[go: up one dir, main page]

WO2017019767A1 - Inhibition de cxcl12 dans l'immunothérapie anticancéreuse - Google Patents

Inhibition de cxcl12 dans l'immunothérapie anticancéreuse Download PDF

Info

Publication number
WO2017019767A1
WO2017019767A1 PCT/US2016/044254 US2016044254W WO2017019767A1 WO 2017019767 A1 WO2017019767 A1 WO 2017019767A1 US 2016044254 W US2016044254 W US 2016044254W WO 2017019767 A1 WO2017019767 A1 WO 2017019767A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
cxcl12
antibody
tumor
immune response
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
PCT/US2016/044254
Other languages
English (en)
Inventor
Douglas Fearon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Myosotis LLC
Original Assignee
Myosotis LLC
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 Myosotis LLC filed Critical Myosotis LLC
Publication of WO2017019767A1 publication Critical patent/WO2017019767A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons

Definitions

  • the present invention is concerned with therapy of tumors.
  • the invention is concerned with reducing or preventing immune suppression and increasing T-cell recruitment and accumulation in the cancerous tumor microenvironment, in order to overcome the exclusion and death of CD3+ T-cells, and preferably CD3+ effector T-cells from the tumor and the suppression of anti-tumor T-cell activity.
  • Cancer is the second leading cause of death in the United States, exceeded only by heart disease.
  • surgery and radiotherapy may be curative if a cancer is found early, but current drug therapies for metastatic disease are mostly palliative and seldom offer a long-term cure.
  • new chemotherapies entering the market, the need continues for new drugs effective in monotherapy or in combination with existing agents as first line therapy, and as second and third line therapies in treatment of resistant tumors.
  • Cancer cells are by definition heterogeneous. For example, within a single tissue or cell type, multiple mutational 'mechanisms' may lead to the development of cancer. As such, heterogeneity frequently exists between cancer cells taken from tumors of the same type that have originated in different individuals and even between cancer cells from different regions of a tumor in a single individual. Frequently observed mutational 'mechanisms' associated with some cancers may differ between one tissue type and another (e.g., frequently observed mutational 'mechanisms' leading to colon cancer may differ from frequently observed 'mechanisms' leading to leukemias). It is therefore often difficult to predict whether a particular cancer will respond to a particular chemotherapeutic agent.
  • CXCL12 is a chemokine that localizes to human PDA.
  • CXCL12 antagonizing CXCL12, or its receptor, CXCR4
  • McCandless et al. increases T-cell trafficking across the blood-brain barrier and improves survival rates from West Nile virus disease
  • anti-CXCL12 therapy might be useful for the treatment of ovarian cancer, because CXCL12 inhibition leads to a reduction in FoxP3+ regulatory T-cells in ovarian tumors (Righi et al., Cancer Res. 2011 Aug 15; 71(16):5522-34).
  • the present invention addresses the continued need to improve and develop new cancer treatments.
  • the present invention relates to a method of inducing an immune response to an epithelial tumor characterized by bound CXCL12 comprising administering to a subject with the tumor one or more CXCL12-targeted immune response agents; wherein administration of the CXCL12-targeted immune response agent enhances or induces a tumor antigen- specific immune response against the epithelial tumor.
  • the CXCL12-targeted immune response agent is an anti- CXCL12 antibody.
  • the anti-CXCL12 antibody belongs to immunoglobulin isotypes that bind to Fc receptors that mediate phagocytosis by macrophages or that mediate antibody-dependent cell-mediated cytotoxicity by natural killer cells.
  • anti-CXCL12 antibody immunoglobulin isotypes include, but are not limited to: the IgGl and IgG3 immunoglobulin isotypes.
  • the anti-CXCL12 antibody belongs to the human IgGl or IgG3 immunoglobulin isotypes.
  • the anti-CXCL12 antibody triggers the antibody-dependent elimination of epithelial tumor cells.
  • an anti-CXCL12 antibody includes, but is not limited to an anti-SDF-1 antibody.
  • the method also comprises administration of an anti-CD47 antibody.
  • the anti-CD47 antibody disrupts the interaction between CD47 on cancer cells with SIRPa on phagocytic cells.
  • the anti-CD47 antibody enhances the antibody-dependent elimination of epithelial tumor cells.
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with an anti-CD47 antibody.
  • the CXCL12-targeted immune response agent is a bispecific antibody.
  • These bispecific antibodies include, but are not limited to: a bispecific T- cell engager (BiTE) antibody, a dual-affinity retargeting molecule (DART), a CrossMAb antibody, a DutaMabTM antibody, a DuoBody antibody; a Triomab, a TandAb, a bispecific NanoBody, a Tandem scFv, a diabody, a single chain diabody, a HSA body, a (scFv)2 HSA Antibody, an scFv-IgG antibody, a Dock and Lock bispecific antibody, a DVD-IgG antibody, a TBTI DVD-IgG, an IgG-fynomer, a Tetravalent bispecific tandem IgG antibody, a dual-targeting domain antibody, a chemically linked bispecific (Fab')2 molecule,
  • the CXCL12-targeted immune response agent is a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12.
  • the CAR T-cell causes apoptosis of the epithelial tumor cells.
  • the CXCL12-targeted immune response agent is a T- cell preloaded with bispecific antibody.
  • the T-cell preloaded with bispecific antibody is polyclonally- activated.
  • the anti-CXCL12 antibody or fragment thereof is fused to a heterologous molecule, substance, or agent.
  • the heterologous molecule, substance, or agent binds to a cytokine.
  • cytokine examples include, but are not limited to: IL-2, IL-6, IL-7, IL-10, IL-12, IL-15, IL-17, IL-18, IL-21, GM-CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL.
  • the cytokine is a pro-inflammatory cytokine that promotes inflammatory responses.
  • the anti-CXCL12 antibody or fragment thereof is fused to two molecules that bind to cytokines.
  • the two molecules may bind the same cytokine, while in other embodiments, the two molecules may be distinct and may bind different cytokines.
  • any cytokine or combination of cytokines capable of stimulating an immune response may bind to the heterologous molecule, substance, or agent, to be delivered to tumor cells characterized by CXCL12.
  • the anti-CXCL12 antibody or fragment thereof is fused to a radionuclide.
  • radionuclides include, but are not limited to 13 iodine, 9a yYttrium, 177 Lutetium, 188 Rhenium, 67 Copper, 211 Astatine, 213 Bismuth, 125 Iodine, or i n Indium.
  • the anti-CXCL12 antibody or fragment thereof is fused to a toxin.
  • toxins include, but are not limited to Pseudomonas exotoxin, staphylococcal enterotoxin A, ricin A-chain, or plant ribosome-inactivating proteins such as saporin. Additional examples of toxins include, but are not limited to, diphtheria toxin and pokeweed antiviral toxin. Other plant and bacterial toxins are contemplated for attachment to the anti-CXCL12 antibody or fragment thereof.
  • the anti-CXCL12 antibody or fragment thereof is fused to a pro-apoptotic protein.
  • pro-apoptotic proteins include, but are not limited to caspase-3, FOXP3, or death ligand TNF-related apoptosis-inducing ligand (TRAIL).
  • pro-apoptotic proteins include, but are not limited to: bad, bax, bcl-2, bcl-w, BID, BIM, Caspase 8, CD40, CD40 Ligand, cIAP-2, Cytochrome-C, DR6, Fas, Fas Ligand, HSP27, HSP60, HSP70, HTRA, IGF-1, IGF-2, IGFBP-1, IGFBP-2, IGFBP-3, IGFBP-4, IGFBP-5, IGFBP-6, IGF-lsR, livin, p21, p27, p53, SMAC, Survivin, sTNFRI, sTNFRII, TNF alpha, TNF beta, TRAIL Rl, TRAIL R2, TRAIL R3, TRAIL R4, and XIAP.
  • the anti-CXCL12 antibody or fragment thereof is fused to a chemotherapeutic agent/anti-cancer compound.
  • chemotherapeutic agents/anti- cancer compounds are provided herein.
  • the anti-CXCL12 targeting agent is used to deliver a second molecule to the tumor cells that inhibits the growth of or eliminates the tumor cells.
  • the second molecule can be a pro-apoptotic protein that triggers apoptosis, a radionuclide or toxin that kills tumor cells, or any heterologous molecule, substance or agent that binds to a chemokine or cytokine that stimulates the immune system to identify and eliminate the tumor cells.
  • the anti-CXCL12 targeting agent is fused/covalently bound to a pro-apoptotic protein, to a radionuclide or toxin, or to a chemotherapeutic agent. In other embodiments, the anti-CXCL12 targeting agent forms a non-covalent complex with a pro-apoptotic protein, a radionuclide or toxin, a chemotherapeutic agent, or a cytokine or chemokine.
  • the anti-CXCL12 targeting agent is a bispecific antibody or fragment thereof that binds to CXCL12 and also binds to a cytokine or chemokine that activates the immune system to inhibit the growth of or eliminate the tumor cells.
  • Cytokines include, but are not limited to: IL-2, IL-6, IL-7, IL-10, IL-12, IL-15, IL-17, IL-18, IL-21, IL-27, GM-CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL.
  • the anti-CXCL12 targeting agent and the second molecule are administered in a therapeutically effective amount to a patient in need thereof.
  • the second molecule can be a pro-apoptotic protein, a radionuclide or toxin, a chemotherapeutic or any heterologous molecule, substance or agent capable of binding to a chemokine or cytokine to activate the immune system to eliminate the tumor cell.
  • the anti-CXCL12 targeting agent may be an anti-CXCL12 antibody conjugated to a chemotherapeutic agent to form an antibody-drug conjugate (ADC) for delivery to the tumor cells
  • ADC antibody-drug conjugate
  • the anti-CXCL12 targeting agent may be an anti-CXCL12 antibody conjugated to a chemotherapeutic agent to form an antibody-drug conjugate (ADC) for delivery to the tumor cells
  • ADC antibody-drug conjugate
  • the anti-CXCL12 targeting agent may be an anti-CXCL12 antibody conjugated to a chemotherapeutic agent to form an antibody-drug conjugate (ADC) for delivery to the tumor cells
  • ADC antibody-drug conjugate
  • the method inhibits cancer cell growth. In an even further preferred embodiment, the method eliminates cancer cells. In an even further preferred embodiment, the method reduces tumor mass. In a further preferred embodiment, the tumor is resistant to immunotherapy.
  • the tumor is an adenocarcinoma, sarcoma, skin cancer, melanoma, bladder cancer, brain cancer, breast cancer, uterine cancer, ovarian cancer, prostate cancer, lung cancer, colorectal cancer, cervical cancer, liver cancer, head and neck cancer, esophageal cancer, pancreas cancer, pancreatic ductal adenocarcinoma (PDA), renal cancer, stomach cancer, multiple myeloma or cerebral cancer.
  • the method also comprises administering a checkpoint antagonist.
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with a checkpoint antagonist, including for example, an antibody directed to a checkpoint protein.
  • the method also comprises administering a PD-1 signaling inhibitor.
  • the PD-1 signaling inhibitor is a PD-1 antagonist.
  • the PD-1 antagonist is an anti-PD-1 antibody.
  • the PD-1 signaling inhibitor is a PD-L1 antagonist.
  • the PD-L1 antagonist is an anti-PD-Ll antibody.
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with a PD-1 signaling inhibitor, and preferably with a PD-1 antagonist, including for example, an anti-PD-1 antibody, or a PD-L1 antagonist, including for example, an anti-PD-Ll antibody.
  • a PD-1 signaling inhibitor and preferably with a PD-1 antagonist, including for example, an anti-PD-1 antibody, or a PD-L1 antagonist, including for example, an anti-PD-Ll antibody.
