OKT3, autologous feeder cells and low-dose IL-2 maintains the same percentage of antigen-specific T cells recorded at the end of the second restimulation step, while increasing the total number of T cells ⁇ 25-fold.
  • CD3 + T cells 45 (29-64) 83 (59-98)
  • CD8 + T cells 17 ( 8-31) 84 (60-96)
  • CD15 + /CD16 + neutrophils 2 0.5-9) 0.5 (0.5-1)
  • the clinical endpoints were: 1) safety and tolerability of reinfused autologous CTLs after in vitro immunization; 2) determination of the kinetics of the infused CTLs in the systemic circulation by limiting dilution analysis; 3) whole body disposition of ' ' ' indium-labelled CTLs by radioscintigraphy; 4) cell composition of biopsied nodules by immunohistochemical analysis (CTLs, TH, NK, B cells) and 5) regression of measurable lesions and duration of response over two months.
  • CTLs immunohistochemical analysis
  • TTP time to progression
  • Interferon-alpha has a broad spectrum of immunomodulatory and antiproliferative effects in a variety of malignancies. It is believed that one mechanism of action of IFN- ⁇ is the upregulation of tumor antigen expression on melanoma cells. It has the ability to enhance the expression of immunologically important molecules on the surface of the tumor. These include the MHC antigens, accessory molecules, as well as tumor-associated antigens. 25'27 These immunomodulatory effects may improve the activity of the immune system, including both antibodies and lymphocytes, to recognize and attack tumor cells in vivo. Active, specific immunotherapy has demonstrated significant clinical responses in the treatment of disseminated melanoma.
  • IL-2 Human recombinant interIeukin-2
  • IL-2 is a lymphokine produced by recombinant DNA technology that has been shown to exhibit a variety of biological activities. IL-2 stimulates the immune system and exerts its biological effects following binding to specific receptors on the surface of target cells. In vitro, it has been shown to enhance T cell proliferation and lymphocyte cytotoxicity, induce the killer activity of both lymphokine- activated and natural killer cells, and induce interferon-gamma production.
  • Administration of high-dose IL-2 to 283 patients produced a remission rate of 7% with 9 complete responses, and the patients remained disease free from 9 to greater than 91 months.
  • a regimen of cyclophosphamide and fludarabine originally used in renal cell cancer patients receiving HLA-matched allotransplants 32 has recently been used in metastatic melanoma patients 16 ' 31 .
  • the rationale for the incorporation of a lymphodepleting regimen prior to adoptive transfer of T cells was to possibly destroy regulatory cells, disrupt homeostatic T cell regulation ("making space"), or abrogate other normal tolerogenic mechanisms which potentially could enhance the engraftment of the transferred T cells. No treatment-related mortality was observed, and therefore non-myeloablative chemotherapy in combination with antitumor lymphocytes plus high-dose IL-2 should be safe.
  • the non-myeloablation was induced with fludarabine (25 mg/m 2 ) and cyclophosphamide (60 mg/kg) and was followed by a lymphocyte infusion and high dose IL-2 (720,000 IU/kg).
  • chemotherapeutic agents cyclophosphamide and fludarabine while not completely myeloablative, has been demonstrated to be myelosuppressive, affecting neutrophils, lymphocytes, platelets, and red blood cells.
  • Neutrophil levels which fell to a nadir on day 10 following the initiation of chemotherapy, were recorded at 6/mm 3 and recovered to above 500/mm 3 on day 14 with support of filgrastim (G-CSF). Lymphocyte levels had a nadir of 6/mm 3 and recovered to above 200/mm 3 during the same time period. Patients usually displayed neutrophil counts above 500/mm 3 and platelet counts above 20,000/mm 3 , 2-3 weeks after the initiation of the chemotherapy.
  • T reg T regulatory cells
  • DAB 389 IL-2 denileukin diftitix
  • RNA-DC vaccine was administered and a 10-fold enhancement in the immune response to the vaccine antigens was noted in the patients receiving both DAB IL-2 and vaccine versus vaccine alone.
  • the same dose (18 ⁇ g/kg) six days prior to an adoptive transfer of the antigen-specific CTLs is a preferred dose for use in regimens in accordance with the invention.
  • Subjects for the study are human patients diagnosed with metastatic melanoma who are HLA-A2 positive.
  • a monoclonal antibody (BB7.2) is used to analyze PBMC samples by FACS analysis and further analysis is performed using the Olerup SSPTM PCR test (Geno Vision) to determine the HLA-A*0201 subtype.
  • Figure 2 depicts the treatment regimens described in more detail below.
  • the patients are divided into three cohorts of subjects, with 10 in each cohort.
  • CTLs plus cytokines are administered.
  • CTLs plus cytokines are administered in the Cohort A regimen.
  • CTLs plus cytokines are administered in the Cohort B regimen.
  • patients receive cladribine (days 0-4) plus CTLs (day 36) plus cytokines.
  • patients receive DAB IL-2 (day 30) plus CTLs (day 36) plus cytokines.
  • the experimental regimens are described in more detail below.
  • Cytotoxic T lymphocyte therapy for malignant melanoma utilizes autologous in vitro-ac ⁇ vs ⁇ Q ⁇ CD8 + T cells to destroy melanoma cells bearing melanoma-associated antigenic epitopes.
  • CTLs are prepared from leukapheresis samples generated at the clinical site and transferred to a facility where CTLs are generated under GMP guidelines and returned for infusion.
  • leukapheresis On day 0, patients with metastatic melanoma undergo leukapheresis (approximately 10-15 liter run, depending on the patient's weight and condition). The obtained cells are shipped overnight at room temperature for processing at a GMP manufacturing facility. At appropriate times, leukapheresis is performed to obtain peripheral blood lymphocytes for analysis. These samples are used to monitor the presence of antigen-specific T cells and the presence of circulating melanoma tumor cells. The monitoring leukapheresis procedure processes approximately 5 liters of peripheral blood (or one-third the volume collected at the initial leukapheresis to obtain CD8 cells for subsequent infusion) at the clinical site and is shipped to the GMP manufacturing facility.
  • a PCR assay is used to determine a decrease in the level of circulating melanoma cells following therapy.
  • the detection of circulating melanoma cells can be measured by a sensitive, quantitative real time PCR assay 39"41 .
  • the presence of antigen-specific T cells is evaluated with peptide-specific/HLA-A2 tetramers.
  • the final CTL preparation for infusion contains 1 x 10 10 autologous CTLs in 30OmL, contained in 76% (v/v) Lactated Ringer's Solution, 4% (v/v) of D5NS and 20% (v/v) of 25% human serum albumin (HSA).
  • the CTL product is prepared in a sterile bag, suitable for infusion, held on ice and infused over 30 minutes, within forty-two (42) hours of preparation.
  • the CTLs Prior to infusion, the CTLs are monitored for lytic activity (both peptide-loaded target cells and melanoma tumor cell lysis), viability, and purity by FACS analysis. The absence of both Drosophila cell DNA and insect viral RNA is checked with sensitive quantitative, real-time PCR assays. Sterility is determined by BacT/Alert® analysis, mycoplasma testing and Gram stain analysis.
  • Patients receive a CTL infusion of approximately 10 10 autologous T cells, which have been stimulated and expanded ex vivo to become cytotoxic T lymphocytes capable of targeting to cells in vivo expressing the antigens to which the CTLs are directed.
  • the cells are infused after the end of the IFN-alpha treatment and immediately precede the start of the IL-2 regimen.
  • Interleukin-2 is obtained from Chiron Corporation (Emeryville, CA). It is supplied as single-use vials containing 22 million IU ( ⁇ 1.3 mg) of IL-2 as a sterile, white to off-white lyophilized cake plus 50 mg mannitol and 0.18 mg sodium dodecyl sulfate, buffered with approximately 0.17 mg monobasic and 0.89 mg dibasic sodium phosphate to a pH of 7.5 (range 7.2 to 7.8). The powder is reconstituted with 1.2 mL of Sterile Water for Injection, USP, and the resultant concentration is 18 million IU/ml or 1.1 mg/mL.
  • Intact vials are stored in the refrigerator (2° - 8°C) and protected from light. Reconstituted IL-2 is further diluted with 2.4 mL of D5% water. The final concentration (6MU/mL), once drawn into plastic syringes, is good for 14 days after reconstitution, if kept refrigerated at 2-8°C. The final dilution of IL-2 is self-administered on an outpatient basis subcutaneously at a dose of 3 million units (3 MIU), initiating immediately following the CTL infusion and daily until disease progression. INTRON- A ® is obtained from Schering-Plough Corporation (Kenilworth, NJ).
  • This IFN-alpha-2b product is supplied either as a lyophilized powder or a solution for injection in sterile 5ml vials.
  • a vial contains approximately 3 x 10 U or l ⁇ x lO U IFN.
  • the lyophilized powder is reconstituted preferably just prior to use, as there is no bacteriostatic agent included; unused, lyophilized powder is stored in a refrigerator or freezer (-4 0 C to -20 0 C). When the agent is supplied as an injectable solution, it is stored in the refrigerator (2°-8°C or 36°-46°F).
  • the final dilution of IFN- O is self-administered on an outpatient basis at 10MU/m 2 on days 31-35 as a subcutaneous injection.
  • DAB38 9 ⁇ L-2 (denileukin diftitox, ONTAK ® ) is a recombinant DNA-derived cytotoxic protein composed of the amino acid sequences for diphtheria toxin fragments A and B followed by the sequences for interleukin 2 that is expressed in E coli. It is a targeted drug, which binds to cells expressing CD25 (IL-2R) on their surface. It interacts with the high-affinity IL-2 receptor on the surface of malignant or normal T regulatory (T reg ) cells to inhibit intracellular protein synthesis, rapidly leading to cell death.
  • DAB389IL-2 is supplied in single use vials as a sterile, frozen solution intended for intravenous (IV) administration.
  • Each 2 mL vial of ONTAK contains 300 meg of recombinant DAB-IL2 in a sterile solution of citric acid (2OnM), EDTA (0.05mM) and polysorbate 20 ( ⁇ 1%) in water for injection, USP.
  • the solution has a pH of 6.9-7.2.
  • Intact vials are stored frozen or -10 0 C but not refrozen.
  • the material is brought to room temperature (25°C or 77°F) before preparing the dose by thawing in a refrigerator for not more than 24 hours or at room temperature for 1 -2 hours, and diluted with NS to a concentration of >15 mcg/mL.
  • DAB 3 89IL-2 is administered by intravenous injection, preferably by infusion over at least 15 minutes.
  • Patients assigned to Cohort C receive a single subcutaneous injection of DAB IL-2 on day 30, which is one day before the initiation of IFN-Dand six days prior to the injection of the CTLs.
  • the dose is 18 ⁇ g/kg.
  • Leustatin® (2-chloro-2'deoxy-DD-adenosine) is a synthetic antineoplastic agent. It is a purine nucleoside analog resistant to the action of adenosine deaminase, which results in preferential lymphocytoxicity.
  • adenosine deaminase e.g., lymphocytes and monocytes
  • cladribine is phosphorylated into the active triphosphate deoxynucleotide, 2-CdATP, which accumulates, causing a disruption of cellular metabolism, DNA damage, and subsequent cell death.
  • the drug is a clear, colorless, sterile, preservative-free, isotonic solution supplied in single-use vials containing lOmg (lmg/mL) of cladribine.
  • lOmg lmg/mL
  • cladribine contains lmg of the active ingredient and 9 mg (0.15mEq) of sodium chloride as an inactive ingredient.
  • the solution has a pH range of 5.5 to 8.0.
  • Phosphoric acid and/or dibasic sodium phosphate may be added to adjust the pH to 6.3 ⁇ 0.3.
  • Intact vials are stored refrigerated (2-8°C).
  • the dose of cladribine for Cohort B is 0.12 mg/kg/day for a total of 5 days, fractionated into 3-4 sites where a maximum of 3.0 cc is administered at a single site.
  • the total cladribine dose is 0.6 mg/kg.
  • Leustatin has been found to be 12% higher than reported in previous clinical studies compared with a cladribine stock synthesized in a laboratory. The dose that was reported to be 0. lmg/kg is now estimated to actually have been only 0.087 mg/kg 33 .
  • a study in relapsing-remitting multiple sclerosis patients used 0.07 mg/kg/day X 5 days, monthly for 6 months for a total cumulative dose of 2.1 mg/kg 3 . A total of 27 patients randomized to the treatment drug arm.
  • Infections were limited to an episode of mild segmental herpes zoster that occurred in two cladribine- treated patients and in one patient receiving placebo. There were no individual cases of significant thrombocytopenia, anemia granulocytopenia or generalized marrow suppression.)
  • Patients may receive any of the following to abrogate IFN-alpha and IL-2 toxicity during treatment and thereafter: acetaminophen (650 mg PO q4h prn), diphenhydramine (50 mg IM or PO q4h prn), indomethacin (25 mg PO TID or 75 mg SR qd), prochlorperazine (10 mg IV or PO q4h prn nausea), Zantac ® (150 mg BID gastritis).
  • Other anti-inflammatory and antiemetic agents not specifically prescribed may be substituted for any of the above supportive drugs.
  • Van der Bruggen P Traversari C, Chomez P 5 et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 254: 1643- 1647, 1991. 7. Gaugler B, Van der Eynde B, Van der Bruggen P, et al. Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J Exp Med 179:921-930, 1994.

Abstract

In a cancer treatment combining cell therapy with chemotherapy, autologous CD8+ T cells are obtained from a patient, activated ex vivo by contacting them with xenogenic antigen presenting cells loaded with selected peptide antigen, thereby generating antigen- specific activated cytotoxic T lymphocytes. Such activated CTLs are administered to the patient in conjunction with a lymphodepletion and CTL maintenance regimen comprising a non-myeloblative but lymphdepleting agent, such as cladribine or denileukin diftitox, and interleukin-2 and interferon-α-2b stimulatory cytokines.