  • the method also comprises administering a CTLA-4 antagonist.
  • the CTLA-4 antagonist is an anti-CTLA-4 antibody.
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with a CTLA-4 antagonist, including for example, an anti-CTLA-4 antibody.
  • the method also comprises administering a TIM-3 antagonist.
  • the TIM-3 antagonist is an anti-TIM-3 antibody.
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with a TIM-3 antagonist, including for example, an anti-TIM-3 antibody.
  • the method also comprises administering a LAG3 antagonist.
  • the LAG3 antagonist is an anti-LAG3 antibody.
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with a LAG3 antagonist, including for example, an anti-LAG3 antibody.
  • the method also comprises administering an agonist to a T-cell co-receptor.
  • agonists to a T-cell co-receptor include, but are not limited to an agonistic antibody to a T-cell co-receptor.
  • T-cell co-receptors include, but are not limited to, 4-1BB (CD137) and ICOS (CD278).
  • the CXCL12-targeted immune response agent of the present invention is administered in combination with an agonist to a T-cell co-receptor, and preferably to an agonistic antibody to a T-cell co-receptor, and even more preferably, an agonistic antibody to 4- 1BB (CD137) or ICOS (CD278).
  • the method also comprises administering other anticancer therapies.
  • other anti-cancer therapies include, but are not limited to: chemotherapeutic agents, radiation therapy, cancer therapy, immunotherapy, or cancer vaccines.
  • immunotherapies include, but are not limited to adoptive T-cell therapies or cancer vaccine preparations designed to induce T lymphocytes to recognize tumor cells.
  • the cancer vaccine recognizes one or more tumor antigens expressed on the cancer cells.
  • tumor antigens include, but are not limited to MAGE-A1, MAGE-A2, MAGE- A3, MAGE-A4, MAGE-A5, MAGE-A6, MAGE-A7, MAGE-A8, MAGE-A9, MAGE-A10, MAGE- Al l, MAGE-A12, GAGE-I, GAGE-2, GAGE- 3, GAGE-4, GAGE- 5, GAGE-6, GAGE-7, GAGE- 8, BAGE-I, RAGE- 1, LB33/MUM-1, PRAME, NAG, MAGE-Xp2 (MAGE-B2), MAGE-Xp3 (MAGE-B3), MAGE-Xp4 (MAGE-B4), MAGE- C1/CT7, MAGE-C2, NY-ESO-I, LAGE-I, SSX-I, SSX-2(HOM-MEL-40), SSX-3, SSX-4
  • the anti-cancer therapy includes, but is not limited to: aspirin, sulindac, curcumin, alkylating agents including: nitrogen mustards, such as mechlor- ethamine, cyclophosphamide, ifosfamide, melphalan and chlorambucil; nitrosoureas, such as carmustine (BCNU), lomustine (CCNU), and semustine (methyl-CCNU); thylenimines/methylmelamine such as thriethylenemelamine (TEM), triethylene, thiophosphoramide (thiotepa), hexamethylmelamine (HMM, altretamine); alkyl sulfonates such as busulfan; triazines such as dacarbazine (DTIC); antimetabolites including folic acid analogs such as methotrexate and trimetrexate, pyrimidine analogs such as 5-fluorouracil, fluorodeoxyuridine, gemcitabine
  • alkylating agents including:
  • the CXCL12-targeted immune response agent and the PD-1 signaling inhibitor and/or the anti-cancer therapy are administered simultaneously, separately, or sequentially.
  • the patient is a human.
  • the "patient” or “subject suitable for treatment” may be a mammal, such as a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g.
  • a rodent e.g. a guinea pig, a hamster, a rat, a mouse
  • murine e.g. a mouse
  • canine e.g. a dog
  • feline e.g. a cat
  • equine e.g. a horse
  • primate e.g. a
  • non-human mammals especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.
  • the method is effective to reduce the growth rate and immune evasion of a tumor.
  • a method for treating a cancer comprising administering to a subject in need thereof a pharmaceutically effective amount of a CXCL12- targeted immune response agent.
  • the cancer is an epithelial tumor characterized by bound CXCL12.
  • the cancer is a pancreatic tumor.
  • the pancreatic tumor is a pancreatic ductal adenocarcinoma (PDA).
  • the cancer is colorectal cancer.
  • the cancer is ovarian cancer.
  • the cancer is non-small cell lung cancer (NSCLC).
  • a CXCL12-targeted immune response agent for inducing an immune response to the cancer cells of an epithelial tumor characterized by bound CXCL12.
  • a CXCL12-targeted immune response agent bound to a second molecule may be a pro-apoptotic protein, a radionuclide or toxin, any heterologous molecule, substance or agent capable of binding to a chemokine or cytokine, or a chemotherapeutic agent.
  • the CXCL12-targeted immune response agent is a bispecific antibody or fragment thereof that binds CXCL12 and one or more cytokines.
  • the cytokine(s) induce an immune response to the epithelial tumor cells characterized by bound CXCL12.
  • the cytokine is a pro-inflammatory cytokine.
  • the cytokine is IL-12.
  • IL-12 synergizes with other cytokines, including, but not limited to, IL- 2, IL-6, IL-7, IL-10, IL-15, IL-17, IL-18, IL-21, GM-CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL, etc., to stimulate the immune system and/or enhance the production of tumor-infiltrating effector cells.
  • cytokines including, but not limited to, IL- 2, IL-6, IL-7, IL-10, IL-15, IL-17, IL-18, IL-21, GM-CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL, etc.
  • the tumor is an epithelial tumor characterized by bound CXCL12.
  • the tumor is a pancreatic tumor.
  • the tumor is a pancreatic ductal adenocarcinoma (PDA).
  • the tumor is a colorectal tumor.
  • the tumor is an ovarian tumor.
  • the invention provides a method of promoting T-cell infiltration into cancerous tumor tissue characterized by bound CXCL12 by administering a CXCL12-targeted immune response agent to the individual.
  • the invention provides the use of a CXCL12-targeted immune response agent in the manufacture of a medication for inducing an immune response to an epithelial tumor, preferably, an epithelial tumor characterized by bound CXCL12.
  • Figure 1(A) shows CXCL12 association with cancer cells of mouse and human cancers. Confocal microscopy of serial sections of an orthotopic mouse NSCLC stained with CD3 (white), CD8 (white), CXCL12 (red), and cytokeratin (green). CD3+ T cells are excluded from cytokeratin-positive cancer cells, which are associated with CXCL12.
  • Figure 1(B) shows staining of human NSCLC with anti-CXCL12, demonstrating that the chemokine also associates with cancer cells of the human form of the disease.
  • T-cells or "CD3+ T-cells” are defined as those lymphocyte lineage cells that express the cell surface marker CD3, which includes CD4+ T-cells, CD8+ T-cells, and Foxp3+ regulatory T-cells, and gamma-delta T cells.
  • Effective CD3+ T-cells are defined as those mature T-cell population groups that assist with the activity of other immune cells by releasing T-cell cytokines or have direct cytotoxic function. Such cells include CD4+ T-cells, CD8+ T-cells, and Foxp3+ regulatory T-cells.
  • CXCL12 is a chemokine also known as stroma-derived factor (SDF- 1/CXCL12), is a primary ligand of chemokine receptor CXCR4, and plays significant roles in promoting tumor progression.
  • CXCL12 is constitutively expressed in numerous key organs including lung, liver, kidney, and bone marrow. Many tumors are also associated with high levels of CXCL12 expression.
  • the interaction of CXCL12 and CXCR4 induces intracellular signaling transduction and previous studies have shown that CXCL12/CXCR4 pathway is correlated to oncogenic events such as cancer cell survival, the communication within tumor microenvironment, metastasis, angiogenesis, and evasion of anti-cancer immune responses.
  • the CXCL12/CXCR4 axis represents a promising therapeutic target for cancers.
  • Developing an antibody against CXCL12 can directly target the tumors that are physically associated with CXCL12 and therefore may distinguish normal epithelial cells from those of a malignant organ origin.
  • Zong et al has developed a high-affinity humanized monoclonal antibody specific for CXCL12 in preclinical stage (hu30D8) and has shown antitumor effects in xenograft and orthotopic tumor models (Zong et al., Clin Cancer Res; 19(16): 4433-45, 2013).
  • a "CXCL12-targeted immune response agent” is an exogenous factor, such as a pharmaceutical compound, biological molecule or modified biological material that enhances or induces tumor antigen- specific immune responses, including direct cytotoxicity and Fc receptor-mediated phagocytosis and cytotoxicity, by binding specifically to CXCL12.
  • the CXCL12-targeted immune response agent may be fused to (e.g., covalently attached) or non-covalently attached to a pro-apoptotic protein, a radionuclide or toxin, or a small molecule/chemotherapeutic agent capable of inhibiting or eliminating the tumor cells.
  • the CXCL12-targeted immune response agent may be a bispecific antibody that binds to CXCL12 and a cytokine or chemokine to activate the immune system to destroy the tumor cells.
  • Suitable CXCL12-targeted immune response agents may be identified using standard in vitro or ex vivo assays, such as by staining tissue sections with an anti-CXCL12 antibody.
  • CXCL12-targeted immune response agents include, but are not limited to, anti-CXCL12 antibodies, bispecific antibodies, CAR T-cells engineered to express a chimeric antigen receptor against CXCL12, and T-cells preloaded with bispecific antibodies.
  • T-cells are preloaded with bispecific antibodies that deliver a cytokine to the tumor cell.
  • anti-CXCL12 antibody is defined as a monoclonal antibody that exclusively recognizes the antigen, CXCL12, and thereby elicits immune responses, such as Fc receptor-mediated phagocytosis and antibody-dependent cell-mediated cytotoxicity.
  • Preferred examples of anti-CXCL12 antibodies include, but are not limited to, MAB310 (R&D Systems) and hu30D8.
  • bispecific antibodies are recombinant monoclonal antibodies and antibody-like molecules that combine the specificities of two distinct antibodies in one molecule. Thus, they can therefore simultaneously target two distinct antigens.
  • one of the antigens targeted by the bispecific antibody is CD3 which would cause the bispecific antibody to bind to T-cells.
  • the other antigen targeted by the bispecific antibodies is CXCL12 which would cause the bispecific antibody to bind to CXCL12-coated cancer cells.
  • bispecific antibodies include, but are not limited to, bispecific T- cell engager (BiTE) antibodies, dual-affinity retargeting molecules (DARTs), CrossMAb antibodies, DutaMabTM antibodies, DuoBody antibodies; Triomabs, TandAbs, bispecific NanoBodies, T-cells preloaded with bispecific antibodies, polyclonally- activated T-cells preloaded with bispecific antibodies, Tandem scFvs, diabodies, single chain diabodies, HSA bodies, (scFv)2 HSA antibodies, scFv-IgG antibodies, Dock and Lock bispecific antibodies, DVD-IgG antibodies, TBTI DVD IgG antibodies, IgG-fynomers, Tetravalent bispecific tandem IgG antibodies, dual-targeting domain antibodies, chemically linked bispecific (Fab')2 molecules, crosslinked mAbs, dual-action Fab IgG antibodies (DAF-IgGs), orthoFab-IgG antibodies, bispecific
  • Dual-Affinity Retargeting (DART) platform technology is a type of bispecific antibody developed by MacroGenics.
  • the platform is capable of targeting multiple different epitopes with a single recombinant molecule and is specifically engineered to accommodate various region sequences in a "plug-and-play" fashion.
  • a proprietary covalent linkage is developed and thus, the molecule possesses exceptional stability, optimal heavy and light chain pairing, and predictable antigen recognition.
  • the DART platform is believed to reduce the probability for immunogenicity.
  • Cross monoclonal antibodies are a type of bispecific antibody invented by Roche.
  • the purpose of this technology is to create a bispecific antibody that closely resembles a natural IgG mAb as a tetramer consisting of two light chain-heavy chain pairs, and to solve the problem of light chain mispairing.
  • This technology is believed to prevent unspecific binding of the light chain to its heavy counterpart thereby prevent unwanted side products.
  • this method leaves the antigen-binding regions of the parental antibodies intact and thus can convert any antibodies into a bispecific IgG.
  • a DutaMab is a type of bispecific antibody invented by Dutalys (recently acquired by Roche). This platform differs by developing fully human bispecific antibodies that show high affinity in each arm and simultaneously bind both targets. DutaMabs are also believed to possess excellent stability and good manufacturing properties.
  • Duobody antibodies are a type of bispecific antibodies created by Genmab. This platform generates stable bispecific human IgGl antibodies and is able to fully retain IgGl structure and function. Two parental IgGl monoclonal antibodies are first separately produced, each containing single matched mutations in the third constant domain. Subsequently, these IgGl antibodies are purified according to standard processes for recovery and purification. After production and purification (post-production), the two antibodies are recombined under tailored laboratory conditions resulting in a bispecific antibody product with a very high yield (typically >95%) (Labrijn et al, PNAS 2013;110(13):5145-5150). The Duobody platform is believed to have minimal immunogenicity and can combine any antigen binding sequence derived from any antibody-generating platform to generate a bispecific product.
  • CAR T-cells also known as chimeric antigen receptor T-cells
  • T-cells are produced by using adoptive cell transfer technique. T-cells are first collected from patients' blood and recombinant receptors are introduced into these T-cells using genetic engineering methods such as retroviruses. CAR T-cells are then infused into the patient, the tumor-associated antigen is recognized by the CAR T-cell, and is destroyed. Thus, CAR T-cells enhance tumor specific immunosurveillance.
  • the structure of CAR most commonly incorporates a single-chain variable fragment (scFv) derived from a monoclonal antibody that links to intracellular signaling domains and forms a single chimeric protein.
  • the CAR T-cell is developed specifically against CXCL12.
  • CD47 also known as Integrin Associated Protein, is a transmembrane receptor that belongs to the immunoglobulin superfamily and is ubiquitously expressed on the surface of normal and solid tumor cells. CD47 is also involved in numerous normal and pathological processes including immunity, apoptosis, proliferation, migration, and inflammation. Previous studies have demonstrated the expression of CD47 on various cancer cells and revealed its role in promoting cancer progression. By binding with signal regulatory protein (SIRPa), the primary ligand of CD47 expressed on phagocytic cells (dendritic cells, macrophages, and neutrophils), CD47 prohibits phagocytosis and thus allows tumor cells to evade immune surveillance. Thus, CD47 appears as an important therapeutic target for cancer treatments. Anti-CD47 monoclonal antibodies for clinical uses are currently being developed by Stanford University (phase I, cancer treatment), by the Ukraine Antitumor Center (phase I, cancer treatment), and by Vasculox, Inc. (Preclinical, organ transplantation).
  • SIRPa signal regulatory protein
  • anti-CD47 antibody is defined as a monoclonal antibody that exclusively recognizes and binds to the antigen, CD47. Binding prevents the interaction between CD47 and SIRPa, a protein on phagocytes, thereby reversing the inhibition of phagocytosis normally caused by the CD47/ SIRPa interaction.
  • the anti-CD47 antibody When coupled with a cancer antigen- specific antibody, the anti-CD47 antibody eliminates the "don't eat me signal" and allows the cancer antigen-specific antibody to more efficiently induce a tumor antigen-specific immune response.
  • antibody-dependent cell-mediated cytotoxicity is a mechanism of cell- mediated immune defense whereby an effector cell of the immune system actively lyses a target cell, whose membrane-surface antigens have been bound by specific antibodies.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that immunospecifically binds an antigen.
  • antibody encompasses not only whole antibody molecules, but also antibody fragments as well as variants (including derivatives) of antibodies and antibody fragments.
  • antibody examples include, but are not limited to: single chain Fvs (scFvs), Fab fragments, Fab' fragments, F(ab')2, disulfide linked Fvs (sdFvs), Fvs, and fragments comprising or alternatively consisting of, either a VL or a VH domain.
  • scFvs single chain Fvs
  • Fab fragments fragments
  • Fab' fragments fragments
  • F(ab')2 disulfide linked Fvs
  • sdFvs disulfide linked Fvs
  • Fvs fragments comprising or alternatively consisting of, either a VL or a VH domain.
  • Antibodies of the invention include, but are not limited to, monoclonal, multispecific, bi-specific, human, humanized, mouse, or chimeric antibodies, single chain antibodies, Fab fragments, F(ab') fragments, antiidiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies to antibodies of the invention), and epitope-binding fragments of any of the above.
  • the immunoglobulin molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgGl, IgC2, IgG3, IgG4, IgAl and IgA2) or subclass of immunoglobulin molecule.
  • a "PD-1 signaling inhibitor” is an exogenous factor, such as a pharmaceutical compound or molecule that inhibits or prevents the activation of PD-1 by its ligand PD-L1 and thereby blocks or inhibits PD-1 signaling in cells within the cancerous tumor.
  • a PD-1 signaling inhibitor is defined broadly as any molecule that prevents the negatively regulation by PD- 1 of T-cell activation.
  • Preferred examples of a PD-1 signaling inhibitor include, but are not limited to, a PD-1 antagonist and/or a PD-L1 antagonist.
  • a "PD-1 antagonist” is defined as a molecule that inhibits PD-1 signaling by binding to or interacting with PD-1 to prevent or inhibit the binding and/or activation of PD-1 by PD-L1, thereby inhibiting PD-1 signaling and/or enhancing T-cell activation.
  • Preferred examples of a PD-1 antagonist include, but are not limited to an anti-PD-1 antibody, which are well known in the art. See, Topalian, et al. NEJM 2012.
  • a "PD-L1 antagonist” is defined as a molecule that inhibits PD- 1 signaling by binding to or inhibiting PD-L1 from binding and/or activating PD-1, thereby inhibiting PD-1 signaling and/or enhancing T-cell activation.
  • Preferred examples of a PD-L1 antagonist include, but are not limited to an anti-PD-Ll antibody which are well known in the art. See, Brahmer, et al. NEJM 2012.
  • a "CTLA-4 antagonist” is defined as a molecule that inhibits CTLA-4 signaling by binding to or inhibiting CTLA-4 from binding and/or activating to B7 molecules, known in the art to be present on antigen-presenting cells, thereby preventing interactions of B7 molecules with the co-stimulatory molecule CD28, and inhibiting T-cell function.
  • Preferred embodiments of a CTLA-4 antagonist include, but are not limited to anti- CTLA-4 antibodies.
  • a "LAG3 antagonist” is defined as a molecule that inhibits LAG3 signaling by binding to or inhibiting LAG3 from binding and/or activating MHC molecules and any other molecule, known in the art to be present on antigen-presenting cells, thereby preventing LAG3 interactions and promoting T-cell function.
  • Preferred embodiments of a LAG3 antagonist include, but are not limited to anti-LAG3 antibodies.
  • a "TIM-3 antagonist” is defined as a molecule that inhibits the CD8+ and CD4+ Thl-specific cell surface protein, TIM-3, which, when ligated by galectin-9, for example, causes T-cell death.
  • Preferred embodiments of a TIM-3 antagonist include, but are not limited to anti-TIM-3 antibodies that block interaction with its ligands.
  • a PD-1 antagonist, a CTLA-4 antagonist, a TIM-3 antagonist, and a LAG3 antagonist are T-cell checkpoint antagonists.
  • Other examples of checkpoint antagonists are well known in the art. Blocking CXCR4 with any CXCR4 signaling inhibitor, leads to the uncovering of the anti-cancer effects of the T-cell checkpoint antagonists.
  • a "T-cell co-receptor” is a cell surface receptor that binds to ligands on antigen- resenting cells that are distinct from the peptide-MHC complex that engages the T-cell receptor. Ligation of T-cell co-receptors enhance the antigen-specific activation of the T-cell by recruiting intracellular signaling proteins (e.g., NFkappaB and PI3 -kinase) inside the cell involved in the signaling of the activated T lymphocyte.
  • intracellular signaling proteins e.g., NFkappaB and PI3 -kinase
  • Preferred embodiments of a T-cell co-receptor antagonist include, but are not limited to anti-T-cell co-receptor antibodies, such as, for example, antibodies directed to 4-lBB(CD137) and ICOS (CD278).
  • the CXCL12-targeted immune response agent of the present invention whether it be an anti-CXCL12 antibody, a bispecific antibody, a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12, or a T-cell preloaded with bispecific antibodies, has synergistic activity with a PD-1 signaling inhibitor of the present invention.
  • the CXCL12-targeted immune response agent is for example, MAB310 or hu30D8 and has synergistic activity with a PD-1 signaling inhibitor of the present invention, such as, for example, an anti-PD-1 antibody or an anti-PD-Ll antibody.
  • the CXCL12-targeted immune response agent has synergistic activity with one or more cytokines, including but not limited to: IL-2, IL-6, IL-7, IL- 10, IL-12, IL-15, IL-17, IL-18, IL-21, IL-27, GM-CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL.
  • Cytokines may stimulate the immune system to inhibit the growth of or identify and eliminate tumor cells.
  • a cytokine is a secreted protein that is involved in cell signalling, including regulation of the immune system.
  • Cytokines are typically produced by immune cells, including macrophages, B lymphocytes, T lymphocytes and mast cells, and other cells such as endothelial cells, fibroblasts, and various stromal cells.
  • a given cytokine may be produced by different types of cells. Some cytokines are responsible for activating immune responses against tumors while other cytokines downregulate the immune system to maintain homeostasis and promote self-tolerance.
  • Cytokines include interleukins, lymphokines, monokines, interferons (IFN), colony stimulating factors (CSF), chemokines, and a variety of other proteins.
  • chemokines are a subset of cytokines that activate the immune system to attract immune cells to the site of inflammation.
  • Chemokines are generally pro-inflammatory cytokines.
  • a pro-apoptotic protein is a protein that activates cellular process leading to apoptosis. Examples of such pro-apoptotic proteins include, but are not limited to caspase-3, FOXP3, or death ligand TNF-related apoptosis-inducing ligand (TRAIL).
  • pro-apoptotic proteins include, but are not limited to, bad, bax, bcl-2, bcl-w, BID, BIM, Caspase 8, CD40, CD40 Ligand, cIAP-2, Cytochrome-C, DR6, Fas, Fas Ligand, HSP27, HSP60, HSP70, HTRA, IGF-1, IGF-2, IGFBP-1, IGFBP-2, IGFBP-3, IGFBP-4, IGFBP-5, IGFBP-6, IGF-lsR, livin, p21, p27, p53, SMAC, Survivin, sTNFRI, sTNFRII, TNF alpha, TNF beta, TRAIL Rl, TRAIL R2, TRAIL R3, TRAIL R4, and XIAP.
  • a toxin is a molecule capable of killing a cell, typically by inhibiting protein synthesis.
  • Toxins include but are not limited to bacterial toxins, such as diphtheria toxins, staphylococcal enterotoxin A, and Pseudomonas exotoxins, plant toxins (e.g., ricin, abrin, mistletoe lectin, modeccin, hemitoxins or class I ribosome inactivating proteins such as PAP, pokeweed antiviral toxin, saporin, bryodin 1, bouganin, and gelolin), etc. (Kreitman 2006).