Description

CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES
CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority to United States Provisional Application No. 60/778,516, filed March 1, 2006.
FIELD OF THE INVENTION
The invention relates to methods for treating cancer in patients involving the administration of activated cytotoxic T lymphocytes, cytokines such as IL-2 and IFN- alpha-2b, and cladribine or denileukin diftitox as a lymphodepleting agent.
BACKGROUND OF THE INVENTION
To facilitate an appreciation of the invention, this section may discuss the historical and technical background leading to the development of the invention, including observations, conclusions, and viewpoints that may be unique to an inventor. Accordingly, the background statements herein should not be construed as an admission regarding the content of the prior art.
A number of therapies have been developed to treat a variety of cancers. Many of these efforts have centered around chemotherapeutic regimens. In one particular combination chemotherapy regimen designed as a treatment for metastatic melanoma, response rates of 35-50% were reported with the "Dartmouth regimen" (a of combination DTIC, cisplatin, BCNU and tamoxifen), but the duration of survival has remained at 6 to 10 months. High rates of remission also have been reported for aggressive high-dose intensity chemotherapy1 and repletion of hematopoeisis with autologous bone marrow transplants. Nevertheless, the median duration of survival was short, approximately four months2.
Significant improvements in survival on the order of several years have been noted in a small percentage of melanoma patients undergoing certain immunotherapies. This includes active specific immunotherapy with cancer vaccines, as well as the use of nonspecific boosters of the immune system, such as interleukin-2 (IL-2) and interferon- alpha (IFN-α)Λ5
The identification of T-cell defined tumor antigens in melanoma has led to clinical trials that target cancer cells by attempting to augment the antigen-specific cellular immune response. This approach has been pursued in numerous vaccination strategies in which the antigens are delivered in an immunogenic context in an attempt to induce potent T cell responses in vivo. Although some clinical responses have been observed in the vaccine trials, the magnitude of the induced T-cell response has generally been low, or undetectable and correlated poorly with clinical responses. Immunization of melanoma patients with cancer antigens may increase the number of circulating CTL precursors; however it has not correlated with clinical tumor regression, suggesting a defect in function or activation in vivo.
Studies in mouse tumor models have demonstrated that adoptive immunotherapy, which involves in vitro immunization of T cells specific for one or more tumor antigens, may be efficacious with minimal toxicity. An obstacle in applying this strategy to the treatment of human tumors has been the identification of immunogenic antigens that render the tumor cells susceptible to CTL-mediated destruction. The isolation of tumor-reactive T cells from melanoma patients has led to the identification of some of the tumor antigens (epitopes) to which CTLs are directed. These include tyrosinase, MART-1/Melan A, gplOO, and MAGE. Of these, tyrosinase and MART-I are nearly universally expressed on melanoma and therefore represent a desired target choice for adoptive immunotherapy.6"13
Adoptive T cell therapy involves the removal of T cells from the host environment where tolerogenic mechanisms are active in vivo in cancer patients and contributes to the ineffective responses demonstrated in this patient population. CD8+ T cells may be stimulated ex vivo to generate antigen-specific CTLs (see, e.g., U.S. Patent No. 6,225,042). Early adoptive immunotherapy approaches used activated lymphocytes as a treatment for various cancers.14 Initially, lymphokine-activated killer cells (LAK), and later tumor- infiltrating lymphocytes (TIL), activated ex vivo with EL-2, were used, but the demonstration of efficacy was equivocal. These early, controlled clinical trials failed to show an advantage to the use of the ex vzvo-activated cells over the direct administration of IL-2 to melanoma patients. More recent studies by Yee et al. (Fred Hutchinson Cancer Research Center)15 and Dudley et al. (NCI)15 have demonstrated the potential for certain adoptive T-cell therapeutic approaches. These studies involved use of either T-cell clones specific for MART-I or gplOO and low-dose IL-2, or TILs expanded ex vivo with allogeneic feeder cells and high-dose IL-2. These studies confirmed that adoptive immunotherapy holds promise as a treatment of cancer, although its full development has been impeded by the lack of reproducible methods for ex vivo generation of therapeutic numbers of antigen-specific CD8+ CTLs.17
Cytotolytic CD8+T cells are a major line of defense against viral infections. CD8+ lymphocytes specifically recognize and lyse host cells that are infected with a virus. Although it would be desirable to harness the cytotoxic activity of CTLs, few in vitro/ex vivo procedures have been available to specifically activate CTLs. The identification of key melanoma-associated antigens and a method for specific in vitro activation of CTLs, allows for an efficient evaluation of adoptive immunotherapy for metastatic melanoma.
While it is possible to use naturally occurring antigen presenting cells (APCs) for CD8 activation in vitro (e.g., dendritic cells, macrophages, autologous tumor cells), the efficiency of activation is low since the MHC Class I molecules of native APCs contain many other peptide epitopes, thus allowing minimal presentation of tumor-associated peptide epitopes. Most of these presented peptides represent normal, innocuous endogenous proteins. A more direct approach to this problem would be to activate CD8+ T cells specifically to those epitopes relevant to combating the disease, in this particular case, melanoma-associated antigens.
Recently, an artificial APC has been developed utilizing a Drosophila melanogaster (fruit fly) embryonic cell line, which expresses the major histocompatibility complex (MHC) Class I molecules.18'19 See also U.S. Patent Nos. 6,225,042 and 6,355,479. Since the insect Drosophila lacks an advanced immune system, the TAP- 1,2 peptide transporters, which are involved in the loading of peptide epitopes into the human Class I molecules, are absent. As a result, the transfected Class I molecules appear on the Drosophila cell surface as empty vessels. By incubating these transfected Drosophila cells with exogenous synthetic peptides that bind to the specific Class I molecules (i.e., tumor antigen T-cell peptide epitopes), all of the available Class I molecules maybe occupied with MHC- restricted, specific peptide epitope(s). The high density expression of the Class I molecules presenting single or multiple epitopes, and the addition of key co-stiniulatory molecules B7-1 (CD80), CD70 , LFA-3 (CD58), and ICAM-I (CD54) on these Drosophila APCs may permits the in vitro generation of potent, autologous cytotoxic CD8 T cells, which are specific for the antigenic peptides.
SUMMARY OF THE INVENTION
Various general aspects and preferred embodiments of the invention are reflected in the claims appended to this specification, which are incorporated by reference herein. Other preferred embodiments, features, and advantages of the various aspects of the invention will become apparent from the detailed description below taken in conjunction with the drawing figures.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 illustrates cytolytic activity of CTLs generated ex vivo directed against peptide-loaded target cells. CDS+ T cells isolated from a melanoma patient were immunized in vitro with Drosophila APCs loaded with five different melanoma-associated peptide epitopes. The activated CD8+ T cells were cultured in vitro using IL-2 and IL-7 to selectively expand the melanoma-specific CTLs. Activity was assessed as specific lysis of 5!Cr-labeled T2 target cells individually loaded with each peptide (Tyr-l689, Tyr-2792, gpl00-l8i7, gplOO-2853(or MART- l81g versus T2 cells loaded with an HLA-A2 control peptide).
Figure 2 provides a schematic illustration of two preferred embodiments of treatment regimens according to the invention along with a control regimen. DETAILED DESCRIPTION OF THE INVENTION AND ITS PREFERRED EMBODIMENTS
The various aspects of the invention are illustrated below through detailed description of specific and preferred embodiments. For the sake of brevity, the disclosures of all patents and other publications cited herein are incorporated by reference. Unless defined otherwise herein or apparent from the context, all technical and scientific terms used herein have the same meaning as commonly used in the art.
The terms "including", "comprising", and "containing" are used herein in their open, non-limiting sense. The therapeutic regimens of the invention, which comprise administering CTLs that have been activated by contacting xAPCs loaded with selected peptide in conjunction with cytokines and at least one lymphodepleting agent selected from cladribine and DAB-IL2 may be employed to treat cancer in a subject in need of such treatment. Preferably, the cancer is selected from malignant melanoma, multiple myeloma, prostate cancer, lymphoma, non-Hodgkin's lymphoma, leukemia, acute lymphoblastic leukemia, acute myeloid leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, Burkitt's lymphoma, thyroid cancer, uterine cancer, kidney cancer, ovarian cancer, lung cancer, breast cancer, liver cancer, pancreatic cancer, prostate cancer, colon cancer, skin cancer, stomach cancer, and cervical cancer.
Thus, in one preferred embodiment the invention provides an adoptive CTL therapy regimen for treating or ameliorating a metastatic melanoma comprising: obtaining naϊve CD8+ T cells from a subject; contacting the naϊve CD8+ T cells with xenogenic antigen presenting cells (xAPCs) that have been loaded with selected peptide antigen, thereby generating activated CTLs that target cells expressing the selected peptide antigen; administering the activated CTLs back to the subject; administering at least one lymphodepleting agent selected from cladribine and DAB-IL2; and administering at least two cytokines that effect CTL persistence. The adoptive CTL therapy regimen preferably employs autologous CD8+ T cells that are activated ex vivo, which when activated and administered to a subject in accordance with the invention are capable of in vivo destruction of tumor cells bearing cancer-associated antigenic epitopes,
The term "subject" in this context refers to a mammalian patient in need of treatment for a cancer. For example, a subject may be a human diagnosed with a melanoma, such as an advanced malignant metastatic melanoma, e.g., a patient who has been diagnosed to be HLA- A2 positive and have Stage III/IV unresectable disease.
The CTL agent is prepared from xAPCs. Exemplary xenogenic antigen presenting cells (xAPCs) that are suitable for use may include the following components: an exogenous MHC I molecule; one or more exogenous assisting molecules to assist in the activation of naϊve T cells; and host cells capable of expressing the exogenous molecules their surface. Preferably, the exogenous molecules are encoded by xenogenic nucleic acid that has been introduced into host cells. The xAPCs preferably also express exogenous co- stimulator and adhesion molecules, which potentiate T cell activation capability of the xAPCs. Preferably, the host cells are insect cells, more preferably Drosophila cells, such as Schneider 2 (S2) cells. Exemplary xAPCs and methods for their manufacture are described in, for example, U.S. Patent Nos. 6,225,042 and 6,355,479.
The xAPCs may be loaded with peptide antigen by a variety of methods known or available in the art. Peptides are selected that are capable of binding to the empty MHC Class I molecules. Selected peptides preferably correspond to epitopes comprising antigenic or immunogenic amino acid sequences derived from protein expressed on the surface of cells, which will serve as targets for the T cells employed in the adoptive CTL therapy. In order to load empty MHC Class I molecules with selected peptide, one or more antigenic or immunogenic peptide species that binds to such empty MHC Class I molecules may be contacted with the xAPCs under suitable conditions for the binding to occur.
One or more antigenic or immunogenic peptide species may be selected. If more than one species is selected, they may be contacted with the xAPCs simultaneously or at distinct instances, resulting in multi-antigenic or multi-immunogenic MHC-peptide complexes produced on the xAPCs.
Loading of the selected peptide onto empty MHC molecules preferably occurs under conditions that approximate biological binding conditions, which may be approximated in vitro, ex vivo, or in vivo. In selecting peptides, the artisan may consider one or more factors such as thermodynamic, electrostatic, energetic, and entropic considerations, as well as specific amino acids within selected peptides that are required for efficacious binding to MHC molecules.
Preferred peptides include, for example, the peptides corresponding to amino acid sequences selected from a tyrosinase protein, a gplOO protein, and a MART-I protein. Other preferred peptides include YMNGTMSQV (SEQ ID NO:1), YMDGTMSQV (SEQ ID NO:2), AAGIGILTV (SEQ ID NO:3), ITDQVPFSV (SEQ ID NO:4), YLEPGPVTA (SEQ ID NO:5), and KTWGQYWQV (SEQ ID NO:6). Additional exemplary peptides that maybe selected include, for example, the following amino acid sequences, where the protein from which each peptide is derived is noted parenthetically: SILSLKEAST (C- Lectin; SEQ ID NO:1), KMASRSMRL (C-Lectin; SEQ ID NO:2 ), ALALAALLW (Pec 60; SEQ ID NO: 3), ALLWDREV (Pec60; SEQ ID NO: 4), YMNGTMSQV (Tyrosinase; SEQ ID NO:5), YMDGTMSQV (Tyrosinase; SEQ ID NO:6), ITDQVPFSV (gplOO; SEQ ID NO:7), YLEPGPVTA (gplOO; SEQ ID NO:8), AAGIGILTV (MART-I; SEQ ID NO:9), ELAGIGILTV (MART-I; SEQ ID NO: 10), CLTSTVQLV (Her-2/neu; SEQ ID NO:11), HLYQGCQW (Her-2/neu; SEQ ID NO: 12), KIFGSLAFL (Her-2/neu; SEQ ID NO: 13), IIS A VVGIL (Her-2/neu; SEQ ID NO: 14), PLTSIISAV (Her-2/neu; SEQ ID NO: 15), VMAGVGSPYV (Her-2/neu; SEQ ID NO: 16), VLVKSPNHV (Her-2/neu; SEQ ID NO: 17), ELVSEFSRM (Her-2/neu; SEQ ID NO: 18), YLSGANLNL (CEA; SEQ ID NO: 19), GPLTPLPV (AES; SEQ ID NO:20), SLLMWITQC (NY-ESO-I; SEQ ID NO:21), KALFAGPPV (CA- 125; SEQ ID NO:22), YLETFREQV (CA-125; SEQ ID NO:23), GLQSPKSPL (CA-125; SEQ ID NO:24), VLLKLRRPV (CA-125; SEQ ID NO:25), ELYIPSVDL (CA-125; SEQ ID NO:26), SLLMWITQV (NY-ESO-I; SEQ ID NO:27), ILAKFLHWL (Telomerase; SEQ ID NO:28), STAPPVHNV (MUC-I; SEQ ID NO:29), FLWGPRALV (MAGE-3; SEQ ID NO:30), FMWGNLTLA (CA-125; SEQ ID NO:31), RLVDDFLLV (Telomerase; SEQ ID NO:32), HLSTAFARV (G250; SEQ ID NO:33), QLSLLMWIT (NY-ESO-I; SEQ ID NO:34), ELWTHSYKV (FBP; SEQ ID NO:35), KVAELVHFL (MAGE-3; SEQ ID NO:36), YIFATCLGL (MAGE-3; SEQ ID NO:37), HLYIFATCL (MAGE-3; SEQ ID NO:38), MLMAQEALAFL (CAMEL; SEQ ID NO:39), STLEKINKT (SSX-4; SEQ ID NO:40), KASEKIFYV (SSX-2; SEQ ID NO:41), SLLMWITQCFL (NY-ESO-I; SEQ ID NO:42), ELTLGEFLKL (Survivin; SEQ ID NO:43), LTLGEFLKL (Survivin; SEQ ID NO:44), SLLEKREKT (SP 17; SEQ ED NO:45), TLGEDDPWL (SART-I; SEQ ID NO:46), KLGLKPLEV (SART-I; SEQ ID NO:47), YLWTSAKNT (SCP-I ; SEQ ID NO:48), STAPPAHGV (MUC-I; SEQ ID NO:49), GMGSEELRL (LIVIN; SEQ ID NO:50), SLGSPVLGL (LIVIN; SEQ ID NO:51), YLFFYRKSV (hTRT; SEQ ID NO:52), CQQEETFLL (CA-125; SEQ ID NO:53), TLAKFSPYL (PRAME; SEQ ID NO:54), NLTHVLYPV (PRAME; SEQ ID NO:55), STFKNWPFL (Survivin; SEQ ID NO:56), SLLQHLIGL (PRAME; SEQ ID NO:57), FLDQRVFFV (gplOO; SEQ IDNO:58), FLDQRVFW (gplOO; SEQ ID NO:59), FLDQVAFVV (gplOO; SEQ ID NO:60), GLDREQLYL (MUC-16; SEQ ID NO:61), VMQHLLSPL (MUC-16; SEQ ID NO:62), QQTHGITRL (MUC-16; SEQ IDNO:63), LQPLSGPGL (MUC-16; SEQ ID NO:64), TLDRDSLYV (MUC-16; SEQ ID NO:65), QLYLELSQL (MUC-16; SEQ ID NO:66), KVLEYVIKV (MAGE-I; SEQ IDNO:67), KVADLVGFL (MAGE-I; SEQ ID NO:68), and KTWGQYWQV (SEQ ID NO:70).
Selected peptides may be presented to the cells via a variety of means and methods in the art. Selected peptides may be presented in a manner that allows them to enter an intracellular pool of peptides. For example, peptides may be presented via osmotic loading. Preferably, peptides are added to the xAPC system culture medium. The peptides may be added to the culture medium in the form of an intact polypeptide or protein that is subsequently degraded via cellular processes, such as, for example, enzymatic degradation. Alternatively, the intact polypeptide or protein may be degraded via some other means such as chemical digestion (e.g., cyanogen bromide) or proteases (e.g., trypsin and chymotrypsin) prior to addition to the xAPC system culture medium. Alternatively, an entire protein or polypeptide sequence may be cloned into an appropriate vector and inserted into a prokaryotic cell, whereby the cell generates significant amounts of the antigenic polypeptide that are then harvested, purified, and digested into peptides that are then added to the xAPC system culture medium.
A sufficient amount of each selected peptide may be added to the cell culture to allow the Class I MHC molecules to bind and subsequently present a large density of the peptide on the surface of human Class I MHC-expressing cells.
The xAPCs may be assayed for enhanced APC function compared to the APC function of nonxenogenic or endogenous antigen presenting cells. Enhanced APC function may be determined by measuring any of a variety of parameters of CD8+ T cell activation, such as, for example, a degree of one or more cell surface proteins expression, which is indicative of CD8+T cell activation, such as CD69 cell surface expression, a degree of differentiation, a degree of cyotoxic killing ability, a degree of specific cell lysis, and a degree cytokine production.
Purification of proteins and peptides maybe achieved through various techniques that are known in the art, such as immunoaffinity chromatography, affinity chromatography, protein precipitation, buffer exchanges, ionic exchange chromatography, hydrophobic interaction chromatography, size exclusion chromatography. Antigen- stimulated CTLs may be detected or isolated by peptide-MHC pMHC tetramer staining, wherein detected CTLs are specific for a selected peptide presented by the xAPCs. Peripheral blood leukocytes (PBLs) are obtained from a subject, and preferably substantially purified. Methods for purification of PBLs include, methods emplying Ficoll gradients may be utilized for this purpose. The purified PBLs are then mixed with xAPCs cells preincubated with the appropriate antigenic peptides. Preferably, PBLs are purified by magnetic bead purification systems that are in the art, such as Miltenyi beads (Myltenyi Biotec) and Dynabead systems (Dynal Biotech). PBLs may also be purified via cell sorting procedures, such as with fluorescence assisted cell sorter (FACS)-based methods, or other appropriate cell sorting devices and methodology. Such cell sorting methods or sorting of red blood cells by using green fluorescent protein (GFP) as a marker for various cell- specific proteins.
Naive T cells are incubated in culture with the appropriate xAPCs and loaded with selected peptide for a time period sufficient to activate and further enrich for a population of CD8+ cells. For example, U.S. Patent No. 4,690,915 describes a method of obtaining large numbers of lymphocytes via lymphocytopheresis. Preferably, the CD8+ cells are activated in an antigen-specific manner. The ratio of resting or precursor CD8 (effector) cells to antigen-presenting cells may vary from individual to individual and may further depend upon variables such as the amenability of an individual's lymphocytes to culturing conditions and the nature and severity of the cancer. Preferably, however, the lymphocyte:antigen-presenting cell (e.g., Drosophila cell) ratio is preferably in the range of about 30:1 to 300:1. For example, in one embodiment, 3xlO7 human PBL and IxIO6 live Drosophila cells are admixed and maintained in 20 ml of RPMI 1640 culture medium.
The effector/antigen-presenting culture may be maintained for as long a time as is necessary to activate and enrich for a population of a therapeutically useable or effective number of CD8+ cells. With a maximum specific CD8+ activation level generally being observed after one to ten days of culture, e.g., after five days of culture, a preferred time is from about three to seven days. In one embodiment of the present invention, in vitro activation of CD8+ T cells may be detected within a brief period of time after transfection of a cell line. In one embodiment, transient expression in a transfected cell line capable of activating CD8+ T cells is detectable within 48 hours of transfection. Thus, either stable or transient cultures of transformed cells expressing human Class I MHC molecules are effective in activating CD8+ T cells. Activated cytotoxic T lymphocytes may be effectively separated from the xAPCs (e.g., Drosophila cells) using a suitable method known or available in the art. For example, monoclonal antibodies specific for the artificial APCs, for the peptides loaded onto the artificial APCs, or for the CTLs (or a segment thereof) may be utilized to bind their appropriate complementary ligand. Antibody-tagged cells may then be extracted from the stimulator-effector cell admixture by any of a variety of methods in the art, such as, for example, immunoprecipitation and immunoassay methods. Alternatively, a separation step may be omitted completely and the inactivated xAPCs may be left in culture with the activated CTLs.
Therapeutically effective, cytotoxic amounts of the activated CTLs may be selected as suitable for the described in vitro and in vivo use, e.g., in view of the amount and type of cells that are the ultimate target of these CTL cells. The amount will also be selected in view of the condition of the patient and maybe determined via consideration of all appropriate factors by the practitioner. Preferably, about IxIO6 to about IxIO12, more preferably about 1x108 to about 1 x 10π, and even more preferably, about IxIO9 to about IxIO10, activated CD8+ cells are utilized for adult humans, compared to about 5xlO6 -5x107 cells used in mice.
Preferably, the activated CD8+ cells, which are CTLs, are harvested as described above from the xAPC culture prior to administration of the CTLs cells to the individual being treated. The cell culture system is preferably not tumorigenic. Therefore, if complete separation of Drosophila cells and activated CD8+ cells is not achieved, there should be no inherent danger associated with the administration of a small number of Drosophila cells.
Naϊve CD4+ T cells or CD8+ T cells, or both CD4+ T cells and CD8+ T, cells may be preferably extracted from a subject prior to incubation with the xAPC cultures. Subjects may undergo any of a variety of known or available blood cell separation procedures (e.g., leukopheresis) to collect white blood cells.
Naϊve T cells may be harvested from a subject prior to the initiation of other treatment or therapy that may interfere with, attenuate, or in any way limit the specific activation of naϊve T cells methods and uses as provided in the present invention. For example, in the treatment of an individual with a neoplasia or tumor, a sample of naϊve T cells may be obtained prior to the initiation of chemotherapy or radiation treatment and kept in culture. After the naϊve T cells are activated with peptide-loaded xAPCs, the naϊve T cells may be expanded and activated, and the activated CTLs may be introduced back into the subject. Alternatively, naive T cells may be activated, and the activated CTLs may be introduced back into the subject from whom the naϊve T cells were obtained before, after, or in conjunction with other optional forms of treatment, such as chemotherapy or radiation.
The activated CTLs may also be suspended into an appropriate vehicle for delivery and infusion into a subject. Techniques of reintroducing cellular components are known in the art and include procedures such as those exemplified in U.S. Pat. Nos. 4,844,893 and 4,690,915. For example, administration of activated CTLs cells via intravenous infusion may be employed. Multiple infusions may be required, and these infusions may occur over a period of several weeks or longer.
In the treatment regimens of the invention, cytokines are also administered to attain CTL persistence and thereby enhance half-life, activity, potency, and/or selectivity properties of the CTLs that are administered to a subject. Such persistence may result from a direct effect on the CTLs or from an indirect effect that involves upregulation of antigen expression on target cells of the CTLs. Preferred cytokines are IFN-α-2b and IL-2.
Cladribine (2-CdA, Leustatin®) and/or DAB IL-2 (ONTAK®) lymphodepleting agents are also administered in the inventive treatment regimens. These agents are non- myeloablative and elicit a transient immunosuppression in subjects receiving the CTL therapy.
The timing and duration of the administration of each of the CTL, cytokine, and lymphodepleting agents may be selected by the artisan based on routine experimentation and the guidance herein, including the examples below.
To illustrate various aspects and features of the invention, the following examples are provided.
EXAMPLES Ex-Vivo Preparation of CTLs
Xenogenic APC (xAPC) lines are generated from Schneider S2 cells (S2 cells), which were established in 1969 from several hundred 20- to 24-hr old Oregon-R (wild type) Drosophila melanogaster (Oregon-R) embryos (ATCC CRL-1963), according to published procedures (Schneider, J. Embryol. Morph. 27:353-365,1972). The S2 cell line has been deposited with the American Type Culture Collection (CRLl 0974). The original supply of S2 cells used to derive the cell lines from which the xAPCs derived are obtained from this source. In order to generate xAPCs, S2 cells are transfected with vectors derived from plasmid vector pRMHa-3 (see, e.g., U.S. Patent No. 6,225,042). One xAPC line, designated clone A, is transfected with vectors encoding HLA- A2.1 Class I, B7.1 and ICAM-I . A second xAPC line, designated clone B, is transfected with vectors encoding HLA-A2.1 Class I, B7.1, B7.2, ICAM-I and LFA-3. A third xAPC cell line, designated clone C, is transfected with vectors encoding HLA-A2.1 Class I5 B7.1, ICAM-I, LFA-3, and CD70. Thus, clone A expresses HLA-A2, B7.1, and ICAM-I, clone B expresses HLA-A2.1 Class I, B7.1, B7.2, ICAM-I and LFA-3, and clone C expresses HLA- A2.1 Class I, B7.1, ICAM-I, LFA-3, and CD70.B7.2 and LFA-3.
Independent continuous cultures of clone A- and clone B-descended cells are maintained in Schneider's medium supplemented with 10% fetal calf serum and 500 μg/ml geneticin (G418) and are split twice a week with fresh media added during each split to adjust cell density to approximately 1 x 106 cells /mL. Approximately one day prior to induction (day —2 to -4; day 0 defined as the day cells are tested for expression of exogenous molecules and are loaded with peptide), 3 x T75 flasks are inoculated with a volume of cell suspension maintained in stock cultures equivalent to 1.5 x 107 cells/flask. Complete Drosophi Ia-SFM medium without G418 is added to bring the volume up to 15 ml/flask. Flasks are then incubated in a chamber at approximately 27°C. On approximately day -1 to —3, cells are then induced by addition of copper sulfate (CuSO4) to a final concentration of 1.0 mM (1:200 dilution of 200 mM stock OfCuSO4; 75 μl OfCuSO4 for each T75 flask containing 15 ml of cell suspension) and returned to the 27°C chamber. The induction time lasts for approximately 24 to 72 hours.
On day 0, flasks containing induced cell cultures are checked visually and microscopically for evidence of contamination. Uncontaminated flasks are pooled and viable cells counted. Samples of pooled cell cultures of approximately 6x106 cells are evaluated by flow cytometry using fluorescence assisted cell sorter (FACS) analysis to determine the level of expression of exogenous molecules. Cell cultures (approximately 1 xlO7 cells/mL) are then tested to verify expression exogenous HLA-A2.1, B7.1 and ICAM-I (for clone A cells) or HLA-A2.1, B7.1, B7.2, ICAM-I and LFA-3 (for clone B cells) prior to peptide loading. Once expression of exogenous molecules is verified, each cell culture is washed by splitting each culture into two sterile 50 ml conical tubes. Each tube is then filled with HYQ SFX-Insect medium (Hyclone) and centrifuged at 1,700 rpm (600 x g) for approximately seven minutes to pellet the cells. Supernatants are discarded, and the tubes containing cell pellet are again centrifuged at 1,700 rpm (600 x g) for approximately one minute. Supematants are removed with a fine tipped pipette. Pellets from each split cell culture are then recombined and resuspended in 8 mL of SFX Insect medium to a cell density of approximately 1 x10 cell/mL. Approximately 40 μL of a β2 microglobulin stock solution at 1.0 mg/mL and 24 μL of 1 : 50 dilution of a stock peptide combo solution at 1.67 mg/mL for each peptide is added to each resuspended culture. Thus, each cell culture suspension contains β2 microglobulin at a final concentration of approximately 5 μg/mL and each selected peptide to be loaded onto xAPCs at a final concentration of approximately 0.1 μg/mL per peptide. Cell cultures are incubated in the suspension containing β2 microglobulin and peptides for at least four hours, and no more than eight hours, with swirling every 30 minutes at room temperature. After the peptide incubation period, approximately 1 mL aliquots of each cell culture are distributed separately into eight polypropylene tubes (Falcon 2006). Any residual cell culture suspension is discarded.
CD8 cells isolated from leukapheresis samples by positive selection with an anti- CD8 antibody are stimulated against human melanoma-associated peptides (tyrosinase-689.369-377. tyrosinase-792369-3775 MART- 1-81927-35, gpl00-817209-2i7, gplOO- 8I8280-288 and gp 100-853154.162) presented by Drosophila cells expressing human Class I and co-stimulation/adhesion molecules HLA- A2.1 , B7.1 , CD70 , LFA-3 and ICAM- 1. CD8+ cells are restimulated for two rounds with autologous monocytes (pulsed with the described epitopes) in the presence of IL-2 and IL-7. The number of effector cells is then increased by non-specific stimulation with anti-CD3 monoclonal antibody in the presence of gamma-irradiated autologous feeder cells and IL-2. Cytotoxic T lymphocyte activity is measured against peptide-loaded T2 cells and a panel of melanoma cells, while the purity of in v<ϊro-stimulated CD8+ T cells is assessed by flow cytometry. Materials.
Figure imgf000015_0001
Reagents rhIL-7. Recombinant human interleukin-7 (IL-7) is a lymphokine produced in E. coli and purified by the supplier (PeproTech) using high performance liquid chromatography (HPLC) but not antibody. IL-7, received as a powder, is diluted in sterile DPBS containing 1% human serum albumin. The bulk solution is then filtered through a 0.2-μm filter, aliquoted (30,000 U/mL, IOOOX concentration) into sterile vials, and stored at -800C prior to use. rhIL-2. Recombinant human interleukin-2 (IL-2) is produced by recombinant DNA technology and supplied by Chiron Corporation (Proleukin®). IL-2, received as a powder, is diluted in IL-2 diluent (0.5% human serum albumin in 50 mM acetic acid), filtered through a 0.2-μm filter, aliquoted into sterile vials (20,000 U/mL, 1000 X concentration), and then stored at -800C prior to use.
Tyrosinase PeptideYMNGTMSQV. A tyrosinase peptide (tyr 369-377), corresponding to amino acids 369-377 of human tyrosinase, is manufactured and purified using GLP compliance standards (Synpep Corporation). The peptide powder as received from the manufacturer (Synpep Corporation) is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0C to —88 0C prior to use. This stock peptide solution is mixed in equal parts with other peptide stock solutions (also at a concentration of 10 mg/ml) to generate combination peptide solutions for use in loading xAPCs. The combination peptide solutions are aliquoted into sterile vials in a Class 10,000 clean room under aseptic conditions in a Class II biosafety cabinet.
Tyrosinase PeptideYMDGTMSQV. A deamidated form of the tyr 369-377 peptide described above, which contains an aspartic acid residue in place of an asparagine residue at position three of the peptide, is manufactured and purified using GLP compliance standards (Synpep Corporation). This deamidation form is called tyr 369- 377d. The peptide powder received from the manufacturer is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0C to -88 0C prior to use. gplOO Peptide ITDQVPFSV. A gplOO peptide (gpl0020<>-2i7), corresponding to amino acids 209-217 of human gp-100, is manufactured and purified using GLP compliance standards (Synpep Corporation). The peptide powder is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0C to —88 0C prior to use. gplOO Peptide KTWGQYWQV. A gplOO peptide (gp 100154-162), corresponding to amino acids 154-162 of human gplOO, is manufactured and purified using GLP compliance standards. The peptide powder as received from Synpep Corporation is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -720C to -88 °C prior to use. gplOO Peptide YLEPGVTA. A gplOO peptide (gplOO28o-288), corresponding to amino acids 280-288 of human gplOO, is manufactured and purified using GLP compliance standards by Synpep Corporation. The peptide powder is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0C to -88 0C prior to use.
MART-I Peptide AAGIGILTV. A MART-I peptide (MART-127-35), corresponding to amino acids 27- 35 of human MART-I, is manufactured and purified using GLP compliance (Synpep Corporation). The peptide powder is dissolved in dimethylsulfoxide (DMSO) to achieve a stock peptide solution at a concentration of 10 mg/mL, and is stored at -72 0C to -88 0C prior to use.
DYNABEADS® M-450. DYNABEADS® M-450 (SAM) IgG are sterile paramagnetic beads coated with polyclonal sheep anti-mouse IgG that bind the primary mouse IgG. DYNABEADS, available from Baxter Oncology Inc., are stored at 40C prior to use in T cell isolation using the Isolex 300i Magnetic Cell Selector.
Human Serum Albumin. 25 % HSA, USP (Baxter Fenwal Laboratories; the plasma source for each lot tested to be negative for HIV-I HIV-2, HCV, and HBV), is stored at RT prior to use as a source of buffered protein during the following T cell preparation and activation steps steps: purification of CD8+ T cells and CD8' T cells; peptide-loading of adherent cells; and final formulation of activated T cells.
Anti-CD8 Antibody. Anti-CD8 monoclonal antibody (37B1A) is a murine monoclonal antibody directed against the CD8 antigen of T cells, which is used to select CD8+ T cells with the Isolex 300i Magnetic Cell Selector System. The concentrate solution is diluted in sterile DPBS for use in CDS+ T cell isolation or activation processes. The bulk solution is filtered through a 0.2-μm filter and then aliquoted into single-use vials in a Class 10,000 clean room under aseptic conditions in a Class II biosafety cabinet. Aliquots (10.0 mg/mL) are stored at -800C prior to use.
CD8 Alpha Chain Peptide - AAEGLDTQRFSG. CD8 alpha light chain peptide (AAEGLDTQRFSG) is purified and manufactured under GLP compliance standards. The CD8 alpha chain peptide is used in CD8+ T cell isolation processes to release CD8+ T cells captured using CD8 (37B1A) antibody and the Isolex 30Oi Magnetic Cell Selector. Each lot of peptide is manufactured by Synpep Corporation to meet pharmaceutical grade standards, and is tested for peptide sequence, purity, molecular weight, and appearance. The CD8 alpha chain peptide, received as a powder, is further processed to create a stock solution of 10 mg/ml. This stock solution is diluted in DPBS, filtered through a 0.2-μm filter, aliquoted into sterile vials, and stored at -72 0C to -88 0C prior to use. Vialiπg of the peptide reagent is performed in a Class 10,000 clean room under aseptic conditions in a Class II biosafety cabinet.
Human β2M Microglobulin. A concentrate of human beta— 2 microglobulin (β2M) produced by recombinant DNA technology is diluted in sterile DPBS to achieve a concentration of 1.0 mg/mL. The bulk solution is then filtered through a 0.2-μm filter, aliquoted into sterile vials and stored at -8O0C prior to use during the preparation and peptide-loading of xAPCs and peptide loading of adherent cells.