  • Toxins typically have poor selectivity, and thus, need to be conjugated to a molecule having specificity for a tumor cell, e.g., such as an anti-CXCL12 antibody or fragment thereof, in order to target and kill cancer cells, while leaving noncancerous cells intact and functional. Once bound to the tumor cell, the anti-CXCL12 antibody-toxin conjugate is internalized, and the toxin localizes to the cytoplasm to kill the cell.
  • Toxins may be derived from both plant and bacterial sources. Toxins may be genetically fused or chemically conjugated to the CXCL12-targeted immune response agent.
  • a radionuclide is a radioactive atom that emits excess nuclear energy (radiation) which damages cells and triggers apoptosis.
  • radionuclides include, but are not limited to 131 Iodine, 90 ⁇ Yttrium, 177 Lutetium, 188 Rhenium, 67 Copper, 21 Astatine, 213 Bismuth, 125 Iodine, or 11 indium.
  • the radionuclide can deliver a toxic dose of radiation specifically to tumor cells characterized by CXCL12. Radionuclides not only destroy the target cell, but also adjacent tumor cells that may lack CXCL12.
  • toxins, pro- apoptotic proteins, chemokines or cytokines, radionuclides, and chemotherapeutics in combination with the CXCL12-targeted immune response agent have the capacity to eliminate both a primary tumor site as well as cancer that has spread throughout the body, including malignant cell populations that may be undetectable by diagnostic imaging.
  • the CXCL12-targeted immune response agent of the present invention whether it be an anti-CXCL12 antibody, a bispecific antibody, a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12, or a T-cell preloaded with bispecific antibodies, has synergistic activity with a CTLA-4 antagonist of the present invention.
  • the CXCL12-targeted immune response agent is for example, MAB310 or hu30D8 and has synergistic activity with a CTLA-4 antagonist of the present invention, such as, for example, an anti-CTLA-4 antibody.
  • the CXCL12-targeted immune response agent of the present invention whether it be an anti-CXCL12 antibody, a bispecific antibody, a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12, or a T-cell preloaded with bispecific antibodies, has synergistic activity with a LAG3 antagonist of the present invention.
  • the CXCL12-targeted immune response agent is for example, MAB310 or hu30D8 and has synergistic activity with a LAG3 antagonist of the present invention, such as, for example, an anti-LAG3 antibody.
  • the CXCL12-targeted immune response agent of the present invention whether it be an anti-CXCL12 antibody, a bispecific antibody, a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12, or a T-cell preloaded with bispecific antibodies, has synergistic activity with a TIM-3 antagonist of the present invention.
  • the CXCL12-targeted immune response agent is for example, MAB310 or hu30D8 and has synergistic activity with a TIM-3 antagonist of the present invention, such as, for example, an anti-TIM-3 antibody.
  • the CXCL12-targeted immune response agent of the present invention whether it be an anti-CXCL12 antibody, a bispecific antibody, a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12, or a T-cell preloaded with bispecific antibodies, has synergistic activity with a checkpoint antagonist of the present invention.
  • the CXCL12-targeted immune response agent is for example, MAB310 or hu30D8 and has synergistic activity with a checkpoint antagonist of the present invention and those known in the art.
  • the CXCL12-targeted immune response agent of the present invention whether it be an anti-CXCL12 antibody, a bispecific antibody, a CAR T-cell engineered to express a chimeric antigen receptor against CXCL12, or a T-cell preloaded with bispecific antibodies, has synergistic activity with a T-cell co-receptor antagonist of the present invention.
  • the CXCL12-targeted immune response agent is for example, MAB310 or hu30D8 and has synergistic activity with a T-cell co-receptor antagonist of the present invention, such as, for example, an anti-T-cell co-receptor antibody, for example, an anti-4-lBB (CD137) antibody or an anti-ICOS (CD278)antibody.
  • a T-cell co-receptor antagonist of the present invention such as, for example, an anti-T-cell co-receptor antibody, for example, an anti-4-lBB (CD137) antibody or an anti-ICOS (CD278)antibody.
  • a tumor is defined as a population of heterogeneous cells, collectively forming a mass of tissue in a subject resulting from the abnormal proliferation of malignant cancer cells.
  • the tumor may comprise of p53+ (Gene ID; 2191, reference sequence NP_004451.2 GI: 16933540) cancer cells.
  • the tumors are p53 negative.
  • a tumor will contain both normal or “non-cancerous” cells and “cancer” or “cancerous” cells.
  • a tumor typically comprises or is associated with p53+ and/or FAP+ stromal cells and/or inflammatory/immune cells.
  • the cancer cells are often grouped together in "nests", separated by stromal regions containing extracellular matrix (e.g., collagen), immune cells and FAP+ fibroblastic cells.
  • FAP+ stromal cells in a cancerous tumour may be identified using routine techniques, including protein based methods, such as fluorescence microscopy and immunohistology or nucleic acid based methods, such as RT-PCR.
  • protein based methods such as fluorescence microscopy and immunohistology
  • nucleic acid based methods such as RT-PCR.
  • proximity is defined as the distance between the CD3+ T- cells, and even more preferably effector CD3+ T-cells, and the cancer cells within a tumor.
  • one way to measure "proximity” is to cross-section the tumor, such as a PDA tumor, and then stain the tumor with a cancer detecting antibody, such as anti-p53 (loss-of- heterozygosity at the p53 locus cancer cells may be have mutant p53 protein detectable) and anti- CD3epsilon (T-cells are +). The section is then subjected to ARIOL scanning. An instrument then evaluates the image, and calculates for each p53+ cell the distance to the nearest CD3+ cell.
  • a histogram can then be constructed.
  • increases in the proximity of the T-cells among the cancer cells is increased by at least 2 fold (distance between cancer cell and nearest T-cell is decreased by 2 fold), 3 fold (distance between cancer cell and nearest T-cell is decreased by 3 fold), 4 fold (distance between cancer cell and nearest T-cell is decreased by 4 fold) or 5 fold (distance between cancer cell and nearest T-cell is decreased by 5 fold).
  • An alternative assay is to delineate the areas of the tumor that contain cancers cells, identified by monoclonal appropriate antibodies, and calculate the density of CD3+ T cells contained within these areas and outside these areas, which are taken as stromal regions.
  • effector CD3+ T-cells When effector CD3+ T-cells are in close proximity to the cancerous tumor cell, effector response ensues. Otherwise, if a T cell that has a TCR specific for a cancer cell antigen cannot come into contact with the cancer cell, the T cell will not be activated and its anti-cancer functions are not elicited, allowing the tumor to evade the immune recognition.
  • frequency is defined as the quantitative increase in T-cells and even more preferably effector CD3+ T-cells that are found among the cancer cells in the tumor microenvironment.
  • increases in frequency of the T-cells among the cancer cells is increased by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 100%, at least 200%, or at least 300%.
  • tumors include, but are not limited to, sarcomas, skin cancer, melanoma, bladder cancer, brain cancer, breast cancer, uterus cancer, ovary cancer, prostate cancer, lung cancer, colorectal cancer, cervical cancer, liver cancer, head and neck cancer, esophageal cancer, pancreas cancer, renal cancer, stomach cancer, multiple myeloma and cerebral cancer.
  • Preferred embodiments of tumors are adenocarcinomas.
  • the cancer may be pancreatic cancer, for example pancreatic ductal adenocarcinoma.
  • T-cell exclusion in a tumor is defined as those tumor evasion mechanisms known in the art where effector CD3+ T-cell subsets are prevented from being recruited to and accumulating among cancer cells within the tumor microenvironment.
  • Tumor evasion mechanisms include, but are not limited to: (1) immunologic barriers within the tumor microenvironment, including a failure of immunosurveillance in the tumor, (2) non-functional antigen presenting cells, and (3) dysfunctional CD4+ T-cells, CD8+ T-cells, and excessive numbers of Foxp3+ regulatory T-cells,
  • a model of human PDA was developed to replicate a failure of immunosurveillance in the tumor. This failure is attributable to local immunosuppression mediated by the FAP+ stromal cell, which manifests as exclusion and likely death of T-cells from regions of the tumor containing PDA cells and involves its production of CXCL12.
  • CXCL12 is associated with both impairment and promotion of immune control of growth of tumors.
  • the art that demonstrates impairment of immune control indicates that this results from the recruitment of FoxP3+ regulatory T-cells to the tumor by expression of CXCL12.
  • CXCL12 expression results in exclusion of all T-cells, including CD4+ T-cells, CD8+ T-cells, and Foxp3+ regulatory T-cells.
  • the use of CXCL12-targeted immune response agents are expected to overcome this exclusion, thus eliciting tumor antigen- specific immune responses against the tumor cells.
  • the present invention provides a method for recruitment of CD3+ T-cell subsets, including CD4+ T-cells, CD8+ T-cells, and Foxp3+ regulatory T-cells, to cancer cell- containing regions of a tumor in a subject, and methods for treating tumors by restoring immunological control of tumor growth.
  • the present invention overcomes the problem of T-cell exclusion and allows effector CD3+ T-cell subsets to accumulate and recruit to the cancer cells in order to carry out their endogenous function of eliminating the cancer cells.
  • the described method herein increases the recruitment of effector CD3+ T- cell accumulation in the sites of a tumor that contain cancer cells, comprising administering to a subject in need thereof a pharmaceutically effective amount of a CXCL12-targeted immune response agent.
  • the efficacy of the present invention is based on the observation that FAP+ stromal cells secrete CXCL12 which becomes associated with the cancer cells within the tumor.
  • Administration of a CXCL12-targeted immune response agent as described herein, such as for example an anti-CXCL12 antibody, a bispecific antibody, or a CAR T-cell results in CD3+ effector T-cell recruitment to the cancer cell-containing sites of the tumor and elimination of the cancer cells.
  • anti-CXCL12 antibodies are examples of CXCL12-targeted immune response agents that can be used to recruit CD3+ T-cells to the cancer cell-containing sites of tumors and restore immunological regulation of the cancerous tumor cells. This restoration of immunological surveillance of the cancerous tumor leads to the elimination of the cancerous cells.
  • the described invention increases T-cell accumulation and recruitment to the cancerous tumor cells, such as PDA, to reduce the tumor growth and overcome tumor evasion mechanisms.
  • PDA tumors like most solid tumors, PDA tumors contain stromal cells that express fibroblast activation protein (FAP). FAP+ stromal cells are found in both PDA and other tumors and are known to secrete CXCL12.
  • FAP fibroblast activation protein
  • One tumor evasion strategy is for cancer cells to bind CXCL12 and suppress local immune regulation of the tumor by excluding effector T-cells from accumulating amongst the cancer cells.
  • a CXCL12-targeted immune response agent such as, for example, anti-CXCL12 antibodies, bispecific antibodies or CAR T-cells, immune regulation of the tumor is restored.
  • CD3+ T-cells that accumulate to the cancerous cells, such as PDA cells, when in the presence of a CXCL12-targeted immune response agent, such as, for example, anti-CXCL12 antibodies, bispecific antibodies or CAR T-cells, and these T-cells restore immunological regulation of the tumor.
  • a CXCL12-targeted immune response agent such as, for example, anti-CXCL12 antibodies, bispecific antibodies or CAR T-cells
  • this invention provides a method to treat a cancer comprised subject, such as a subject who contains PDA, by administering to a subject in need thereof a pharmaceutically effective amount of a CXCL12-targeted immune response agent.
  • a CXCL12-targeted immune response agent is able to reduce immune suppression, increase infiltration of effector T-cells amongst the cancer cells, restore immunological regulation of the tumor, and effectively reduce and eliminate cancer cells, preferably, in a tumor comprised of FAP+ stromal cells.
  • This invention relates to the use of CXCL12-targeted immune response agents to reduce or abolish tumor immunosuppression in an individual with cancer.
  • the CXCL12-targeted immune response agent described here can be used to initiate or enhance immune responses against cancer cells in a subject, in particular cell-mediated immune responses.
  • the CXCL12-targeted immune response agent as described herein reduces the ability of the cancerous tumor to suppress immune responses, for example by excluding CD3+ T-cell subsets, such that immune responses to the tumor are more effective in the subject. This may have a beneficial therapeutic effect on the cancerous tumor of a human patient.
  • the anti-CXCL12 targeting agent can be used to deliver a second molecule to the tumor cells in order to eliminate or inhibit the growth of the tumor cells.
  • the second molecule is a pro-apoptotic protein that triggers apoptosis, a toxin or radionuclide that destroys the tumor cells, a chemotherapeutic agent/anti-cancer compound or any other heterologous molecule, substance, or agent capable of binding to a chemokine or cytokine that triggers the immune system to eliminate the tumor cell.
  • the anti-CXCL12 targeting agent is a bidirectional antibody molecule or fragment thereof that binds CXCL12 and also binds to a cytokine.
  • the bidirectional antibody may bind to endogenously expressed cytokine(s).
  • the bidirectional antibody may be pre-loaded with a particular cytokine before infusion into the patient.
  • Cytokines stimulate effector cells and stromal cells at the tumor site, thereby, enhancing tumor cell recognition by cytotoxic effector cells (Lee et al., 2011), and have broad anti-tumor activity.
  • IL- 12 regulates both innate and adaptive immune responses, induces anticancer effects, and synergizes with other cytokines for increased immunoregulatory and antitumor activities (Lee et al., 2011).
  • the cytokine is IL- 12.
  • IL-12 may be delivered to the tumor via a bispecific antibody or any other molecule that can bind to IL-12 to deliver IL-12 to the site of the tumor.
  • Other cytokines include IL-2, IL-6, IL-7, IL- 10, IL-15, IL-17, IL- 18, IL-21 , GM- CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL.
  • cytokines are proinflammatory.
  • IL-12 may inhibit tumor growth by activating the immune system.
  • IL- 12 is known to activate T and natural killer (NK) cells, as well as facilitate presentation of tumor antigens through the upregulation of class I and II major histocompatibility (MHC) molecules, and to generate T helper type I (Thl) immune responses (Weiss et al., 2007).
  • IL- 12 also is known to exert potent anti-tumor effects in solid tumors, leukemias, lymphomas and melanomas, as well as induce expression of IFN- ⁇ in developing T cells, thereby priming these T-cells to stably produce IFN- ⁇ (Id.).
  • the CXCL12-targeted immune response agent as described herein may be administered in combination with a cytokine, such as IL- 12.
  • the CXCL12- targeted immune response agent as described herein may bind endogenous cytokine, acting to increase the local concentration of the cytokine, e.g., IL-12.
  • the CXCL12-targeted immune response agent is a bispecific antibody that binds both CXCL12 and a cytokine such as IL-12.
  • inhibition of CXCL12 alleviates suppression of immune responses (specifically, the exclusion of T cells from the tumor microenvironment), while delivery of IL-12 stimulates the immune system.
  • the CXCL12- targeted immune response agent and IL- 12 act synergistically to activate the immune system to inhibit the growth of or eliminate tumor cells.
  • IL- 12 is known to be associated with side effects including toxicity.
  • the CXCL12-targeted immune response agent which can specifically deliver IL-12 to the tumor site, IL-12 can be delivered in a sufficient amount to activate the immune system while reducing or eliminating toxic side effects.
  • any other cytokine having toxic side effects may be bound to the CXCL12-targeted immune response agent to reduce toxicity.
  • the CXCL12-targeted immune response agent as described herein may be administered in combination with multiple cytokines. Cytokines may synergize with other cytokine(s) to reduce immunosuppressive effects from administration of a particular cytokine.
  • IL-12 attenuated other cytokines involved in stimulation of the immune system, including INF- ⁇ (Weiss et al. 2007).
  • IL- 12 may be coadministered with IL-2 to overcome the inhibition of INF- ⁇ production.
  • IL-12 may synergize with cytokines to elicit higher levels of T and NK cell activation and T cell proliferation than if administered individually.
  • IL-12 in combination with IL-2 reportedly induced IFN- ⁇ , TNFa and GM-CSF production at a higher rate in T and NK cells than with IL- 12 alone (Id.).
  • the anti-CXCL12 antibody or fragment thereof and IL- 12 is coadministered with IL-15; the anti-CXCL12 antibody or fragment thereof and IL- 12 is coadministered with IL-7; the anti-CXCL12 antibody or fragment thereof and IL-12 is coadministered with IL-21 ; the anti-CXCL12 antibody or fragment thereof and IL- 12 is coadministered with IL-18; the anti-CXCL12 antibody or fragment thereof and IL- 12 is coadministered with GM-CSF; or the anti-CXCL12 antibody or fragment thereof and IL- 12 coadministered with INF-a (Weiss et al., 2007). Coadministration can alleviate attenuation of cytokines needed for immune system activation as well as synergize with each other to activate the immune system.
  • the anti-CXCL12 antibody or fragment thereof or any other molecule capable of binding to CXCL12 is fused to a radionuclide.
  • radionuclides include, but are not limited to 131 Iodine, 9a yYttrium, 177 Lutetium, 188 Rhenium, 67 Copper, 21 Astatine, 213 Bismuth, 125 Iodine, or 11 indium.
  • the radionuclides emit radiation causing destruction of the tumor cell.
  • the anti-CXCL12 antibody is fused to a toxin.
  • toxins include, but are not limited to Pseudomonas exotoxin, staphylococcal enterotoxin A, ricin A-chain, or plant ribosome-inactivating protein saporin.
  • Other bacterial toxins include diphtheria toxins and other plant toxins include abrin, mistletoe lectin, modeccin, hemitoxins, PAP, pokeweed antiviral toxin, saponin, bryodin 1, bouganin, and gelolin, etc.
  • the toxins may block protein synthesis leading to the destruction of the tumor cell.
  • the anti-CXCL12 antibody is fused to a pro-apoptotic protein.
  • pro-apoptotic proteins include, but are not limited to caspase-3, FOXP3, or death ligand TNF-related apoptosis-inducing ligand (TRAIL).
  • pro-apoptotic proteins include but are not limited to bad, bax, bcl-2, bcl-w, BID, BIM, Caspase 8, CD40, CD40 Ligand, cIAP-2, Cytochrome-C, DR6, Fas, Fas Ligand, HSP27, HSP60, HSP70, HTRA, IGF-1, IGF-2, IGFBP-1, IGFBP-2, IGFBP-3, IGFBP-4, IGFBP-5, IGFBP-6, IGF-lsR, livin, p21, p27, p53, SMAC, Survivin, sTNFRI, sTNFRII, TNF alpha, TNF beta, TRAIL Rl, TRAIL R2, TRAIL R3, TRAIL R4, and XIAP.
  • pro-apoptotic proteins when triggered, lead to the death of the tumor cell.
  • kits for cancer immunotherapy in an individual in need thereof, which comprise administering to the individual a CXCL12-targeted immune response agent as described herein in an amount effective to treat the cancer, for example by increasing the effectiveness of the host immune response against the cancer in the individual.
  • a CXCL12-targeted immune response agent can be used to increase T-cell accumulation and recruitment at the cancer containing sites in a tumor.
  • the present invention also relates to the use of a CXCL12-targeted immune response agent in the manufacture of a medication for use in increasing T-cell accumulation at the cancer containing sites in a tumor.
  • T-cell therapy includes therapies which reduce the rate of tumor growth, that is slow down, but do not necessarily eliminate tumor growth.
  • Reduction in the rate of tumor growth can be, for example, a reduction in at least 10%, 20%, 30%, 40%, 50%, 75%, 100%, 150%, 200% or more of the rate of growth of a tumor.
  • the rate of growth can be measured over 1, 2, 3, 4, 5, 6 or 7 days, or for longer periods of one or more weeks.
  • the invention may result in the arrest of tumor growth, or the reduction in tumor size or the elimination of a tumor.
  • Cancer cells within the tumor in the subject may be immunologically distinct from normal somatic cells in the subject (for example, the tumor may be immunogenic; alternatively, even if it is not immunogenic, it may present different immunological determinants(s) from somatic cells).
  • the cancer cells may be capable of eliciting a systemic immune response in the subject against one or more antigens expressed by the cancer cells.
  • the antigens that elicit the immune response may be tumor antigens or may be shared by normal cells.
  • the tumor although presenting different antigenic determinants, is hidden from the immune system of a subject and displays tumor evasion characteristics.
  • the tumor may exclude immune cells, thus lowering its immunological visibility and sensitivity, and/or preventing the immune system from acting to attack the tumor.
  • CD8+ T-cells that are specific for cancer cells within the cancerous tumor may be present in the subject.
  • CD8+ T-cells may be recruited to the cancerous tumor.
  • the cancer cells may express one or more antigens that are not expressed by normal somatic cells in the subject (i.e. tumor antigens).
  • tumor antigens are known in the art and may elicit immune responses in the subject.
  • tumor antigens may elicit T-cell- mediated immune responses against cancer cells in the subject i.e. the tumor antigens may be recognized by CD8+ T-cells in the subject.
  • Tumor antigens expressed by cancer cells in a cancerous tumor may include, for example, cancer-testis (CT) antigens encoded by cancer-germ line genes, such as MAGE-A1, MAGE-A2, MAGE- A3, MAGE-A4, MAGE-A5, MAGE-A6, MAGE-A7, MAGE-A8, MAGE- A9, MAGE- A 10, MAGE-A11, MAGE-A12, GAGE-I, GAGE-2, GAGE-3, GAGE-4, GAGE-5, GAGE-6, GAGE-7, GAGE- 8, BAGE-I, RAGE- 1, LB33/MUM-1, PRAME, NAG, MAGE-Xp2 (MAGE-B2), MAGE-Xp3 (MAGE-B3), MAGE-Xp4 (MAGE-B4), MAGE- C1/CT7, MAGE- C2, NY-ESO-I, LAGE-I, SSX-I, SSX-2(HOM-MEL-40), SSX-3,
  • CT cancer
  • tumor antigens that may be expressed include, for example, overexpressed or mutated proteins and differentiation antigens particularly melanocyte differentiation antigens such as p53, ras, CEA, MUC1, PMSA, PSA, tyrosinase, Melan-A, MART-1, gplOO, gp75, alpha- actinin-4, Bcr-Abl fusion protein, Casp-8, beta-catenin, cdc27, cdk4, cdkn2a, coa-1, dek-can fusion protein, EF2, ETV6-AML1 fusion protein, LDLR-fucosyltransferaseAS fusion protein, HLA-A2, HLA-A11, hsp70-2, KIAAO205, Mart2, Mum-2, and 3, neo-PAP, myosin class I, OS- 9, pml-RAR.alpha.
  • melanocyte differentiation antigens such as p53, ra