Sodium Citrate Solution. A sterile, nonpyrogenic anticoagulant sodium citrate solution, USP (Baxter Fenwal), is stored at room temperature (RT) prior to use as a buffer additive for running the Isolex 30Oi Magnetic Cell Selector for selection of CD8+T cells and CD8" T cells.
Schneider 's Medium. Schneider's Drosophila medium is a culture medium used for culturing Drosophila cells. Each lot of medium is tested by the supplier (Invitrogen Corporation) for osmolality, pH, sterility, and the ability to sustain the growth of Drosophila cells. Schneider's Drosophila medium (IX concentration) is stored at 20C to 6°C prior to use.
Fetal Bovine Serum. Fetal bovine serum (FBS), which is used as a protein source for the growth of host cells or xAPCs cells, is stored at — 800C. The FBS, available from Gemini Bioproducts, is processed from bovine fetal blood from animals of United States origin. The maternal animals from which the blood is derived are free of infectious and contagious diseases and injurious parasites.
HYQ SFX Insect Medium. Hyclone's SFX Insect Medium (Hyclone Corporation) is a serum- free culture medium (IX concentration) used during the peptide loading of xAPCs, and is stored at 20C to 6 0C prior to use. This medium does not contain products of bovine origin.
Copper Sulfate. Copper (II) sulfate pentahydrate (Aldrich) is used for induction of modified host cells to express human HLA, co-stimulatory, and adhesion molecules. The stock solution is formulated by dissolving the crystalline CuSO4 in endo toxin-free sterile water to achieve a concentration of 200 mM and aseptically filtering the solution through a 0.2-μm filter into a sterile container in a Class II biosafety cabinet. The filtered stock solution is stored at 2 0C to 60C prior to use.
RPMI. RPMI culture medium (available from Invitrogen Corporation or Gibco), which is serum- and antibiotic-free (IX concentration), is used to grow T cells. RPMI culture medium is stored at 2 to 60C prior to use.
Dulbecco 's Phosphate Buffered Saline (DPBS). Sterile, non-pyrogenic Dulbecco's phosphate buffered saline (DPBS) solution (available from Invitrogen Corporation or Gibco, IX concentration) is stored at RT prior to use. DPBS is used for the following procedures: running the Isolex 300i Magnetic Cell Selector instrument during the selection of CDS+T cells and CD8" T cells; washing non-adherent cells during restimulation steps and washing unbound OKT3 monoclonal antibody during non-specific expansion; and diluting human β2 microglobulin, IL-7, CD8 peptide, and OKT3.
Leibovitz' s Medium. Leibovitz's L-15 medium (without L-glutamine; IX concentration), available from Sigma- Aldrich, is stored at 20C to 6 0C prior to use during peptide-loading of the T cell activation process.
OKI® 3 Antibody. Orthoclone OKT®3 (l.Omg/mL), a murine monoclonal antibody specific for the CD3 antigen of T cells supplied in ampoules as a sterile solution approved for clinical use (available from Ortho), is aliquoted into single-use vials under sterile conditions and stored frozen at— 80°C prior to use in the activation of T cells.
Geneticin (G418). Geneticin (Invitrogen Corporation) is a selective antibiotic used in the culture of Drosophila cells for maintaining expression of exogenous molecules encoded by xenogenic nucleic acid. Geneticin is supplied as a sterile stock solution (50 mg/mL), and is stored at 2 to 6 °C prior to use.
Calcium Chloride. Calcium chloride hydrate is used for clotting of autologous plasma obtained from the lymphopheresis product to generate autologous serum used in CD8+ T cell isolation or activation processes. Calcium chloride hydrate is received as a crystalline powder, is compounded into a stock solution (IM), and stored at 2 °C to 6 0C prior to use. The stock solution is formulated by dissolving calcium chloride in endotoxin-free sterile water and aseptically filtering through a 0.2-μm filter into a sterile container in a Class II biosafety cabinet. Acetic Acid. Acetic acid (17.4M) used for the preparation of stock solutions of IL-2 is obtained from Sigma Corporation and stored at RT prior to use.
FICOLL-PAQUE ® Plus. Following isolation of CD8+ T cells and CD8" T cells with the Isolex 300i Cell Selector System, mononuclear cells from the non- CD8+ fraction are further fractionated using FICOLL-PAQUE ® Plus (IX concentration), a Ficoll gradient reagent without any animal components available from Amersham Pharmacia Biotech used to remove dead cells, neutrophils, and red blood cells. The reagent is stored at RT prior to use.
PENTASPAN®. PENTASPAN (B. Braun Medical Inc) is a sterile solution of 10% pentastarch in 0.9% sodium chloride for clinical use (NDC 0264-1972-10), and is stored at RT prior to use. It is used (IX concentration) as a cryoprotectant in the cryopreservation of isolated CD8" T cells and CD8 T cells.
Dimethyl Sulfoxide (DMSO). DMSO is used as a cryoprotectant in the cryopreservation of isolated CD8" T cells and CD8+T cells. The DMSO solution (IX concentration), available from Sigma- Aldrich, is stored at RT prior to use.
L-Glutamine. L-Glutamine (USP), 20OmM (10OX concentration), available from Invitrogen Corporation, is used as an RPMI culture medium supplement, and is stored at -8O0C prior to use.
MEM Sodium Pyruvate Solution. MEM sodium pyruvate solution (10OmM, IOOX concentration), available from Invitrogen Corporation, is used to supplement RPMI medium, and is stored at 2 to 60C prior to use.
Non-Essential Amino-Acids. Non-essential amino-acids (l OmM; IOOX concentration) from Invitrogen Corporation, used to supplement RPMI medium, are stored at 2 to 6°C prior to use.
HEPES Solution. IM (200X concentration) HEPES buffer solution
(Invitrogen Corporation), used to supplement RPMI medium, is stored at 20C to 6 0C prior to use.
X-Vivo 10-Cell Medium. X-vivo 10-cell culture medium, supplied by BioWhittaker, is stored at 2 0C to 60C prior to use. This medium (IX concentration), which is serum-, phenol red-, and antibiotic-free, is used during the phase of non-specific expansion of T cells activated by exposure to peptide-loaded xAPCs. Sodium Chloride Injection. A 0.9% sodium chloride solution, USP, available from Baxter Fenwal Laboratories, is used for cell washing procedures during harvesting of T cells. The solution, which is sterile, non-pyrogenic, and free of animal components, is stored at RT prior to use.
Dextrose + Sodium Chloride Solution. An injectable solution of 5% dextrose and 0.9% sodium chloride, USP (Baxter Fenwal Laboratories), is obtained as a sterile, non-pyrogenic solution free of animal components. The solution, which is used as a storage buffer for activated T cells, is stored at RT prior to use.
Lactated Ringer's Solution. A 0.9 % Lactated Ringer's solution, USP (Baxter Healthcare Laboratories), which is a sterile, low-endotoxin solution of calcium chloride, potassium chloride, sodium chloride, and sodium lactate in water for injection (free of animal components), is stored at RT prior to use in harvesting and suspending T cells.
Distilled Water. Cell culture grade distilled water, which is obtained by membrane- filtering and endotoxin-screening (Invitrogen Corporation), is used as a solvent for the preparation of stock solutions of copper sulfate, calcium chloride, and interleukin— 2 (IL-2) and is stored at RT prior to use.
Other Materials.
Lymphopheresis products are collected from human subjects diagnosed with melanoma and are stored at RT prior to use for the generation of an autologous, patient- specific cell product.
Autologous human serum is used as a protein source for culturing of isolated T cells. Autologous human plasma is prepared from lymphopheresis product by adding calcium chloride to achieve fibrin clotting and then collecting the liquid serum phase. The collected liquid serum phase is stored at 40C for short-term storage and at -8O0C for long- term storage.
A Drosophila xAPC line, derived from the xenogenic Drosophϊla clone B, which is used as a seed stock to create a continuous Drosophila xAPC culture, is obtained as described below. Procedures.
Isolation of Human CD8+ Cells. CD8+ cells are isolated from leukapheresis samples using an Isolex 30Oi machine (Baxter) by positive selection with an anti-CD8 monoclonal antibody (antibody 1) followed by a Dynabeads™ (Dynal) isolation procedure using sheep anti-mouse IgG (antibody2) coated onto magnetic beads (SAM beads). The anti-human CD8 mouse monoclonal antibody is added to washed cells resuspended in Dulbecco's PBS supplemented with 1% HSA (Baxter-Hyland) and 0.2% sodium citrate. Dynal magnetic beads (Dynabeads™) are added at a bead to cell ratio of 1:1 to 1 :2 depending on the number of PBMCs. The isolated CD8+ cells are removed by magnetic separation. The remaining non-CD8 fraction is collected and cryopreserved for future use during the restimulation and non-specific expansion steps. Dissociation of the CD8 cell-antibody l-antibody2 -bead complex is achieved by incubation at 37°C for 45 minutes in the presence of CD8 peptides9-7o (AAEGLDTQRFSG), the peptide to which antibodyl was generated. Released beads are removed magnetically and the CD8+ cells are counted and analyzed by flow cytometry to evaluate purity. Recovery of CD8+ cells is typically greater than 80%. Heat- inactivated serum is prepared by collecting the autologous plasma at the time of the initial cell washing step. Fibrin is clotted using CaCl2 and the fibrin clot removed. Serum is heat-inactivated, filtered, aliquoted and frozen at -800C. Non-CD8+ cells are retained from the positive selection procedure and purified using a Ficoll gradient. These cells are cryopreserved in DMSO, Pentaspan, and heat-inactivated autologous serum and stored in liquid nitrogen (LN2). These cells are used as the source of adherent cells at the time of the restimulations and are peptide-pulsed prior to use as antigen presenting cells.
In vitro immunization of purified human CDS+ T cells
Primary Stimulation. Transfected Drosophila S2 cells are incubated in Schneider's medium (106 cells/mL) supplemented with 10% fetal calf serum and copper sulfate at 27°C for 24 to 72 hours. S2 cells (clone 1120-3-9 ) are harvested, washed and resuspended in HYQ SFX-Insect medium (Hyclone) containing 0.1 μg/mL each of human gpl00i54-i62, gpl00209-2i7, gpl0028θ-288, MART-I27-35, tyrosinase-N369-377 and tyrosinase- D369-377 peptides and 5μg/mL human β2 microglobulin recombinant protein purified from E. coli. Following incubation at room temperature (23-25°C) for four hours, the S2 cells are mixed with CD8+ cells at a ratio of 1:10 {Drosophila cells:T cells) in Roswell Park Memorial Institute (RPMI) medium (Gibco) supplemented with 5-10% autologous serum. The cell mixture is incubated at 37°C during which time the Drosophila cells die off (by 48 hours). On day four, IL-2 (20 U/mL) and IL-7 (30 U/mL) are added to selectively expand the melanoma-specific CTL population.
Restimulations. Autologous, CD8-depleted PBMCs, obtained at the time of lymphapheresis and frozen for future use, are thawed, washed and resuspended at 107 cells/mL in RPMI medium containing 10% autologous serum, 5μg/mL recombinant human β2 microglobulin and 5 μg/mL of gpl00i54-162, gpl00209-2i7, gpl0θ28o-288, MART-l27-3S, tyrosinase-N369-377 and tyrosinase-D369.377 peptides. Following γ-irradiation (5,000 rads), the cells are incubated at 37°C for two hours in the flasks used for the restimulation. Nonadherent cells are removed by washing with Dulbecco's PBS. Adherent monocytes are loaded with the peptide epitopes and incubated for 90 minutes in Leibovitz medium containing 5 μg/mL human β2 microglobulin in 1% HSA and 5 μg/mL of each peptide. The supernatant is removed and the Drosopfa/α-activated CD8+ cell suspension (2.5 x 106 cells/mL in RPMI medium with 10% autologous serum) is added at a ratio of about 10 CD8+ cells to one adherent monocyte. After three to four days of culture at 37°C, IL-2 (20 U/mL) and IL-7 (30 U/mL) are added with a medium change to selectively expand the melanoma-specific CTL population.
Non-specific Expansion. The CD8+ effector cells that have undergone two rounds of restimulation are expanded in cell culture bags along with feeder cells (irradiated autologous non-CD8+ selected cells) after being stimulated with OKT3 antibody. Frozen non-CD8+ selected cells are thawed, washed and then gamma-irradiated (3,500 rads). A ratio of 4: 1 (feeder: effector) is placed in T-225 flasks that have been coated with OKT3 antibody. OKT3 stimulation is performed in complete RPMI medium containing 10% autologous serum supplemented with 20 U/mL of IL-2. Two days later, the stimulated T cells are diluted with fresh media (X-vivo 10 medium) and transferred to cell culture bags for expansion. Fresh media and IL-2 are supplemented approximately every two to three days to feed the rapidly expanding T cells.
Preparation and Release of CTLs Cell Harvest and Final Product Formulation. Harvest of the final cell product is performed by centrifugation to remove culture media and to concentrate the cells. After centrifugation, cells are washed in saline containing 1% human serum albumin (HSA), filtered through a 70-μm filter, and then diluted in infusion medium. The cell product for infusion contains autologous CTLs in 30OmL Lactated Ringer's Injection, USP (76% v/v), 5% Dextrose in 0.9% Sodium chloride (4% v/v) and 25% HSA (20% v/v). Final product cells are packaged in a 1000 mL transfer bag in a chilled insulated shipping container with a temperature data logger.
Product Testing. Tests performed before release of CTL product are listed below. Specific activity directed against individual peptides is assessed by measuring the cytotoxic activity of the effector cells against chromium-labeled T2 cells loaded with each individual peptide used for stimulation. Cytotoxic activity is also measured against chromium-labeled melanoma targets (Malme 3M, M 14), using normal fibroblasts (Malme 3)or a melanoma cell line (01 -KN) negative for tyrosinase, gp 100 and MART-I as controls.
Phenotype: A portion of the cell harvest is used to test the phenotype of the CD8 product prior to shipment. The final product specification is CD3, CD8 and TCR expression. Additional phenotypic evaluation for markers consistent with memory, activation, etc. are measured and recorded.
Identity: The HLA-A2, HLA-C and HLA-DR status of the sample is determined by analysis of a DNA preparation isolated from a PBMC sample prepared at the time of receipt of the initial leukapheresis and from the CD8+ T cells collected at the end of the culture, taken from the harvested cell product. PCR analysis of DNA samples is performed with HLA-specific primer oligos provided by Genovision. The final product specification is identity with day 0 sample.
Cell Counts: The desired cell dose for infusion is 1010 CD8+ T cells. The final product infusion limit is 1010 cells. The total number of cells generated is patient-dependent. Cell doses averaged 9 X 109 in a previous study where the cells were generated under the same ex vivo protocol. The final cell count is made by determining the number of viable cells. Viability: The viability of the final cell dose is >70%. The number of viable cells is established by microscopically counting cells that exclude trypan blue dye.
Mycoplasma Testing: Testing of the cells for mycoplasma is performed before the cells are administered to the patient to confirm negative mycoplasma detection. The Roche PCR kit is used which also includes an ELISA assay. In addition, samples of the T cell cultures are collected at the first cytokine feeding (day 6 ) and sent to BioReliance to fulfill the 28-day culture procedure. The results of the culture are received post T cell infusion.
Endotoxin testing: The endotoxin test on the final cellular product is performed using a Limulus Amebocyte Lysate assay (BioWhittaker, Walkersville, MD). The endotoxin level limit is less than 5 EU/kg infused into the patients. The final cell product specification is <lEU/mL.
In Process Product Sterility Tests: In process testing for sterility is performed using the BacT/ Alert system. Samples are taken during times of media changes. A sample from the final dose is also tested by the BacT/Alert system, and the final product specification is absence of growth.
Drosophila DNA and Insect Viral RNA Detection: DNA is isolated from the final CD8+ T cell product and used as a template for a Drosophila DNA PCR assay, which uses primers specific for the insect vector used to prepare the recombinant Drosophila APCs. This sample is compared to the naϊve CD8+ sample that acts as the negative control and Drosophila cell DNA is used as the positive control. Total RNA is also isolated from the same CD8+ T cell samples and a quantitative real time RT-PCR, which can detect 20 copies/μg of cDNA of three different, known insect viral RNA viruses, is performed. The product specification for both assays is negative detection of insect genomic and viral nucleic acid.
Lytic Activity: Lytic activity is evaluated for the final CD8+ T cell product by a chromium-release assay in both peptide-loaded T2 cell targets, to measure anti- peptide activity, and established or autologous melanoma cell HLA- A2 targets and donor-matched tumor and non-tumor lines (Malme 3 and Malme 3M) to asses melanoma-specific killing. The product specification is specific killing of target cells.
Also, to facilitate monitoring the following tests may also be performed in addition the release tests described above:
Tetrameric Staining: CD8+ T cells for the naϊve leukapheresis-derived sample is evaluated for the presence of antigen-specific T cells with tetrameric molecules which have been designed to enumerate T cells with specificity for the peptide epitopes used to generate the CTLs ex vivo. This allows the monitoring of antigen-specific T cells after treatment.
Melanoma Tumor Cells in Peripheral Blood Samples: Sensitive, quantitative real time PCR assays allow for the monitoring of circulating melanoma cells in patient blood samples, before, during and after drug treatment. While not all stage III/IV melanoma patients demonstrate circulating tumor cells, the detection of these cells can be used as a surrogate marker for response to therapy. CTL products prepared as described above may be used in the combination therapy treatment regimens of the invention as illustrated by the preferred embodiments described in the following examples.