  • fusion protein PTPRK, K-ras, N-ras, Triosephosphate isomeras, GnTV, Herv-K-mel, NA-88, SP17, and TRP2- 2, (MART-I), E2A-PRL, H4-RET, IGH-IGK, MYL- RAR, Epstein Barr virus antigens, EBNA, human papillomavirus (HPV) antigens E6 and E7, TSP-180, MAGE-4, MAGE-5, MAGE-6, pl85erbB2, pl80erbB-3, c-met, nm-23Hl, PSA, TAG- 72-4, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, alpha.-fetoprotein, 13HCG, BCA225, BTAA, CA 125, CA 15-3 (CA 27.29 ⁇ BCAA), CA 195, CA 242, CA-50, CAM43, CD68 ⁇ KP1, CO-029,
  • tumor antigens that may be expressed include out-of-frame peptide-MHC complexes generated by the non-AUG translation initiation mechanisms employed by "stressed" cancer cells (Malarkannan et al. Immunity 1999).
  • tumor antigens that may be expressed are well-known in the art (see for example WO00/20581; Cancer Vaccines and Immunotherapy (2000) Eds Stern, Beverley and Carroll, Cambridge University Press, Cambridge) The sequences of these tumor antigens are readily available from public databases but are also found in WO 1992/020356 Al, WO 1994/005304 Al, WO 1994/023031 Al, WO 1995/020974 Al, WO 1995/023874 Al & WO 1996/026214 Al.
  • a subject suitable for treatment as described above may be a mammal, such as a rodent (e.g. a guinea pig, a hamster, a rat, a mouse), murine (e.g. a mouse), canine (e.g. a dog), feline (e.g. a cat), equine (e.g. a horse), a primate, simian (e.g. a monkey or ape), a monkey (e.g. marmoset, baboon), an ape (e.g. gorilla, chimpanzee, orangutan, gibbon), or a human.
  • a rodent e.g. a guinea pig, a hamster, a rat, a mouse
  • murine e.g. a mouse
  • canine e.g. a dog
  • feline e.g. a cat
  • equine e.g. a horse
  • the subject is a human.
  • non-human mammals especially mammals that are conventionally used as models for demonstrating therapeutic efficacy in humans (e.g. murine, primate, porcine, canine, or rabbit animals) may be employed.
  • the subject may have minimal residual disease (MRD) after an initial cancer treatment.
  • MRD minimal residual disease
  • a subject with cancer may display at least one identifiable sign, symptom, or laboratory finding that is sufficient to make a diagnosis of cancer in accordance with clinical standards known in the art. Examples of such clinical standards can be found in textbooks of medicine such as Harrison's Principles of Internal Medicine, 15th Ed., Fauci AS et al., eds., McGraw-Hill, New York, 2001.
  • a diagnosis of a cancer in a subject may include identification of a particular cell type (e.g. a cancer cell) in a sample of a body fluid or tissue obtained from the subject.
  • a suitable serum concentration of CXCL12-targeted immune response agent for the induction of an immune response to an epithelial tumor characterized by bound CXCL12 may be readily determined from in vivo assays.
  • the CXCL12-targeted immune response agent may be administered together with other anti-cancer therapies, such as conventional chemotherapeutic agents, radiation therapy or cancer immunotherapy.
  • the CXCL12-targeted immune response agent is administered together with an anti-cancer compound.
  • the CXCL12-targeted immune response agent and the anti-cancer compound may be separate compounds or molecules or they may be covalently or non-covalently linked in a single compound, molecule, particle or complex.
  • An anti-cancer compound may be any anti-cancer drug or medicament which has activity against cancer cells.
  • Suitable anti-cancer compounds for use in combination with CXCR4 as disclosed herein may include aspirin, sulindac, curcumin, alkylating agents including: nitrogen mustards, such as mechlor-ethamine, cyclophosphamide, ifosfamide, melphalan and chlorambucil; nitrosoureas, such as carmustine (BCNU), lomustine (CCNU), and semustine (methyl-CCNU); thylenimines/methylmelamine such as thriethylenemelamine (TEM), Methylene, thiophosphoramide (thiotepa), hexamethylmelamine (HMM, altretamine); alkyl sulfonates such as busulfan; triazines such as dacarbazine (DTIC); antimetabolites including folic acid analogs such as methotrexate and trimetrexate, pyr
  • a pharmaceutical composition may comprise, in addition to the CXCL12-targeted immune response agent and/or an anti-cancer compound, one or more pharmaceutically acceptable carriers, adjuvants, excipients, diluents, fillers, buffers, stabilizers, preservatives, lubricants, or other materials well known to those skilled in the art. Suitable materials will be sterile and pyrogen-free, with a suitable isotonicity and stability. Examples include sterile saline (e.g.
  • compositions may further contain auxiliary substances such as wetting agents, emulsifying agents, pH buffering agents or the like.
  • Suitable carriers, excipients, etc. can be found in standard pharmaceutical texts, for example, Remington's Pharmaceutical Sciences, 18th edition, Mack Publishing Company, Easton, Pa., 1990.
  • pharmaceutically acceptable refers to compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • a subject e.g. human
  • Each carrier, excipient, etc. must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • one or both of the CXCL12-targeted immune response agent and anti-cancer compound may be provided in a lyophilized form for reconstitution prior to administration.
  • lyophilized reagents may be re-constituted in sterile water and mixed with saline prior to administration to a subject
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. Such methods include the step of bringing into association the active compound with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active compound with liquid carriers or finely divided solid carriers or both, and then if necessary shaping the product.
  • Formulations may be in the form of liquids, solutions, suspensions, emulsions, elixirs, syrups, tablets, lozenges, granules, powders, capsules, cachets, pills, ampoules, suppositories, pessaries, ointments, gels, pastes, creams, sprays, mists, foams, lotions, oils, boluses, electuaries, or aerosols.
  • other therapeutic or prophylactic agents may be included in a pharmaceutical composition or formulation.
  • Increasing immune response to tumors as described herein may be useful in immunotherapy for the treatment of cancer.
  • Treatment may be any treatment and therapy, whether of a human or an animal (e.g. in veterinary applications), in which some desired therapeutic effect is achieved, for example, the inhibition or delay of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, cure or remission (whether partial or total) of the condition, preventing, delaying, abating or arresting one or more symptoms and/or signs of the condition or prolonging survival of a subject or patient beyond that expected in the absence of treatment.
  • some desired therapeutic effect is achieved, for example, the inhibition or delay of the progress of the condition, and includes a reduction in the rate of progress, a halt in the rate of progress, amelioration of the condition, cure or remission (whether partial or total) of the condition, preventing, delaying, abating or arresting one or more symptoms and/or signs of the condition or prolonging survival of a subject or patient beyond that expected in the absence of treatment.
  • Treatment as a prophylactic measure is also included.
  • a subject susceptible to or at risk of the occurrence or re-occurrence of cancer may be treated as described herein. Such treatment may prevent or delay the occurrence or re-occurrence of cancer in the subject.
  • treatment may include inhibiting cancer growth, including complete cancer remission, and/or inhibiting cancer metastasis.
  • Cancer growth generally refers to any one of a number of indices that indicate change within the cancer to a more developed form.
  • indices for measuring an inhibition of cancer growth include a decrease in cancer cell survival, a decrease in tumor volume or morphology (for example, as determined using computed tomographic (CT), sonography, or other imaging method), a delayed tumor growth, a destruction of tumor vasculature, improved performance in delayed hypersensitivity skin test, an increase in the activity of cytolytic T-lymphocytes, and a decrease in levels of tumor- specific antigens.
  • CT computed tomographic
  • CXCL12-targeted immune response agents may be administered as described herein in therapeutically-effective amounts .
  • terapéuticaally-effective amount refers to that amount of an active compound, or a combination, material, composition or dosage form comprising an active compound, which is effective for producing some desired therapeutic effect, commensurate with a reasonable benefit/risk ratio.
  • appropriate dosages of the active compounds can vary from patient to patient. Determining the optimal dosage will generally involve the balancing of the level of therapeutic benefit against any risk or deleterious side effects of the administration.
  • the selected dosage level will depend on a variety of factors including, but not limited to, the route of administration, the time of administration, the rate of excretion of the active compound, other drugs, compounds, and/or materials used in combination, and the age, sex, weight, condition, general health, and prior medical history of the patient.
  • the amount of active compounds and route of administration will ultimately be at the discretion of the physician, although generally the dosage will be to achieve concentrations of the active compound at a site of therapy without causing substantial harmful or deleterious side-effects.
  • a suitable dose of the active compound is in the range of about 100 ⁇ g to about 250 mg per kilogram body weight of the subject per day.
  • the active compound is a salt, an ester, prodrug, or the like
  • the amount administered is calculated on the basis of the parent compound and so the actual weight to be used is increased proportionately.
  • a CXCL12-targeted immune response agent as described herein such as such as, for example, anti-CXCL12 antibodies, bispecific antibodies, or CAR T-cells may be administered by continuous intravenous infusion in an amount sufficient to maintain the serum concentration at a level that inhibits tumor growth.
  • Other CXCL12-targeted immune response agents described herein can also be used in this same manner.
  • Administration in vivo can be effected in one dose, continuously or intermittently (e.g., in divided doses at appropriate intervals). Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the formulation used for therapy, the purpose of the therapy, the target cell being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the physician. [00156] Administration of anti-cancer compounds and the CXCL12-targeted immune response agent may be simultaneous, separate or sequential. By “simultaneous" administration, it is meant that the anti-cancer compounds and the CXCL12-targeted immune response agents are administered to the subject in a single dose by the same route of administration.
  • the anti-cancer compounds and the CXCL12-targeted immune response agent are administered to the subject by two different routes of administration which occur at the same time. This may occur for example where one agent is administered by infusion or parenterally and the other is given orally during the course of the infusion or parenteral administration.
  • the anti-cancer compounds and the CXCL12-targeted immune response agent are administered at different points in time, provided that the activity of the first administered agent is present and ongoing in the subject at the time the second agent is administered.
  • the anti-cancer compounds may be administered first, such that an immune response against a tumor antigen is generated, followed by administration of the CXCL12-targeted immune response agent, such that the immune response at the site of the tumor is enhanced, or vice versa.
  • a sequential dose will occur such that the second of the two agents is administered within 48 hours, preferably within 24 hours, such as within 12, 6, 4, 2 or 1 hour(s) of the first agent.
  • Multiple doses of the CXCL12-targeted immune response agent may be administered, for example 2, 3, 4, 5 or more than 5 doses may be administered after administration of the anticancer compounds.
  • the administration of the CXCL12-targeted immune response agent may continue for sustained periods of time after administration of the anti-cancer compounds. For example treatment with the CXCL12-targeted immune response agent may be continued for at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month or at least 2 months. Treatment with the CXCL12-targeted immune response agent may be continued for as long as is necessary to achieve complete tumor rejection.
  • Multiple doses of the anti-cancer compounds may be administered, for example 2, 3, 4, 5 or more than 5 doses may be administered after administration of the CXCL12-targeted immune response agent.
  • the administration of the anti-cancer compounds may continue for sustained periods of time after administration of the CXCL12-targeted immune response agent.
  • treatment with the anti-cancer compounds may be continued for at least 1 week, at least 2 weeks, at least 3 weeks, at least 1 month or at least 2 months. Treatment with the anti- cancer compounds may be continued for as long as is necessary to achieve complete tumor rejection.
  • the active compounds or pharmaceutical compositions comprising the active compounds may be administered to a subject by any convenient route of administration, whether systemically/ peripherally or at the site of desired action, including but not limited to, oral (e.g. by ingestion); and parenteral, for example, by injection, including subcutaneous, intradermal, intramuscular, intravenous, intraarterial, intracardiac, intrathecal, intraspinal, intracapsular, subcapsular, intraorbital, intraperitoneal, intratracheal, subcuticular, intraarticular, subarachnoid, and intrasternal; by implant of a depot, for example, subcutaneously or intramuscularly.
  • administration will be by the intravenous route, although other routes such as intraperitoneal, subcutaneous, transdermal, oral, nasal, intramuscular or other convenient routes are not excluded.
  • compositions comprising the active compounds may be formulated in suitable dosage unit formulations appropriate for the intended route of administration.
  • Formulations suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets, each containing a predetermined amount of the active compound; as a powder or granules; as a solution or suspension in an aqueous or nonaqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion; as a bolus; as an electuary; or as a paste.
  • a tablet may be made by conventional means, e.g., compression or molding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active compound in a free-flowing form such as a powder or granules, optionally mixed with one or more binders (e.g. povidone, gelatin, acacia, sorbitol, tragacanth, hydroxypropylmethyl cellulose); fillers or diluents (e.g. lactose, microcrystalline cellulose, calcium hydrogen phosphate); lubricants (e.g. magnesium stearate, talc, silica); disintegrants (e.g.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active compound therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile.
  • Tablets may optionally be provided with an enteric coating, to provide release in parts of the gut other than the stomach.
  • Formulations suitable for parenteral administration include aqueous and nonaqueous isotonic, pyrogen-free, sterile injection solutions which may contain anti-oxidants, buffers, preservatives, stabilizers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to target the compound to blood components or one or more organs.
  • aqueous and nonaqueous isotonic, pyrogen-free, sterile injection solutions which may contain anti-oxidants, buffers, preservatives, stabilizers, bacteriostats, and solutes which render the formulation isotonic with the blood of the intended recipient
  • aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents, and liposomes or other microparticulate systems which are designed to
  • Suitable isotonic vehicles for use in such formulations include Sodium Chloride Injection, Ringer's Solution, or Lactated Ringer's Injection.
  • concentration of the active compound in the solution is from about 1 ng/ml to about 10 ⁇ g/ml, for example from about 10 ng/ml to about 1 ⁇ g/ml.
  • the formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules, and tablets.
  • Formulations may be in the form of liposomes or other microparticulate systems which are designed to target the active compound to blood components or one or more organs.
  • compositions comprising anti-cancer compounds and/or CXCL12-targeted immune response agents may be prepared in the form of a concentrate for subsequent dilution, or may be in the form of divided doses ready for administration.
  • the reagents may be provided separately within a kit, for mixing prior to administration to a human or animal subject.
  • the CXCL12-targeted immune response agent may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the individual circumstances.
  • CXCL12-targeted immune response agents as described herein may be administered in combination with one or more additional active compounds.
  • the CXCL12-targeted immune response agents as described herein may be administered in combination with one or more additional cytokines.
  • Cytokines include, but are not limited to: IL-2, IL-6, IL-7, IL-10, IL-12, IL-15, IL-17, IL-18, IL-21, GM- CSF, TNF, IFN-a, IFN- ⁇ , IFN- ⁇ , or FasL.
  • the treatment of a subject using a CXCL12-targeted immune response agent as described herein may further comprise administering one or more additional immuno therapeutic agents to the subject.
  • An immunotherapeutic agent may facilitate or enhance the targeting of cancer cells by the immune system, in particular T-cells, through the recognition of antigens expressed by the cancer cells.
  • Suitable agents include cancer vaccine preparations designed to induce T lymphocytes (T-cells) recognizing a localized region of an antigen or epitope specific to the tumor cell.
  • a cancer vaccine is an agent, a cell-based agent, molecule, or immunogen which stimulates or elicits an endogenous immune response in a subject or subject against one or more tumor antigens.
  • Suitable cancer vaccines are known in the art and may be produced by any convenient technique.
  • Cancer cells from the subject may be analyzed to identify a tumor antigen expressed by the cancer cells.
  • a method as described herein may comprise the step of identifying a tumor antigen which is displayed by one or more cancer cells in a sample obtained from the subject.
  • a cancer vaccine comprising one or more epitopes of the identified tumor antigen may then be administered to the subject whose cancer cells express the antigen.
  • the vaccine may induce or increase an immune response, preferably a T-cell mediated immune response, in the subject against the cancer cells expressing the identified tumor antigen.
  • the cancer vaccine may be administered before, at the same time, or after the CXCL12- targeted immune response agent is administered to the subject as described here.
  • Adoptive T-cell therapy involves the administration to a subject of tumor- specific T- cells to a subject.
  • the T-cells were previously isolated from the subject and expanded ex vivo.
  • Suitable adoptive T-cell therapies are well known in the art (J. Clin Invest. 2007 June 1 ; 117(6): 1466-1476.)
  • adoptive T-cell therapy using CAR T-cells would be greatly improved if used in combination with a CXCL12-targeted immune response agent.
  • CAR T-cells must migrate into a tumor to get in proximity to the cancer cells within the tumor in order to mediate their killing activity.
  • the present invention such as such as, for example, AMD3100, BMS-936564/MDX-1338, AMD11070, or LY2510924, used in combination with CAR T-cells may improve this type of immunotherapy.
  • the treatment of an individual using a CXCL12-targeted immune response agent may further comprise administering one or more tumor therapies to treat the cancerous tumor.
  • Such therapies include, for example, tumor medicaments, radiation and surgical procedures.
  • a tumor medicament is an agent which is administered to a subject for the purpose of treating a cancer. Suitable medicaments for the treatment of tumors are well known in the art.
  • Suitable medicaments for use in combination with CXCL12-targeted immune response agent as disclosed herein may include aspirin, sulindac, curcumin, alkylating agents including: nitrogen mustards, such as mechlor-ethamine, cyclophosphamide, ifosfamide, melphalan and chlorambucil; nitrosoureas, such as carmustine (BCNU), lomustine (CCNU), and semustine (methyl-CCNU); thylenimines/methylmelamine such as thriethylenemelamine (TEM), Methylene, thiophosphoramide (thiotepa), hexamethylmelamine (HMM, altretamine); alkyl sulfonates such as busulfan; triazines such as dacarbazine (DTIC); antimetabolites including folic acid analogs such as methotrexate and trimetrexate, pyrimidine analogs such as 5-fluorouracil, fluorodeoxyur
  • T-cell checkpoint antagonists like Lag-3, or inhibitors of ID01/ID02 (indoleamine 2,3-dioxygenase) could also be used in combination with the present invention. These enzymes catabolize tryptophan in the tumor microenvironment, which impairs T-cell function.
  • a CXCL12-targeted immune response agent such as for example, anti- CXCL12 antibodies, bispecific antibodies, or CAR T-cells
  • a T-cell checkpoint antagonist may synergistically increase cancer cell killing within a tumor.
  • compositions comprising the CXCL12-targeted immune response agent and optionally one or more other agents co-formulated or in admixture with each other and further discloses a kit or unit dose containing the CXCL12-targeted immune response agent.
  • such compositions, kits or doses further comprise one or more carriers in admixture with the agent or co-packaged for formulation prior to administration to an individual.
  • methods of treatment may comprise the administration of a CXCR4 signaling inhibitor in combination with a CXCL12-targeted immune response agent, as described above, for the treatment of cancer.
  • the CXCR4 signaling inhibitor and CXCL12-targeted immune response agent may be administered in the absence of the PD- 1 signaling inhibitor.
  • Suitable CXCL12-targeted immune response agents, immunotherapeutic agents and methods of treatment are described mutatis mutandis above.
  • Various embodiments are also disclosed above for combinations of a PD-1 signaling inhibitor and a CXCL12-targeted immune response agent. Aspects and embodiments of the invention relating to combinations of a PD-1 signaling inhibitor and a CXCL12-targeted immune response agent and optionally one or more other agents disclosed above include disclosure of the administration of the compounds or agents separately (sequentially or simultaneously) or in combination (co-formulated or mixed).
  • the specification further discloses a composition comprising the PD-1 signaling inhibitor and CXCL12-targeted immune response agent and optionally one or more other agents co-formulated or in admixture with each other and further discloses a kit or unit dose containing the PD-1 signaling inhibitor and CXCL12-targeted immune response agent packaged together, but not in admixture.
  • such compositions, kits or doses further comprise one or more carriers in admixture with one or both agents or co-packaged for formulation prior to administration to a subject.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