Treatment of Patients with Metastatic Melanoma Using Autologous CD8+ Lymphocytes Stimulated Ex Vivo with Drosophila Cells Loaded with Melanoma-Associated Peptides (MART-I, gplOO, and Tyrosinase) And Subcutaneous Administration ofIFNa-2b andIL-2, With and Without Administration of 2-CdA or DAB IL- 2
These examples illustrate treatments comprising the administration of autologous cytotoxic T lymphocytes (CTLs) that are specific for peptide epitopes derived from melanoma-associated antigens in combination with a lymphodepletion agent selected from cladribine and DAB IL2 as well as cytokines IL-2 and IFN-α— 2b, as well as a control treatment. The cell therapy agent, which comprises autologous CTLs that have been activated such that they specifically target melanoma cells expressing MART-I, gplOO, and tyrosinase by contacting Drosophila xAPCs loaded with MART-I-, gplOO-, and tyrosinase-derived antigenic peptides, is designed to enhance and maintain a patient's immune targeting of melanoma cells expressing MART-I, gpl 00, and tyrosinase. The addition of immunomodulators IFN-α-2b and IL-2 is included to augment the response of the CTLs. A non-myeloablative, but lymphocyte-depleting, preparative regimen (cladribine) or a specific T cell subset depletion denileukin diflitox (DAB IL-2) is administered prior to the CTL infusion to enhance the engraftment of the CTLs. The treatment is designed to cause tumor regression and clinical benefit through persistence of melanoma-specific CTLs.
More particularly, clinical patients with advanced, malignant melanoma that are HLA- A2 positive are administered a non-myeloablative, but lymphocyte-depleting, preparative regimen consisting of either cladribine (0.12 mg/kg/day x 5 days) or DAB IL-2 (a single injection of 18μg/kg). In addition, a concomitant regimen of interferon-α-2b (IFN-α-2b; 10MIU/m2) is administered daily, for 5 consecutive days, by subcutaneous injection prior to CTL infusion. Subsequently, the patients receive a single dose of ex vivo- generated, Drosophila xAPC-stimulated, autologous CD8+ T cells, which display the CTL phenotype. The cell infusion is immediately followed by subcutaneous daily administration of low-dose (e.g., 3MIU) IL-2, for twenty-eight (28) days, followed by tumor assessment. Subcutaneous, daily administration of low-dose (e.g., 3MIU) IL-2 is continued in patients without evidence of disease progression.
The CTLs are generated by a method in which purified naϊve CD8+ T cells are stimulated with Drosophila xAPCs presenting six different melanoma-associated T cell peptide epitopes in the context of human MHC class I restriction, resulting in CTLs that display multiple specificities for MART-I, gplOO, and tyrosinase, after approximately 34 days in culture.
Blood is collected at screening to qualify patients for the study and the preparative leukapheresis procedure takes place on day 0 or day 1. Patients undergo a standard 2.0- 3.0X the blood volume leukapheresis to obtain enough white blood cells required to prepare the cell therapy agent. At indicated times, additional leukapheresis procedures (< of third the amount used to obtain the initial CD8 sample) is be performed to obtain peripheral blood cells for analysis. These samples are used to monitor the presences of antigen-specific T cells and possible circulating melanoma tumor cells. Plasma and blood cells are also collected to meet the xenotransplantation requirement for archiving pre- and post-treatment samples. Leukapheresis products are transported to a facility where CD8+ T cells are isolated, stimulated and cultured for approximately 34 days. An evaluation of measurable lesions is conducted at both 4 and 8 weeks following the CTL infusion. An objective of the study is to determine whether administration of a full non-myeloablative, lymphocyte-depleting (cladribine) or a selective T cell subset depletion (DAB IL-2), prior to the T-cell infusion and in conjunction with an established immune-enhancing cytokine regimen, will result in antigen-specific T cell persistence with accompanying objective tumor regression.
Preparation of Cytotoxic Lymphocytes
CDS+ cells isolated from leuJcapheresis samples by positive selection with an anti- CD8 antibody are stimulated with human melanoma-associated antigenic peptides
(tyrOSinaSe369-377(native), tyrθSinase369-377(modι(Ted D37i), MART-127-35, gpl00i54-l63, gpl00209-2I7 and gpl0028o-288) presented by Drosophila xAPCs expressing human class I and co- stimulatory molecules (HLA-A2.1, β2 microglobulin, B7.1, CDlO , ICAM-I and LFA-3). These same CD8+ cells are restimulated by two rounds of autologous, peptide-pulsed mononuclear cells, in the presence of IL-2 and IL-7. A non-specific expansion step with anti-CD3 mAb (OKT3) is also included to increase the total number of cells: generally 25- fold greater than that obtained at the end of the second restimulation step. Cytolytic T cell activity is measured against peptide-loaded T2 cells and a panel of A2+ melanoma cells, while the purity of the in vz7ro-stimulated CD8+ T cells is assessed by flow cytometry. Additionally, interferon-gamma production, in response to antigen-specific stimulation and peptide-specifϊc tetrameric analysis, confirms the effector function and specificity of the generated CTLs, respectively.
Potency of CTLs Generated from Naϊve CDS^ T Cells Isolated from Melanoma Cancer Patients
It has been demonstrated in melanoma patients that antigen-specific CTLs, isolated from peripheral blood samples, are part of a heterogeneous population of cells that range from low to high avidity for the specific peptide/MHC complex to which the TCR is directed. However, the vast majority of these CTLs are of low avidity, and only the CTLs with high avidity demonstrate significant tumor cell lysis.21 In addition, tumor-infiltrating lymphocytes (TILs) have been isolated from tumor masses in patients with melanoma. The ex vivo expansion of these TILs in the presence of high-dose IL-2 has resulted in objective responses in melanoma patients; however, these responses were of short duration and the highly-reactive cloned T cells originating from the TILs failed to generate any complete responses.16
Ih contrast, CTLs generated ex vivo with Drosophila-xAVCs are potent, high- avidity, antigen-specific T cells capable of peptide-specific lysis (see, e.g., Figure 1) and strong melanoma tumor cell killing. The reproducible generation of the CTLs, with strong cytolytic activity for tumor cells, is believed to be attributable to several contributing factors. First, CD8+ purification with a peptide-specific, anti-CD8 monoclonal antibody selected for cells with a high density of CDS , while the antibody release in the presence of specific peptide, avoided non-specific stimulation of the T cells. Second, the highly- purified CD8+ T cells (Table I) are stimulated with xAPCs presenting specific T-cell epitopes in the context of MHC molecules, where a high density of antigen presentation results in a potent primary T-cell immune response. The inclusion of multiple costimulation molecules on the APCs optimizes the T cell stimulation. Third, the restimulation steps incorporate autologous, mononuclear cells that have been loaded with the same melanoma- associated T cell epitopes used in the primary stimulation and provide a boost to the proliferating T cells. Finally, a single non-specific expansion step with OKT3, autologous feeder cells and low-dose IL-2 maintains the same percentage of antigen-specific T cells recorded at the end of the second restimulation step, while increasing the total number of T cells ~ 25-fold.
Table I: Purification of CD8+ Cells by Positive Selection and Analyzed by Flow Cytometry (data represents summary of total of 28 patients)
PBMC Post-selection
Cell Type (%) (Range) (%) (Range)
CD3+ T cells 45 (29-64) 83 (59-98)
CD4+ T cells 27 (16-46) 2 (0.5-4)
CD8+ T cells 17 ( 8-31) 84 (60-96)
CD14+ monocytes 27 01-43) 5 (0.5-17)
CD15+/CD16+neutrophils 2 (0.5-9) 0.5 (0.5-1)
CD16"7CD15" NK cell- 15 ( 8-47) 8 ( 2-20)
CD^ B cells 1 (0.5-2) 1 (0.5-3)
This ex vivo xAPC CD8+ T cell stimulation protocol resulted in CTLs with potent tumor cell lysis, high avidity for the peptide/MHC complex, and antigen-specificity as assessed by enumeration with peptide-specific tetrameric molecules, interferon-gamma release in response to specific antigenic stimulus and specific tumor lysis.
Evaluation of CIL ΩierOpyinMdigimtiMetastaficMelamiiMβvmPr^^
Two phase I trials and one phase II trials have been conducted, which involved a total of 55 subjects with Stage III or Stage IV melanoma. Study 1, "Autologous Cytotoxic T Lymphocytes (CD8*) Serially Cultured Ex Vivo with Drosophila Antigen Presenting Cells, Interleukin-2, Interleukin-7, then Adherent Monocytes, and Tyrosinase Peptide", was an open-label study in 10 Stage IV melanoma patients. The clinical endpoints were: 1) safety and tolerability of reinfused autologous CTLs after in vitro immunization; 2) determination of the kinetics of the infused CTLs in the systemic circulation by limiting dilution analysis; 3) whole body disposition of ' ' ' indium-labelled CTLs by radioscintigraphy; 4) cell composition of biopsied nodules by immunohistochemical analysis (CTLs, TH, NK, B cells) and 5) regression of measurable lesions and duration of response over two months. 22 In Study 2, "Pilot Study of Subcutaneous Interferon- Alpha with Infusion of Drosophila Cell Stimulated Autologous CD8+ Lymphocytes for the Treatment of Advanced Melanoma", a total of 15 Stage III/IV melanoma patients were infused with autologous CD8+ T cells on background maintenance therapy with IFNα-2b. CTL therapy was evaluated by: 1) monitoring the safety and toleration of the reinfused CTLs; 2) cell composition of biopsied nodules; 3) regression of measurable lesions and 4) duration of response over three months. Subjects who had stable disease or demonstrated a clinical response at the follow-up evaluation (four weeks after the first infusion) were offered a second-cycle of treatment. Eight (8) of the fifteen (15) subjects underwent a second cycle of CTL therapy, four (4) subjects entered a third cycle and one (1) subject was treated with four cycles of T cell therapy.23
In Study 3, "Randomized Phase II Trial of Subcutaneous Interferon-α-2b (IFN) and Interleukin-2 (IL-2) with or without Infusion of Drosophila Cell-stimulated Autologous CD8 Lymphocytes for the Treatment of Advanced Melanoma", a total of 30 Stage ΪWW melanoma patients were treated with cytokines alone (IFNα and IL-2) or cytokines plus T cells. It was a randomized study in which patients entered the cytokines-only arm (Arm A) or cytokines plus T cells arm (Arm B). Patients who entered and progressed on Arm A were offered an opportunity to cross over to the cytokines plus T-cell arm (Arm C). The primary end point of this study was to compare the time to progression (TTP) of disease between the two groups (Arm A versus Arm B). Statistical significance was reached in TTP in patients entering Arm B versus those entering Arm A (Figure 11). In patients who crossed over from Arm A to Arm C, statistical significance in TTP was also detected in patients receiving T cells. Safety and tolerability of IFNα and IL-2 at the doses and on the schedule prescribed were also monitored.24
Rationale for Use of Interferon- a-2b (IFN-a-2b)
Interferon-alpha (IFN-oc) has a broad spectrum of immunomodulatory and antiproliferative effects in a variety of malignancies. It is believed that one mechanism of action of IFN-α is the upregulation of tumor antigen expression on melanoma cells. It has the ability to enhance the expression of immunologically important molecules on the surface of the tumor. These include the MHC antigens, accessory molecules, as well as tumor-associated antigens.25'27 These immunomodulatory effects may improve the activity of the immune system, including both antibodies and lymphocytes, to recognize and attack tumor cells in vivo. Active, specific immunotherapy has demonstrated significant clinical responses in the treatment of disseminated melanoma. The results of immune function studies discussed above have demonstrated that melanoma vaccine treatment increases the frequency of anti-melanoma CTLs. It is believed that these two modalities of immunotherapy may act synergistically. A five (5) day course of IFN-αpOMU/m2; subcutaneously) was included in Study 2 , and the results of IHC stains of tissue demonstrated that the timing and dose was sufficient to upregulate both class I and melanoma-associated antigen expression in serial biopsy samples obtained from a single patient at the specified time frames. The same 5-day course in clinical study was included in Study 3.
Rationale for the Use ofInterleukin-2
Human recombinant interIeukin-2 (IL-2) is a lymphokine produced by recombinant DNA technology that has been shown to exhibit a variety of biological activities. IL-2 stimulates the immune system and exerts its biological effects following binding to specific receptors on the surface of target cells. In vitro, it has been shown to enhance T cell proliferation and lymphocyte cytotoxicity, induce the killer activity of both lymphokine- activated and natural killer cells, and induce interferon-gamma production. Administration of high-dose IL-2 to 283 patients produced a remission rate of 7% with 9 complete responses, and the patients remained disease free from 9 to greater than 91 months.28 Although high-dose IL-2 appeared to be more effective than low-dose continuous infusions, high doses of IL-2 are also more toxic. The most common side effects were flu- like symptoms. The most severe side effects were hypotension, capillary leak syndrome, and reduced organ perfusion. Subcutaneous administration of low-dose IL-2 (at 3MIU/day X 28 days) was used in part of Study 3 , to enhance and maintain the level of adoptively- transferred autologous T cells. It was added afer a short course of IFN-α and immediately following the CTL infusion to enhance lymphocyte mitogenesis, lymphocyte cytotoxicity, and interferon-gamma production in an effort to maintain the antigen-specific T cells in vivo. Adoptive Transfer of Lymphocytes Following a Non-Myeloablative
Immunosuppressive Chemotherapy Regimen
Mouse studies demonstrated that induction of immunosuppression with chemotherapy prior to the administration of T cells was essential to enable the adoptive transfer of lymphocytes to mediate maximal tumor regression29'30. Based on these studies, clinical protocols were initiated to treat patients with the adoptive transfer of lymphocytes following the application of a non-myelo ablative chemotherapeutic regimen16'31. Non- myeloablative preparative regimens have been used to treat patients receiving allogeneic bone marrow transplants, and these regimens appear ideally suited to induce the transient immunosuppression that may enhance the effect of adoptively transferred T lymphocytes. A regimen of cyclophosphamide and fludarabine originally used in renal cell cancer patients receiving HLA-matched allotransplants32 has recently been used in metastatic melanoma patients16'31. The rationale for the incorporation of a lymphodepleting regimen prior to adoptive transfer of T cells was to possibly destroy regulatory cells, disrupt homeostatic T cell regulation ("making space"), or abrogate other normal tolerogenic mechanisms which potentially could enhance the engraftment of the transferred T cells. No treatment-related mortality was observed, and therefore non-myeloablative chemotherapy in combination with antitumor lymphocytes plus high-dose IL-2 should be safe. The non-myeloablation was induced with fludarabine (25 mg/m2) and cyclophosphamide (60 mg/kg) and was followed by a lymphocyte infusion and high dose IL-2 (720,000 IU/kg).
The combination of chemotherapeutic agents cyclophosphamide and fludarabine, while not completely myeloablative, has been demonstrated to be myelosuppressive, affecting neutrophils, lymphocytes, platelets, and red blood cells. Neutrophil levels, which fell to a nadir on day 10 following the initiation of chemotherapy, were recorded at 6/mm3 and recovered to above 500/mm3 on day 14 with support of filgrastim (G-CSF). Lymphocyte levels had a nadir of 6/mm3 and recovered to above 200/mm3 during the same time period. Patients usually displayed neutrophil counts above 500/mm3 and platelet counts above 20,000/mm3, 2-3 weeks after the initiation of the chemotherapy. No patients needed a stem cell transfusion to rescue marrow function, and therefore the overall treatment appears to be safe. However, CD4 counts did remain persistently low (the mean CD4 count at approximately day 200 was 156/mm3, with a range between 46/mm3 and 320/mm3), which is a known side effect of the immunosuppression induced with fludarabine. The development of opportunistic infections did occur (i.e., transient herpes zoster outbreaks), which resolved following the termination of treatment.
Cladribine (2-CdA, Leustatin®), a purine analog similar to fludarabine, is approved for use in the treatment of hairy cell leukemia and has been evaluated also in multiple sclerosis (MS) patients in an effort to reduce autoreactive T lymphocytes. In MS patients a specific decrease in T cell subsets was correlated with different doses of cladribine.33"35 It has a very favorable toxicity profile relative to other Iymphocytolytic drugs, while inducing a long-lasting lymphopenia. Lymphocyte levels plunge immediately after the first infusion of cladribine, reach a nadir at 5 months and begin to recover by 6 months when administered for 4 monthly courses33'34. Lymphopenia was observed in all patients, and infectious complications were not a serious problem. The drug may be given by the convenient subcutaneous route without causing local irritation and with pharmacokinetic and therapeutic results that appear entirely equivalent to the IV route36'37.