L'invention concerne un procédé permettant d'augmenter l'accumulation de lymphocytes T effecteurs dans des sites contenant des cellules cancéreuses d'une tumeur, consistant à administrer à un sujet qui en a besoin une quantité pharmaceutiquement efficace d'un agent de réponse immunitaire ciblant CXCL12. Dans certains modes de réalisation, l'agent de réponse immunitaire ciblant CXCL12 est administré avec une autre molécule qui inhibe ou détruit la cellule tumorale.
PCT/US2016/044254 2015-07-27 2016-07-27 Inhibition de cxcl12 dans l'immunothérapie anticancéreuse Ceased WO2017019767A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562197455P 2015-07-27 2015-07-27
US62/197,455 2015-07-27

Publications (1)

Publication Number Publication Date
WO2017019767A1 true WO2017019767A1 (fr) 2017-02-02

Family

ID=56611613

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/044254 Ceased WO2017019767A1 (fr) 2015-07-27 2016-07-27 Inhibition de cxcl12 dans l'immunothérapie anticancéreuse

Country Status (1)

Country Link
WO (1) WO2017019767A1 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
CN111065410A (zh) * 2017-09-06 2020-04-24 弗雷德哈钦森癌症研究中心 用于改善过继细胞疗法的方法
US10639368B2 (en) 2016-05-27 2020-05-05 Agenus Inc. Anti-TIM-3 antibodies and methods of use thereof
CN111315783A (zh) * 2017-10-18 2020-06-19 四十七公司 用抗cd47和抗pd-l1治疗卵巢癌
WO2020177733A1 (fr) * 2019-03-06 2020-09-10 江苏恒瑞医药股份有限公司 Protéine de fusion bifonctionnelle et son utilisation pharmaceutique
CN115068487A (zh) * 2021-03-11 2022-09-20 深圳埃格林医药有限公司 包含己酸羟孕酮的抗肿瘤联合制剂及其用途
US11547694B2 (en) 2017-03-23 2023-01-10 The General Hospital Corporation CXCR4/CXCR7 blockade and treatment of human papilloma virus-associated disease
WO2023128494A1 (fr) * 2021-12-27 2023-07-06 주식회사 에피바이오텍 Anticorps contre cxcl 12 et composition le comprenant pour le traitement de la chute des cheveux
US11802153B2 (en) 2017-10-18 2023-10-31 Forty Seven, Inc. Anti-CD47 agent-based ovarian cancer therapy
US12246067B2 (en) 2018-06-19 2025-03-11 Biontech Us Inc. Neoantigens and uses thereof

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
C. ZHONG ET AL: "Development and Preclinical Characterization of a Humanized Antibody Targeting CXCL12", CLINICAL CANCER RESEARCH, vol. 19, no. 16, 15 August 2013 (2013-08-15), US, pages 4433 - 4445, XP055260304, ISSN: 1078-0432, DOI: 10.1158/1078-0432.CCR-13-0943 *
ORIMO AKIRA ET AL: "Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion", CELL, CELL PRESS, US, vol. 121, no. 3, 6 May 2005 (2005-05-06), pages 335 - 348, XP009138583, ISSN: 0092-8674, [retrieved on 20050505], DOI: 10.1016/J.CELL.2005.02.034 *
PHILLIPS RODERICK J ET AL: "The stromal derived factor-1/CXCL12-CXC chemokine receptor 4 biological axis in non-small cell lung cancer metastases", AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, AMERICAN LUNG ASSOCIATION, NEW YORK, NY, US, vol. 167, no. 12, 15 June 2003 (2003-06-15), pages 1676 - 1686, XP002481676, ISSN: 1073-449X, DOI: 10.1164/RCCM.200301-071OC *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US12122833B2 (en) 2016-05-11 2024-10-22 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US10639368B2 (en) 2016-05-27 2020-05-05 Agenus Inc. Anti-TIM-3 antibodies and methods of use thereof
US12011481B2 (en) 2016-05-27 2024-06-18 Agenus Inc. Anti-TIM-3 antibodies and methods of use thereof
US10912828B2 (en) 2016-05-27 2021-02-09 Agenus Inc. Anti-TIM-3 antibodies and methods of use thereof
US11839653B2 (en) 2016-05-27 2023-12-12 Agenus Inc. Anti-TIM-3 antibodies and methods of use thereof
US11547694B2 (en) 2017-03-23 2023-01-10 The General Hospital Corporation CXCR4/CXCR7 blockade and treatment of human papilloma virus-associated disease
CN111065410A (zh) * 2017-09-06 2020-04-24 弗雷德哈钦森癌症研究中心 用于改善过继细胞疗法的方法
US11802153B2 (en) 2017-10-18 2023-10-31 Forty Seven, Inc. Anti-CD47 agent-based ovarian cancer therapy
CN111315783A (zh) * 2017-10-18 2020-06-19 四十七公司 用抗cd47和抗pd-l1治疗卵巢癌
US12246067B2 (en) 2018-06-19 2025-03-11 Biontech Us Inc. Neoantigens and uses thereof
WO2020177733A1 (fr) * 2019-03-06 2020-09-10 江苏恒瑞医药股份有限公司 Protéine de fusion bifonctionnelle et son utilisation pharmaceutique
CN115068487A (zh) * 2021-03-11 2022-09-20 深圳埃格林医药有限公司 包含己酸羟孕酮的抗肿瘤联合制剂及其用途
CN115068487B (zh) * 2021-03-11 2024-01-30 深圳埃格林医药有限公司 包含己酸羟孕酮的抗肿瘤联合制剂及其用途
WO2023128494A1 (fr) * 2021-12-27 2023-07-06 주식회사 에피바이오텍 Anticorps contre cxcl 12 et composition le comprenant pour le traitement de la chute des cheveux

Similar Documents

Publication Publication Date Title
WO2017019767A1 (fr) Inhibition de cxcl12 dans l'immunothérapie anticancéreuse
US20180228894A1 (en) Inhibition of cxcr4 signaling in cancer immunotherapy
US20220213207A9 (en) Immunomodulatory antibodies
JP7384949B2 (ja) 肺癌の処置のための抗pd-1抗体
TWI687227B (zh) 用於t細胞免疫療法之組合及其用途
WO2013083659A1 (fr) Traitement de combinaison comprenant un inhibiteur d'ho-1 et un agent immunothérapeutique
Engel-Riedel et al. A randomized, controlled trial evaluating the efficacy and safety of BTH1677 in combination with bevacizumab, carboplatin, and paclitaxel in first-line treatment of advanced non-small cell lung cancer
EP3941494A1 (fr) Compositions et méthodes de reprogrammation de tcr faisant appel à des protéines de fusion
JP6693873B2 (ja) Gd2陽性がんを処置するための製剤および方法
WO2016094309A1 (fr) Inhibition de la signalisation de tnf en immunothérapie du cancer
US20250032582A1 (en) Compositions and methods for cancer immunotherapy
KR20250155054A (ko) 흑색종 치료에서의 효능 강화를 위한 pd-1 억제제와 lag-3 억제제의 병용
KR20230002554A (ko) T 세포 요법
WO2016154341A1 (fr) Utilisations médicales d'agonistes de crtam
JP7526529B2 (ja) がん治療剤
RU2842292C1 (ru) Способы лечения рака посредством введения ингибитора pd-1
WO2024228167A1 (fr) Compositions liposomales de modulateur de cellules inkt et procédés d'utilisation
JP2024507866A (ja) Pd-1阻害剤を投与することにより肺がんを処置する方法
EA046291B1 (ru) Способы лечения злокачественного новообразования биспецифическими антителами против cd3xmuc16 и антителами против pd-1

Legal Events

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

Ref document number: 16748238

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16748238

Country of ref document: EP

Kind code of ref document: A1