A recent study in humans identifying T regulatory cells (Treg) as immunosuppressive in a vaccine setting demonstrated an enhanced antitumor immune response following depletion of the Treg subset which displays a CD4+/CD25+hl phenotype along with CD28, CTLA-4 and GITR.38 This T cell subset can be selectively depleted (approximately 3 weeks) following a single dose of denileukin diftitix (DAB389IL-2, ONTAK®). Four days following the single dose, 37-77% depletion of Treg cells was noted in eight (8) renal cell carcinoma patients. Following the T cell depletion, an RNA-DC vaccine was administered and a 10-fold enhancement in the immune response to the vaccine antigens was noted in the patients receiving both DAB IL-2 and vaccine versus vaccine alone. Thus, the same dose (18μg/kg) six days prior to an adoptive transfer of the antigen-specific CTLs is a preferred dose for use in regimens in accordance with the invention.
Patient Selection
Subjects for the study are human patients diagnosed with metastatic melanoma who are HLA-A2 positive. A monoclonal antibody (BB7.2) is used to analyze PBMC samples by FACS analysis and further analysis is performed using the Olerup SSP™ PCR test (Geno Vision) to determine the HLA-A*0201 subtype.
Treatment Schema
Figure 2 depicts the treatment regimens described in more detail below. The patients are divided into three cohorts of subjects, with 10 in each cohort. In the Cohort A regimen, a control for the lymphodepleting regimen, CTLs plus cytokines are administered. In the Cohort B regimen, which is a preferred embodiment of the invention, patients receive cladribine (days 0-4) plus CTLs (day 36) plus cytokines. In the Cohort B regimen, which is another preferred embodiment of the invention, patients receive DAB IL-2 (day 30) plus CTLs (day 36) plus cytokines. The experimental regimens are described in more detail below.
CTLs Specific for Melanoma-Associated Antigens
Cytotoxic T lymphocyte therapy for malignant melanoma utilizes autologous in vitro-acύvsΛQά CD8+ T cells to destroy melanoma cells bearing melanoma-associated antigenic epitopes. CTLs are prepared from leukapheresis samples generated at the clinical site and transferred to a facility where CTLs are generated under GMP guidelines and returned for infusion.
On day 0, patients with metastatic melanoma undergo leukapheresis (approximately 10-15 liter run, depending on the patient's weight and condition). The obtained cells are shipped overnight at room temperature for processing at a GMP manufacturing facility. At appropriate times, leukapheresis is performed to obtain peripheral blood lymphocytes for analysis. These samples are used to monitor the presence of antigen-specific T cells and the presence of circulating melanoma tumor cells. The monitoring leukapheresis procedure processes approximately 5 liters of peripheral blood (or one-third the volume collected at the initial leukapheresis to obtain CD8 cells for subsequent infusion) at the clinical site and is shipped to the GMP manufacturing facility. A PCR assay is used to determine a decrease in the level of circulating melanoma cells following therapy. The detection of circulating melanoma cells can be measured by a sensitive, quantitative real time PCR assay39"41. The presence of antigen-specific T cells is evaluated with peptide-specific/HLA-A2 tetramers. The final CTL preparation for infusion contains 1 x 1010 autologous CTLs in 30OmL, contained in 76% (v/v) Lactated Ringer's Solution, 4% (v/v) of D5NS and 20% (v/v) of 25% human serum albumin (HSA). The CTL product is prepared in a sterile bag, suitable for infusion, held on ice and infused over 30 minutes, within forty-two (42) hours of preparation.
Prior to infusion, the CTLs are monitored for lytic activity (both peptide-loaded target cells and melanoma tumor cell lysis), viability, and purity by FACS analysis. The absence of both Drosophila cell DNA and insect viral RNA is checked with sensitive quantitative, real-time PCR assays. Sterility is determined by BacT/Alert® analysis, mycoplasma testing and Gram stain analysis.
Patients receive a CTL infusion of approximately 1010 autologous T cells, which have been stimulated and expanded ex vivo to become cytotoxic T lymphocytes capable of targeting to cells in vivo expressing the antigens to which the CTLs are directed. The cells are infused after the end of the IFN-alpha treatment and immediately precede the start of the IL-2 regimen.
Cytokines
Interleukin-2 (IL-2) is obtained from Chiron Corporation (Emeryville, CA). It is supplied as single-use vials containing 22 million IU (~1.3 mg) of IL-2 as a sterile, white to off-white lyophilized cake plus 50 mg mannitol and 0.18 mg sodium dodecyl sulfate, buffered with approximately 0.17 mg monobasic and 0.89 mg dibasic sodium phosphate to a pH of 7.5 (range 7.2 to 7.8). The powder is reconstituted with 1.2 mL of Sterile Water for Injection, USP, and the resultant concentration is 18 million IU/ml or 1.1 mg/mL. Intact vials are stored in the refrigerator (2° - 8°C) and protected from light. Reconstituted IL-2 is further diluted with 2.4 mL of D5% water. The final concentration (6MU/mL), once drawn into plastic syringes, is good for 14 days after reconstitution, if kept refrigerated at 2-8°C. The final dilution of IL-2 is self-administered on an outpatient basis subcutaneously at a dose of 3 million units (3 MIU), initiating immediately following the CTL infusion and daily until disease progression. INTRON- A® is obtained from Schering-Plough Corporation (Kenilworth, NJ). This IFN-alpha-2b product is supplied either as a lyophilized powder or a solution for injection in sterile 5ml vials. A vial contains approximately 3 x 10 U or lδ x lO U IFN. The lyophilized powder is reconstituted preferably just prior to use, as there is no bacteriostatic agent included; unused, lyophilized powder is stored in a refrigerator or freezer (-40C to -200C). When the agent is supplied as an injectable solution, it is stored in the refrigerator (2°-8°C or 36°-46°F). The final dilution of IFN- O is self-administered on an outpatient basis at 10MU/m2 on days 31-35 as a subcutaneous injection.
DΛB389IL-2
DAB389ΪL-2 (denileukin diftitox, ONTAK®) is a recombinant DNA-derived cytotoxic protein composed of the amino acid sequences for diphtheria toxin fragments A and B followed by the sequences for interleukin 2 that is expressed in E coli. It is a targeted drug, which binds to cells expressing CD25 (IL-2R) on their surface. It interacts with the high-affinity IL-2 receptor on the surface of malignant or normal T regulatory (Treg) cells to inhibit intracellular protein synthesis, rapidly leading to cell death. DAB389IL-2 is supplied in single use vials as a sterile, frozen solution intended for intravenous (IV) administration. Each 2 mL vial of ONTAK contains 300 meg of recombinant DAB-IL2 in a sterile solution of citric acid (2OnM), EDTA (0.05mM) and polysorbate 20 (<1%) in water for injection, USP. The solution has a pH of 6.9-7.2.
Intact vials are stored frozen or -100C but not refrozen. The material is brought to room temperature (25°C or 77°F) before preparing the dose by thawing in a refrigerator for not more than 24 hours or at room temperature for 1 -2 hours, and diluted with NS to a concentration of >15 mcg/mL. DAB389IL-2 is administered by intravenous injection, preferably by infusion over at least 15 minutes. Patients assigned to Cohort C receive a single subcutaneous injection of DAB IL-2 on day 30, which is one day before the initiation of IFN-Dand six days prior to the injection of the CTLs. The dose is 18 μg/kg.
Cladribine
Leustatin® (2-chloro-2'deoxy-DD-adenosine) is a synthetic antineoplastic agent. It is a purine nucleoside analog resistant to the action of adenosine deaminase, which results in preferential lymphocytoxicity. In cells with a high ratio of deoxycytidine kinase to deoxynucleotidase (e.g., lymphocytes and monocytes), cladribine is phosphorylated into the active triphosphate deoxynucleotide, 2-CdATP, which accumulates, causing a disruption of cellular metabolism, DNA damage, and subsequent cell death. The drug is a clear, colorless, sterile, preservative-free, isotonic solution supplied in single-use vials containing lOmg (lmg/mL) of cladribine. Each mL of cladribine contains lmg of the active ingredient and 9 mg (0.15mEq) of sodium chloride as an inactive ingredient. The solution has a pH range of 5.5 to 8.0. Phosphoric acid and/or dibasic sodium phosphate may be added to adjust the pH to 6.3 ± 0.3. Intact vials are stored refrigerated (2-8°C). The dose of cladribine for Cohort B is 0.12 mg/kg/day for a total of 5 days, fractionated into 3-4 sites where a maximum of 3.0 cc is administered at a single site. The total cladribine dose is 0.6 mg/kg. (Cladribine, administered subcutaneously on this same schedule (5 consecutive days) and at a dose of 0.14 mg/kg/day (total dose = 0.7 mg/kg) has been reported to be safe in hairy cell leukemia patients37. Additionally, the cladribine administered on this schedule and dose to sixty patients produced approximately 70% lymphodepletion after a single cycle with clinically safe myelosuppression. As a result of a change in standardization, Leustatin has been found to be 12% higher than reported in previous clinical studies compared with a cladribine stock synthesized in a laboratory. The dose that was reported to be 0. lmg/kg is now estimated to actually have been only 0.087 mg/kg33. Regarding the safety of subcutaneous administration with the Leustatin® formulation fractionated into 2-3 sites, a study in relapsing-remitting multiple sclerosis patients used 0.07 mg/kg/day X 5 days, monthly for 6 months for a total cumulative dose of 2.1 mg/kg3 . A total of 27 patients randomized to the treatment drug arm. Infections were limited to an episode of mild segmental herpes zoster that occurred in two cladribine- treated patients and in one patient receiving placebo. There were no individual cases of significant thrombocytopenia, anemia granulocytopenia or generalized marrow suppression.)
Optional Supportive Care
Patients may receive any of the following to abrogate IFN-alpha and IL-2 toxicity during treatment and thereafter: acetaminophen (650 mg PO q4h prn), diphenhydramine (50 mg IM or PO q4h prn), indomethacin (25 mg PO TID or 75 mg SR qd), prochlorperazine (10 mg IV or PO q4h prn nausea), Zantac® (150 mg BID gastritis). Other anti-inflammatory and antiemetic agents not specifically prescribed may be substituted for any of the above supportive drugs.
Although the invention has been described in detail above in reference to illustrative examples and preferred embodiments, the artisan will understand that the scope of the invention is defined not by the foregoing description, but by the appended claims as properly construed under principles of patent law.
References
1. Hryniuk W, Bush H. The importance of dose intensity in chemotherapy of metastatic breast cancer. J Clin Oncol 4: 1162-1170, 1986.
2. Herzig, RH. Dose-intensive therapy for advanced melanoma. In: J.O. Armitage, K.H. Antman (eds.), High-Dose Cancer Therapy. Pharmacology, Hematopoietic, Stem Cells; Baltimore: Williams and Wilkins pp750-754, 1992.
3. Mitchell MS, Harel W, Kan-Mitchell J, et al. Active specific immunotherapy of melanoma with allogeneic cell lysates: rationale, results and possible mechanisms of action. In: J.-C. Bystryn, S. Ferrone and P. Livingston (eds.), Specific Immunotherapy of Cancer with Vaccines: Ann. N.Y. Acad. Sci. ppl53-166, 1993.
4. Quan WDY Jr, Mitchell MS. Principles of biologic therapy. In: CM. Haskell, ed. Cancer Treatment; Philadelphia: W.B. Saunders pp57-69, 1995.
5. Mitchell MS, Jakowatz J, Harel W, et al. Increased effectiveness of interferon-alfa 2b following active specific immunotherapy for melanoma. J CKn Oncol 12:402-411, 1994.
6. Van der Bruggen P, Traversari C, Chomez P5 et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 254: 1643- 1647, 1991. 7. Gaugler B, Van der Eynde B, Van der Bruggen P, et al. Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J Exp Med 179:921-930, 1994.
8. Kawakami Y, Eliyahu S, Sakaguchi K, et al. Identification of the immunodominant peptides of the MART-I human melanoma antigen recognized by the majority of HLA- A2 -restricted tumor infiltrating lymphocytes. J Exp Med 180:347-352, 1994.
9. Brichard V, Van Pel A, Wolfel T, et al. The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA- A2 melanomas. J Exp Med 178:489-495, 1993.
10. Robbins PF, el-Gamil M, Kawakami Y, Stevens E, Yannelli JR, Rosenberg SA. Recognition of tyrosinase by tumor-infiltrating lymphocytes from a patient responding to immunotherapy. Cancer Res 54:3124-3126, 1994.
11. Bakker AB, Schreurs MW, de Boer AJ, et al. Melanocyte lineage-specific antigen gplOO is recognized by melanoma-derived tumor-infiltrating lymphocytes. J Exp Med 179:1005-1009, 1994.
12. Wolfel T, Van Pel A, Brichard V, et al. Two tyrosinase nonapeptides recognized on HLA- A2 melanoma by autologous cytolytic T lymphocytes. Eur J Immnol 24:759-764, 1994.
13. Visseren MJW, Van Elsas A, Van der Voort EIH, et al. CTL specific for the tyrosinase autoantigen can be induced from healthy donor blood to lyse melanoma cells. J Immunol 154:3991-3998, 1995.
14. Rubin JT, Lotze MT. Adoptive cellular immunotherapy of cancer. In: M.S. Mitchell (ed.), Biological Approaches to Cancer Treatment. Biomodulation; New York: McGraw-Hill pp379-410, 1993. 15. Yee, C, Thompson, JA, Byrd, D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones from the treatment of patients with metastatic melanoma: In vivo persistence, migration and antitumor effect of transferred T cells. PNAS 99:16168-16173,2002.
16. Dudley ME, Wunderlich, JR, Robbins PF, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science 298:850-854, 2002.
17. Oelke M, Maus MV, Didiano, D et al. Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells. Nat Med 9:619-624. 2003.
18. Leturcq DL, Richards JM, Jackson MR5 Peterson PA and Moriarty AM. Ex Vivo Generation of Potent Cytotoxic T Lymphocytes for the Treatment of Cancer: A Novel Antigen Presentation System. Society of Biological Therapy 17th Annual Meeting; Abstract #40,2002.
19. Jackson MR, Song ES, Yang Y, et al. Empty and peptide-containing conformers of class I major histocompatibility complex molecules expressed mDrosophila melanogaster cells. Proc Natl Acad Sci. USA 89:12117-12121, 1992.
20. Cai Z, Brunmark A, Luxembourg AT, et al. Probing the activation requirements for naive CD8+ T cells with Drosophila cell transfectants as antigen presenting cells. Immunol Rev 165:249-265, 1998.
21. Yee, C, Savage, P.A., Lee, P.P et al. Isolation of high avidity melanoma- reactive CTL from heterogeneous populations using peptϊde-MHC tetramer. J Immunol 162:227-2234, 1999.
22. Mitchell MS, Darrah D, Yeung D et al. Phase I trial of adoptive immunotherapy with cytolytic T lymphocytes immunized against a tyrosinase epitope. J Clin Oncol 20:1075-1086. 2002. 23. Richards JM, Moriarty AM, Leturcq DL, Jackson MR and Peterson PA. Treatment of advanced malignant melanoma with ex vzvo-generated autologous T cells with specificity for melanoma-associated target antigens. ASCO Annual Meeting. San Francisco, CA, May 2001.
24. Richards JM, Moriarty AM, Leturcq DL et al. Treatment of advanced malignant melanoma with autolgous, ex vjvø-generated T cells with specificity for melanoma-associated target antigens. ASCO Annual Meeting. Chicago, IL, June 2003.
25. Dorval, T, Palangies, T, Jouve M et al. Clinical phase II trial of recombinant DNA interferon (interferon alpha 2b) in patients with metastatic malignant melanoma. Cancer_58:215-218, 1986.
26. Sertoli MR, Bernengo MG, Ardizzoni A, et al. Phase II trial of recombinant alpha-2b interferon in the treatment of metastatic skin melanoma. Oncology 46:96-98, 1989.
27. Robincon WA, Mughal TI, Thormas MR, et al. Treatment of metastatic mealignant melanoma with recombinant interferon alpha 2. Immunobiologv 172:275-282, 1986.
28. Rosenberg, SA, Yang JC, Topalxan SL, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin-2. JAMA 271:907-9113, 1994.
29. North, RJ. Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on the elimination of tumor induced suppressor cells. J Exp Med 155:1063-1074.
30. Mills, CD., and North, RJ. : Expression of passively transferred immunity against an established tumor depends on generation of cytolytic T cells in recipient. J Exp Med 157:1448-1460, 1983.
31. Dudley, M., J. Wunderlich, J. C. Yang, et al. A Phase I study of non- myeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes inpatients with metastatic melanoma. J Immunother 25:243-251, 2002. 32. Childs. ., Chernoff, A., Contentin, N. et al. Regression of metastatic renal- cell carcinoma after nonmyeloablative allogeneic peripheral -blood stem-cell transplantation. NEJM 343:750-758, 2000.
33. Beυtler, E. et al. Marrow suppression produced by repeated doses of cladribine. Acta Haematol 91:10-15. 1994.
34. Beutler, E. et al. The treatment of chronic progressive multiple sclerosis with cladribine. Proc Natl Acad Sci. U S A 93 : 1716- 1720, 1996.
35. Rice, GPA et al. Cladribine and progressive MS: Clinical and MRI outcomes of a multicenter controlled trial. Neurology 54:1145-1155, 2000.
36. Romine, JS, Sipe, JC, Koziol, JA et al. A double-blind, placebo-controlled, reandomized trial of cladribine in relapsing-remitting multiple sclerosis. Proc Assoc Amer Physicians 111:35-44, 1999.
37. von Rohr, A, Schmitz, S-FH, Tichelli, A et al. Treatment of hairy cell leukemia with cladribine (2-chlorodeoxyadenosine) by subcutaneous bolus injection: a phase II study. Annals of Oncology 13:1641-1649, 2002.
38. View, J, Su, Z, "and Dannull, J. Enhancement of antitumor immunity following depletion of CD4+CD25+ regulatory T cells. Annual Meeting Proceedings ASCO Abst 2506. 2004.
39. Keilholz, U., Goldin-Lang, P, Bechrakis, NE et al. Quantitative detection of circulating tumor cells in cutaneous and ocular melanoma and quality assessment by realtime reverse transcriptase-polymerase chain reaction. Clinical Cancer Research 10: 1605- 1612, 2004.
40. Hoon, DSB, Bostick, P, Kuo, C et al. Molecular markers in blood as surrogate prognostic indicators of melanoma recurrence. Cancer Research 60:2253-2257, 2000.
41. Goydos, JS and Reintgen, DS. A molecular technique useful in the detection of occult metastases in patients with melanoma. In: BJ. Nickoloff (ed.) Methods in Molecular Medicine, VoI 61: Melanoma: Methods and Protocols; Totowa, NJ.Ηumana Press pp.301 -320.

Claims

WHAT IS CLAIMED IS:
1. A method for treating a subject in need of treatment for cancer, comprising:
(a) obtaining naϊve CD8+ T cells from the subject;
(b) contacting the naive CD8+ T cells with xenogenic antigen presenting cells loaded with one or more peptide antigens, thereby generating activated CTLs that target cells expressing said one or more peptide antigens;
(c) administering to the subject the activated CTLs;
(d) administering to the subject at least two cytokines that effect CTL persistence; and
(e) administering to the subject a lymphodepleting agent selected from the group consisting of cladribine and denileukin diftitox.
2. A method as defined in claim 1, wherein said at said at least two cytokines comprise interferon-α-2b and interleukin-2.
3. A method as defined in claim 2, wherein said one or more peptide antigens comprise an amino acid sequence derived from a protein selected from gplOO, tyrosinase, and MART-I.
4. A method as defined in claim 1, wherein said one or more peptide antigens consists of peptide antigens derived from human gplOO, tyrosinase, and MART-I proteins.
5. A method as defined in claim 1, wherein each of said one or more peptide antigens is selected from the group consisting of YMNGTMSQV (SEQ ID NO:1), YMDGTMSQV (SEQ ID NO:2), AAGIGILTV (SEQ ID NO:3), ITDQVPFSV (SEQ ID NO:4), YLEPGPVTA (SEQ ID NO: 5), and KTWGQYWQV (SEQ ID NO:6).
6. A method as defined in claim 5, wherein said cancer is a melanoma.
7. A method as defined in claim 1, wherein the administration of the lymphodepleting agent begins before the administration of the activated CTLs.
PCT/US2007/004841 2006-03-01 2007-02-23 CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES WO2007103009A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
AU2007222080A AU2007222080B2 (en) 2006-03-01 2007-02-23 Cancer treatment combining lymphodepleting agent with CTLs and cytokines
MX2008011302A MX2008011302A (en) 2006-03-01 2007-02-23 CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES.
US12/281,197 US7993638B2 (en) 2006-03-01 2007-02-23 Cancer treatment combining lymphodepleting agent with CTLs and cytokines
EP07751592.2A EP1996232B1 (en) 2006-03-01 2007-02-23 CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES
JP2008557314A JP2009531303A (en) 2006-03-01 2007-02-23 Cancer treatment combining lymphocyte depleting agents with CTLs and cytokines
CA2643337A CA2643337C (en) 2006-03-01 2007-02-23 Cancer treatment combining lymphodepleting agent with ctls and cytokines
BRPI0708446-3A BRPI0708446A2 (en) 2006-03-01 2007-02-23 cancer treatment by combining lymph node depletion agent with ctls and cytokines
ES07751592.2T ES2579762T3 (en) 2006-03-01 2007-02-23 Cancer treatment by combining lympho - reducing agent with CLTs and cytokines
IL193710A IL193710A (en) 2006-03-01 2008-08-26 Combinations of activated cytotoxic t lymphocytes, cytokines and cladribine for treating cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US77851606P 2006-03-01 2006-03-01
US60/778,516 2006-03-01

Publications (2)

Publication Number Publication Date
WO2007103009A2 true WO2007103009A2 (en) 2007-09-13
WO2007103009A3 WO2007103009A3 (en) 2008-10-09

Family

ID=38475344

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/004841 WO2007103009A2 (en) 2006-03-01 2007-02-23 CANCER TREATMENT COMBINING LYMPHODEPLETING AGENT WITH CTLs AND CYTOKINES

Country Status (14)

Country Link
US (1) US7993638B2 (en)
EP (1) EP1996232B1 (en)
JP (1) JP2009531303A (en)
KR (1) KR20080098066A (en)
CN (1) CN101437542A (en)
AU (1) AU2007222080B2 (en)
BR (1) BRPI0708446A2 (en)
CA (1) CA2643337C (en)
ES (1) ES2579762T3 (en)
IL (1) IL193710A (en)
MX (1) MX2008011302A (en)
RU (1) RU2447900C2 (en)
WO (1) WO2007103009A2 (en)
ZA (1) ZA200808334B (en)

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8075895B2 (en) 2009-09-22 2011-12-13 Janssen Pharmaceutica N.V. Identification of antigenic peptides from multiple myeloma cells
WO2013163171A1 (en) 2012-04-24 2013-10-31 Kaufman Dan S Method for developing natural killer cells from stem cells
WO2015164594A1 (en) 2014-04-23 2015-10-29 Board Of Regents, The University Of Texas System Chimeric antigen receptors (car) for use in therapy and methods for making the same
WO2015140172A3 (en) * 2014-03-17 2015-12-10 Bundesrepublik Deutschland Letztvertreten Durch Das Robert Koch-Institut Vertreten Durch Seinen Präsidenten A medicament for use in a method of inducing or extending a cellular cytotoxic immune response
WO2016073755A2 (en) 2014-11-05 2016-05-12 Board Of Regents, The University Of Texas System Gene modified immune effector cells and engineered cells for expansion of immune effector cells
WO2016073629A1 (en) 2014-11-05 2016-05-12 Board Of Regents, The University Of Texas System Chimeric antigen receptors (car) to selectively target protein complexes
WO2017075147A1 (en) 2015-10-27 2017-05-04 Board Of Regents, The University Of Texas System Chimeric antigen receptor molecules and uses thereof
WO2017214333A1 (en) 2016-06-08 2017-12-14 Intrexon Corporation Cd33 specific chimeric antigen receptors
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018132494A1 (en) 2017-01-10 2018-07-19 Intrexon Corporation Modulating expression of polypeptides via new gene switch expression systems
EP3656864A1 (en) 2014-02-14 2020-05-27 Board of Regents, The University of Texas System Chimeric antigen receptors and methods of making
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
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
WO2020200481A1 (en) * 2019-04-05 2020-10-08 Biontech Rna Pharmaceuticals Gmbh Treatment involving interleukin-2 (il2) and interferon (ifn)
EP3783098A1 (en) 2013-05-14 2021-02-24 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (car) t-cells
WO2021163191A1 (en) 2020-02-10 2021-08-19 Board Of Regents, The University Of Texas System Methods for rapid cloning and expression of hla class i cells
US11118168B2 (en) 2017-06-07 2021-09-14 Precigen, Inc. Expression of novel cell tags
WO2021240240A1 (en) 2020-05-27 2021-12-02 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
US20210369828A1 (en) * 2020-05-28 2021-12-02 Case Western Reserve University Plant virus based cancer antigen vaccine
KR20220095228A (en) 2019-11-06 2022-07-06 베이롤 칼리지 오브 메드신 Methods of Producing Cytotoxic Effector Memory T Cells for the Treatment of T Cells in Cancer
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
RU2689558C1 (en) * 2013-11-22 2019-05-28 Селлектис Method of constructing allogenic and drug resistant t-cells for immunotherapy
JP2017507950A (en) 2014-02-27 2017-03-23 リセラ・コーポレイションLycera Corporation Adoptive cell therapy using retinoic acid receptor-related orphan receptor gamma agonists and related therapeutic methods
US20170044500A1 (en) 2014-04-24 2017-02-16 Board Of Regents, The University Of Texas System Application of induced pluripotent stem cells to generate adoptive cell therapy products
US9896441B2 (en) 2014-05-05 2018-02-20 Lycera Corporation Tetrahydroquinoline sulfonamide and related compounds for use as agonists of RORγ and the treatment of disease
US10189777B2 (en) 2014-05-05 2019-01-29 Lycera Corporation Benzenesulfonamido and related compounds for use as agonists of RORγ and the treatment of disease
JP2018513123A (en) 2015-03-12 2018-05-24 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Methods for treating cancer using ROR gamma inhibitors
CA2982847A1 (en) 2015-05-05 2016-11-10 Lycera Corporation Dihydro-2h-benzo[b][1,4]oxazine sulfonamide and related compounds for use as agonists of ror.gamma. and the treatment of disease
KR20180025894A (en) 2015-06-11 2018-03-09 라이세라 코퍼레이션 Benzo [B] [1,4] oxazinesulfonamides and related compounds for use as agonists of RORY and the treatment of diseases
WO2017087768A1 (en) * 2015-11-18 2017-05-26 Duke University Targeted delivery of interleukin-10 to tumor infiltrating lymphocytes
CN105505873A (en) * 2016-02-29 2016-04-20 时宏珍 Preparation method of HLA-A0201 restriction anti-Her2-neu antigen-specific CTL (cytotoxic lymphocyte)
DE102018108612A1 (en) 2018-03-21 2019-09-26 Immatics US, Inc. METHOD FOR INCREASING PERSISTENCE OF ADOPTIVELY INFUNDED T CELLS
SG11202101888YA (en) 2018-09-04 2021-03-30 Treos Bio Ltd Peptide vaccines
WO2021219990A1 (en) * 2020-04-28 2021-11-04 Achilles Therapeutics Uk Limited T cell therapy
WO2023281097A1 (en) 2021-07-09 2023-01-12 Immunic Ag Methods for treating cancer

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US4844893A (en) 1986-10-07 1989-07-04 Scripps Clinic And Research Foundation EX vivo effector cell activation for target cell killing
US6225042B1 (en) 1995-03-08 2001-05-01 The Scripps Research Institute Antigen presenting system and methods for activation of T-cells
US6355479B1 (en) 1996-05-23 2002-03-12 The Scripps Research Institute MHC class II antigen-presenting systems and methods for activating CD4+ T cells

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6660276B1 (en) * 1994-02-16 2003-12-09 The University Of Virginia Patent Foundation Peptides recognized by melanoma-specific cytotoxic lymphocytes, and uses therefor
US6515016B2 (en) * 1996-12-02 2003-02-04 Angiotech Pharmaceuticals, Inc. Composition and methods of paclitaxel for treating psoriasis
EP1276896B1 (en) 2000-04-28 2010-07-21 Mannkind Corporation Epitope synchronization in antigen presenting cells
US20040071671A1 (en) * 2001-02-20 2004-04-15 Leturcq Didier J. Cell therapy method for the treatment of tumors
WO2003057823A2 (en) * 2001-11-07 2003-07-17 Mannkind Corporation Epitope synchronization in antigen presenting cells
US20050043233A1 (en) * 2003-04-29 2005-02-24 Boehringer Ingelheim International Gmbh Combinations for the treatment of diseases involving cell proliferation, migration or apoptosis of myeloma cells or angiogenesis
JP2005139118A (en) * 2003-11-07 2005-06-02 Ortho Mcneil Pharmaceut Inc Cell therapy method for treating tumor

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4690915A (en) 1985-08-08 1987-09-01 The United States Of America As Represented By The Department Of Health And Human Services Adoptive immunotherapy as a treatment modality in humans
US4844893A (en) 1986-10-07 1989-07-04 Scripps Clinic And Research Foundation EX vivo effector cell activation for target cell killing
US6225042B1 (en) 1995-03-08 2001-05-01 The Scripps Research Institute Antigen presenting system and methods for activation of T-cells
US6355479B1 (en) 1996-05-23 2002-03-12 The Scripps Research Institute MHC class II antigen-presenting systems and methods for activating CD4+ T cells

Non-Patent Citations (43)

* Cited by examiner, † Cited by third party
Title
BAKKER AB; SCHREURS MW; DE BOER AJ ET AL.: "Melanocyte lineage-specific antigen gp100 is recognized by melanoma-derived tumor-infiltrating lymphocytes", J EXP MED, vol. 179, 1994, pages 1005 - 1009
BEUTLER, E. ET AL.: "Marrow suppression produced by repeated doses of cladribine", ACTA HAEMATOL, vol. 91, 1994, pages 10 - 15, XP009183282
BEUTLER, E. ET AL.: "The treatment of chronic progressive multiple sclerosis with cladribine", PROC NATL ACAD SCI, USA, vol. 93, 1996, pages 1716 - 1720, XP002339785, DOI: doi:10.1073/pnas.93.4.1716
BRICHARD V; VAN PEL A; WOLFEL T ET AL.: "The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas", J EXP MED, vol. 178, 1993, pages 489 - 495
CAI Z; BRUNMARK A; LUXEMBOURG AT ET AL.: "Probing the activation requirements for naive CD8+ T cells with Drosophila cell transfectants as antigen presenting cells", IMMUNOL REV, vol. 165, 1998, pages 249 - 265, XP000960281
CHILDS, .; CHERNOFF, A.; CONTENTIN, N. ET AL.: "Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation", NEJM, vol. 343, 2000, pages 750 - 758, XP009049067, DOI: doi:10.1056/NEJM200009143431101
DORVAL, T; PALANGIES, T; JOUVE M ET AL.: "Clinical phase II trial of recombinant DNA interferon (interferon alpha 2b) in patients with metastatic malignant melanoma", CANCER, vol. 58, 1986, pages 215 - 218
DUDLEY ME; WUNDERLICH, JR; ROBBINS PF ET AL.: "Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes", SCIENCE, vol. 298, 2002, pages 850 - 854, XP009113075, DOI: doi:10.1126/science.1076514
DUDLEY, M.; J. WUNDERLICH; J. C. YANG ET AL.: "A Phase I study of non-myeloablative chemotherapy and adoptive transfer of autologous tumor antigen-specific T lymphocytes inpatients with metastatic melanoma", J IMMUNOTHER, vol. 25, 2002, pages 243 - 251
GAUGLER B; VAN DER EYNDE B; VAN DER BRUGGEN P ET AL.: "Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes", J EXP MED, vol. 179, 1994, pages 921 - 930
GOYDOS, JS; REINTGEN, DS.: "Methods in Molecular Medicine, Vol 61: Melanoma: Methods and Protocols", vol. 61, HUMANA PRESS, article "A molecular technique useful in the detection of occult metastases in patients with melanoma", pages: 301 - 320
HERZIG, RH.: "High-Dose Cancer Therapy. Pharmacology, Hematopoietins, Stem Cells", 1992, WILLIAMS AND WILKINS, article "Dose-intensive therapy for advanced melanoma", pages: 750 - 754
HOON, DSB; BOSTICK, P; KUO, C ET AL.: "Molecular markers in blood as surrogate prognostic indicators of melanoma recurrence", CANCER RESEARCH, vol. 60, 2000, pages 2253 - 2257
HRYNIUK W; BUSH H.: "The importance of dose intensity in chemotherapy of metastatic breast cancer", J CLIN ONCOL, vol. 4, 1986, pages 1162 - 1170
JACKSON MR; SONG ES; YANG Y ET AL.: "Empty and peptide-containing conformers of class I major histocompatibility complex molecules expressed in Drosophila melanogaster cells", PROC NATL ACAD SCI, USA, vol. 89, 1992, pages 12117 - 12121, XP000941368, DOI: doi:10.1073/pnas.89.24.12117
KAWAKAMI Y; ELIYAHU S; SAKAGUCHI K ET AL.: "Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes", J EXP MED, vol. 180, 1994, pages 347 - 352, XP000565650, DOI: doi:10.1084/jem.180.1.347
KEILHOLZ, U.; GOLDIN-LANG, P; BECHRAKIS, NE ET AL.: "Quantitative detection of circulating tumor cells in cutaneous and ocular melanoma and quality assessment by real-time reverse transcriptase-polymerase chain reaction", CLINICAL CANCER RESEARCH, vol. 10, 2004, pages 1605 - 1612, XP002534865, DOI: doi:10.1158/1078-0432.CCR-0610-3
LETURCQ DL; RICHARDS JM; JACKSON MR; PETERSON PA; MORIARTY AM: "Ex Vivo Generation of Potent Cytotoxic T Lymphocytes for the Treatment of Cancer: A Novel Antigen Presentation System", SOCIETY OF BIOLOGICAL THERAPY 17TH ANNUAL MEETING, 2002
MILLS, C.D.; NORTH, R.J.: "Expression of passively transferred immunity against an established tumor depends on generation of cytolytic T cells in recipient", J EXP MED, vol. 157, 1983, pages 1448 - 1460
MITCHELL MS; DARRAH D; YEUNG D ET AL.: "Phase I trial of adoptive immunotherapy with cytolytic T lymphocytes immunized against a tyrosinase epitope", J CLIN ONCOL, vol. 20, 2002, pages 1075 - 1086, XP002354241, DOI: doi:10.1200/JCO.20.4.1075
MITCHELL MS; HAREL W; KAN-MITCHELL J ET AL.: "Specific Immunotherapy of Cancer with Vaccines", 1993, ANN. N.Y. ACAD. SCI., article "Active specific immunotherapy of melanoma with allogeneic cell lysates: rationale, results and possible mechanisms of action", pages: L53 - L66
MITCHELL MS; JAKOWATZ J; HAREL W ET AL.: "Increased effectiveness of interferon-alfa 2b following active specific immunotherapy for melanoma", J CLIN ONCOL, vol. 12, 1994, pages 402 - 411
NORTH, RJ.: "Cyclophosphamide-facilitated adoptive immunotherapy of an established tumor depends on the elimination of tumor induced suppressor cells", J EXP MED, vol. 155, pages 1063 - 1074
OELKE M; MAUS MV; DIDIANO, D ET AL.: "Ex vivo induction and expansion of antigen-specific cytotoxic T cells by HLA-Ig-coated artificial antigen-presenting cells", NAT MED, vol. 9, 2003, pages 619 - 624, XP002352663, DOI: doi:10.1038/nm869
QUAN WDY JR; MITCHELL MS.: "Cancer Treatment", 1995, W.B. SAUNDERS, article "Principles of biologic therapy", pages: 57 - 69
RICE, GPA ET AL.: "Cladribine and progressive MS: Clinical and MRI outcomes of a multicenter controlled trial", NEUROLOGY, vol. 54, 2000, pages 1145 - 1155, XP008047071
RICHARDS JM; MORIARTY AM; LETURCQ DL ET AL.: "Treatment of advanced malignant melanoma with autolgous, ex vivo-generated T cells with specificity for melanoma-associated target antigens", ASCO ANNUAL MEETING, CHICAGO, IL, June 2003 (2003-06-01)
RICHARDS JM; MORIARTY AM; LETURCQ DL; JACKSON MR; PETERSON PA.: "Treatment of advanced malignant melanoma with ex vivo-generated autologous T cells with specificity for melanoma-associated target antigens", ASCO ANNUAL MEETING, SAN FRANCISCO, CA, May 2001 (2001-05-01)
ROBBINS PF; EL-GAMIL M; KAWAKAMI Y; STEVENS E; YANNELLI JR; ROSENBERG SA.: "Recognition of tyrosinase by tumor-infiltrating lymphocytes from a patient responding to immunotherapy", CANCER RES, vol. 54, 1994, pages 3124 - 3126, XP002003915, DOI: doi:10.1016/S0065-230X(08)60399-1
ROBINCON WA; MUGHAL TI; THORMAS MR ET AL.: "Treatment of metastatic mealignant melanoma with recombinant interferon alpha 2", IMMUNOBIOLOGY, vol. 172, 1986, pages 275 - 282
ROHR, A; SCHMITZ, S-FH; TICHELLI, A ET AL.: "Annals of Oncology", vol. 13, 2002, article "Treatment of hairy cell leukemia with cladribine (2-chlorodeoxyadenosine) by subcutaneous bolus injection: a phase II study", pages: 1641 - 1649
ROMINE, JS; SIPE, JC; KOZIOL, JA ET AL.: "A double-blind, placebo-controlled, reandomized trial of cladribine in relapsing-remitting multiple sclerosis", PROC ASSOC AMER PHYSICIANS, vol. 111, 1999, pages 35 - 44
ROSENBERG, SA; YANG JC; TOPALIAN SL ET AL.: "Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin-2", JAMA, vol. 271, 1994, pages 907 - 9113
RUBIN JT; LOTZE MT.: "Biological Approaches to Cancer Treatment. Biomodulation", 1993, MCGRAW-HILL, article "Adoptive cellular immunotherapy of cancer", pages: 379 - 410
SCHNEIDER, J. EMBRYOL. MORPH., vol. 27, 1972, pages 353 - 365
See also references of EP1996232A4
SERTOLI MR; BERNENGO MG; ARDIZZONI A ET AL.: "Phase II trial of recombinant alpha-2b interferon in the treatment of metastatic skin melanoma", ONCOLOGY, vol. 46, 1989, pages 96 - 98
VAN DER BRUGGEN P; TRAVERSARI C; CHOMEZ P ET AL.: "A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma", SCIENCE, vol. 254, 1991, pages 1643 - 1647, XP002628352, DOI: doi:10.1126/science.1840703
VIEW, J; SU, Z; DANNULL, J.: "Enhancement of antitumor immunity following depletion of CD4+CD25+ regulatory T cells", ANNUAL MEETING PROCEEDINGS, 2004
VISSEREN MJW; VAN ELSAS A; VAN DER VOORT EIH ET AL.: "CTL specific for the tyrosinase autoantigen can be induced from healthy donor blood to lyse melanoma cells", J IMMUNOL, vol. 154, 1995, pages 3991 - 3998, XP002052117
WOLFEL T; VAN PEL A; BRICHARD V ET AL.: "Two tyrosinase nonapeptides recognized on HLA-A2 melanoma by autologous cytolytic T lymphocytes", EUR J IMMNOL, vol. 24, 1994, pages 759 - 764
YEE, C.; SAVAGE, P.A.; LEE, P.P ET AL.: "Isolation of high avidity melanoma-reactive CTL from heterogeneous populations using peptide-MHC tetramer", J IMMUNOL, vol. 162, 1999, pages 227 - 2234
YEE, C; THOMPSON, JA; BYRD, D ET AL.: "Adoptive T cell therapy using antigen-specific CD8+ T cell clones from the treatment of patients with metastatic melanoma: In vivo persistence, migration and antitumor effect of transferred T cells", PNAS, vol. 99, 2002, pages 16168 - 16173, XP002505178, DOI: doi:10.1073/PNAS.242600099

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8075895B2 (en) 2009-09-22 2011-12-13 Janssen Pharmaceutica N.V. Identification of antigenic peptides from multiple myeloma cells
US8232101B2 (en) 2009-09-22 2012-07-31 Janssen Pharmaceutica N.V. Identification of antigenic peptides from multiple myeloma cells
US8323965B2 (en) 2009-09-22 2012-12-04 Janssen Pharmaceutica Nv Identification of antigenic peptides from multiple myeloma cells
US8323655B2 (en) 2009-09-22 2012-12-04 Janssen Pharmaceutica Nv Identification of antigenic peptides from multiple myeloma cells
WO2013163171A1 (en) 2012-04-24 2013-10-31 Kaufman Dan S Method for developing natural killer cells from stem cells
EP3567102A1 (en) 2012-04-24 2019-11-13 Dan S. Kaufman Method for developing natural killer cells from stem cells
EP3783098A1 (en) 2013-05-14 2021-02-24 Board Of Regents, The University Of Texas System Human application of engineered chimeric antigen receptor (car) t-cells
EP3974520A1 (en) 2013-05-14 2022-03-30 Board of Regents, The University of Texas System Human application of engineered chimeric antigen receptor (car) t-cells
EP3656864A1 (en) 2014-02-14 2020-05-27 Board of Regents, The University of Texas System Chimeric antigen receptors and methods of making
WO2015140172A3 (en) * 2014-03-17 2015-12-10 Bundesrepublik Deutschland Letztvertreten Durch Das Robert Koch-Institut Vertreten Durch Seinen Präsidenten A medicament for use in a method of inducing or extending a cellular cytotoxic immune response
US10849963B2 (en) 2014-03-17 2020-12-01 Bundesrepublik Deutschland letztvertreten durch das Robl Koch-Institut vertreten durch seinen Präsidenten Medicament for use in a method of inducing or extending a cellular cytotoxic immune response
US10688165B2 (en) 2014-03-17 2020-06-23 Bundesrepublik Deutschland letztvertreten durch das Robert-Koch-Institut vertreten durch seinen Präsidenten Medicament for use in a method of inducing or extending a cellular cytotoxic immune response
WO2015164594A1 (en) 2014-04-23 2015-10-29 Board Of Regents, The University Of Texas System Chimeric antigen receptors (car) for use in therapy and methods for making the same
WO2016073755A2 (en) 2014-11-05 2016-05-12 Board Of Regents, The University Of Texas System Gene modified immune effector cells and engineered cells for expansion of immune effector cells
US10570186B2 (en) 2014-11-05 2020-02-25 Board Of Regents, The University Of Texas System Chimeric antigen receptors (CAR) to selectively target protein complexes
WO2016073629A1 (en) 2014-11-05 2016-05-12 Board Of Regents, The University Of Texas System Chimeric antigen receptors (car) to selectively target protein complexes
WO2017075147A1 (en) 2015-10-27 2017-05-04 Board Of Regents, The University Of Texas System Chimeric antigen receptor molecules and uses thereof
WO2017214333A1 (en) 2016-06-08 2017-12-14 Intrexon Corporation Cd33 specific chimeric antigen receptors
WO2018023100A2 (en) 2016-07-29 2018-02-01 Juno Therapeutics, Inc. Anti-idiotypic antibodies and related methods
WO2018132494A1 (en) 2017-01-10 2018-07-19 Intrexon Corporation Modulating expression of polypeptides via new gene switch expression systems
US11118168B2 (en) 2017-06-07 2021-09-14 Precigen, Inc. Expression of novel cell tags
US11919937B2 (en) 2018-01-09 2024-03-05 Board Of Regents, The University Of Texas System T cell receptors for immunotherapy
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
WO2020200481A1 (en) * 2019-04-05 2020-10-08 Biontech Rna Pharmaceuticals Gmbh Treatment involving interleukin-2 (il2) and interferon (ifn)
WO2020201448A1 (en) * 2019-04-05 2020-10-08 Biontech Rna Pharmaceuticals Gmbh Treatment involving interleukin-2 (il2) and interferon (ifn)
CN113747913A (en) * 2019-04-05 2021-12-03 生物技术公司 Treatments involving interleukin-2 (IL2) and Interferon (IFN)
KR20220095228A (en) 2019-11-06 2022-07-06 베이롤 칼리지 오브 메드신 Methods of Producing Cytotoxic Effector Memory T Cells for the Treatment of T Cells in Cancer
WO2021163191A1 (en) 2020-02-10 2021-08-19 Board Of Regents, The University Of Texas System Methods for rapid cloning and expression of hla class i cells
WO2021240240A1 (en) 2020-05-27 2021-12-02 Antion Biosciences Sa Adapter molecules to re-direct car t cells to an antigen of interest
US20210369828A1 (en) * 2020-05-28 2021-12-02 Case Western Reserve University Plant virus based cancer antigen vaccine

Also Published As

Publication number Publication date
KR20080098066A (en) 2008-11-06
EP1996232B1 (en) 2016-04-06
IL193710A (en) 2015-04-30
BRPI0708446A2 (en) 2011-06-07
CN101437542A (en) 2009-05-20
CA2643337A1 (en) 2007-09-13
ZA200808334B (en) 2009-12-30
EP1996232A2 (en) 2008-12-03
AU2007222080A1 (en) 2007-09-13
ES2579762T3 (en) 2016-08-16
CA2643337C (en) 2016-01-12
RU2447900C2 (en) 2012-04-20
WO2007103009A3 (en) 2008-10-09
AU2007222080B2 (en) 2012-08-16
US7993638B2 (en) 2011-08-09
EP1996232A4 (en) 2010-08-04
JP2009531303A (en) 2009-09-03
MX2008011302A (en) 2008-11-04
IL193710A0 (en) 2011-08-01
US20090324539A1 (en) 2009-12-31
RU2008138880A (en) 2010-04-10

Similar Documents

Publication Publication Date Title
US7993638B2 (en) Cancer treatment combining lymphodepleting agent with CTLs and cytokines
Kimura et al. Clinical and immunologic evaluation of dendritic cell–based immunotherapy in combination with gemcitabine and/or S-1 in patients with advanced pancreatic carcinoma
US9222070B2 (en) Cell therapy method for the treatment of tumors
EP2215220B1 (en) Stimulation of anti-tumor immunity using dendritic cell/tumor cell fusions and anti-cd3/cd28
JP2011504101A5 (en)
JP2012097117A (en) Cell therapy method for treatment of tumor
JP2024019229A (en) Activated dendritic cell compositions and methods for immunotherapy treatment for subjects with advanced cancer
Yannelli et al. The large scale generation of dendritic cells for the immunization of patients with non-small cell lung cancer (NSCLC)
Cohen et al. Use of interleukin-7, interleukin-2, and interferon-γ to propagate CD4+ T cells in culture with maintained antigen specificity
US7867488B2 (en) Use of dendritic cells (DCs) expressing interleukin 12 (IL-12)
US11090332B2 (en) Antigen specific mRNA cellular cancer vaccines
JP2005139118A (en) Cell therapy method for treating tumor
WO1998023728A1 (en) Cellular adjuvant
JP2010235611A (en) Method of cell therapy for treatment of tumor
Ngo et al. Breast cancer treatment by transplantations of dendritic cells and cytokine-induced killer cells: An update on clinical trials
AU718873B2 (en) Cellular adjuvant

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 570659

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2643337

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 193710

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2007222080

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2008557314

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/a/2008/011302

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 3693/KOLNP/2008

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 1020087022674

Country of ref document: KR

ENP Entry into the national phase

Ref document number: 2007222080

Country of ref document: AU

Date of ref document: 20070223

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2007751592

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2008138880

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 200780016032.9

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 12281197

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0708446

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20080901