Methods and compositions for treating cancer using P2RX2 inhibitors

Information

  • Patent Grant
  • 10457740
  • Patent Number
    10,457,740
  • Date Filed
    Monday, September 24, 2018
    5 years ago
  • Date Issued
    Tuesday, October 29, 2019
    4 years ago
Abstract
The present invention provides methods for treating cancer using P2RX2 inhibitors, such as P2RX2 inhibitory antibodies, among others. The invention also features compositions containing P2RX2 inhibitors, methods of diagnosing patients with P2RX2-associated cancer, and methods of predicting the response of cancer in a subject to treatment with P2RX2 inhibitors.
Description
BACKGROUND

Cancer is still one of the deadliest threats to human health. In 2012, there were 14 million new cases of cancer worldwide and 8.2 million cancer-related deaths. The number of new cancer cases is expected to rise to 22 million by 2030, and worldwide cancer deaths are projected to increase by 60%. Thus, there remains a need in the field for treatments for cancer.


SUMMARY OF THE INVENTION

The present invention provides methods for treating cancer using purinergic receptor P2X2 (P2RX2) inhibitors, such as P2RX2 inhibitory antibodies, among others. The invention also features compositions containing P2RX2 inhibitors, methods of diagnosing patients with P2RX2-associated cancer, and methods of predicting the response of cancer in a subject to treatment with P2RX2 inhibitors.


In a first aspect, the invention provides a method of treating a subject with cancer, by administering to the subject an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject with cancer by contacting a tumor, tumor microenvironment, site of metastasis, cancer cell, metastatic cancer cell, or stromal cell in a tumor microenvironment with an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject identified as having cancer by administering to the subject an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject identified as having cancer by contacting a tumor, tumor microenvironment, site of metastasis, cancer cell, metastatic cancer cell, or stromal cell in a tumor microenvironment with an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of reducing or inhibiting tumor growth by contacting the tumor with an effective amount of a P2RX2 inhibitor. In some embodiments, the tumor is a P2RX2-associated tumor. In some embodiments, the tumor is a pancreatic tumor.


In another aspect, the invention provides a method of reducing or inhibiting cancer cell proliferation by contacting the cancer cell with an effective amount of a P2RX2 inhibitor. In some embodiments, the cancer cell is a P2RX2-associated cancer cell. In some embodiments, the cancer cell is a pancreatic cancer cell.


In some embodiments of any of the above aspects, the cancer is P2RX2-associated cancer.


In another aspect, the invention provides a method of treating a subject with cancer by: a) identifying a subject with P2RX2-associated cancer; and b) administering to the subject an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject with cancer by: a) identifying a subject with P2RX2-associated cancer; and b) contacting a tumor, tumor microenvironment, site of metastasis, cancer cell, metastatic cancer cell, or stromal cell in a tumor microenvironment with an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject with P2RX2-associated cancer by administering to the subject an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject identified as having P2RX2-associated cancer by administering to the subject an effective amount of a P2RX2 inhibitor.


In another aspect, the invention provides a method of treating a subject with P2RX2-associated cancer by contacting a tumor, tumor microenvironment, site of metastasis, cancer cell, metastatic cancer cell, or stromal cell in a tumor microenvironment with an effective amount of a P2RX2 inhibitor.


In some embodiments of any of the above aspects, the method includes contacting a tumor with an effective amount of a P2RX2 inhibitor. In some embodiments of any of the above aspects, the method includes contacting a tumor microenvironment with an effective amount of a P2RX2 inhibitor. In some embodiments of any of the above aspects, the method includes contacting a site of metastasis with an effective amount of a P2RX2 inhibitor. In some embodiments of any of the above aspects, the method includes contacting a cancer cell with an effective amount of a P2RX2 inhibitor. In some embodiments of any of the above aspects, the method includes contacting a metastatic cancer cell with an effective amount of a P2RX2 inhibitor. In some embodiments of any of the above aspects, the method includes contacting a stromal cell in a tumor microenvironment with an effective amount of a P2RX2 inhibitor.


In some embodiments of any of the above aspects, the P2RX2-associated cancer expresses P2RX2. In some embodiments of any of the above aspects, the P2RX2-associated cancer overexpresses P2RX2.


In another aspect, the invention provides a method of predicting the response of a cancer in a subject to treatment with a P2RX2 inhibitor by contacting a cancer cell isolated from the subject with a P2RX2 inhibitor and evaluating the response of the cancer cell.


In some embodiments of the above aspect, the P2RX2 inhibitor is a P2RX2-specific inhibitor.


In some embodiments of the above aspect, the evaluating includes assessing cancer cell growth, cancer cell proliferation, cancer cell metastasis, cancer cell death, cancer cell migration, cancer cell invasion, cancer cell autophagy, or cancer cell P2RX2 expression.


In another aspect, the invention provides a method of predicting the response of a cancer in a subject to treatment with a P2RX2 inhibitor by: a) isolating a cancer cell from the subject; b) measuring the expression of P2RX2 in the cancer cell; and c) comparing P2RX2 expression in the cancer cell to a reference, wherein increased expression of P2RX2 in the cancer cell as compared to the reference indicates that the subject will respond to treatment with a P2RX2 inhibitor.


In another aspect, the invention provides a method of determining if a cancer cell expresses functional P2RX2 by contacting a cell with ATP and evaluating intracellular calcium levels. In some embodiments, and increase in intracellular calcium levels indicates that the cancer cell expresses functional P2RX2.


In some embodiments of any of the above aspects, the method further includes contacting the cancer cell with a P2RX2 inhibitor.


In another aspect, the invention provides a method of characterizing a cancer in a subject by: a) isolating a cancer cell from the subject; b) measuring the expression of P2RX2 in the cancer cell; and c) comparing P2RX2 expression in the cancer cell to a reference, wherein increased expression of P2RX2 in the cancer cell as compared to the reference indicates that the subject has P2RX2-associated cancer.


In another aspect, the invention provides a method of identifying a subject as having P2RX2-associated cancer by: a) isolating cancer cell from the subject; b) measuring the expression of P2RX2 in the cancer cell; and c) comparing P2RX2 expression in the cancer cell to a reference, wherein increased expression of P2RX2 in the cancer cell as compared to the reference indicates that the subject has P2RX2-associated cancer.


In some embodiments of any of the above aspects, the method further includes providing a P2RX2 inhibitor suitable for administration to the subject. In some embodiments of any of the above aspects, the method further includes administering to the subject an effective amount of a P2RX2 inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2-specific inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor is a P2RX2 function blocker.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor is a P2RX2 signaling inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor reduces P2RX2 expression or activity.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor reduces P2RX2 binding to a binding partner.


In some embodiments of any of the above aspects, the cancer is pancreatic cancer, melanoma, small cell lung cancer, non-small cell lung cancer, gastric cancer, colorectal cancer, head and neck cancer, ovarian cancer, testicular cancer, thymoma, uterine cancer, kidney cancer, acute myeloid leukemia, diffuse large B-cell lymphoma, prostate cancer, breast cancer, or hepatocellular carcinoma. In some embodiments, the cancer is pancreatic cancer.


In some embodiments of any of the above aspects, the cancer is P2RX2-associated cancer. In some embodiments, the P2RX2-associated cancer expresses P2RX2. In some embodiments, the P2RX2-associated cancer overexpresses P2RX2.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor is administered locally. In some embodiments, the P2RX2 inhibitor or P2RX2-specific inhibitor is administered intratumorally. In some embodiments, the P2RX2 inhibitor or P2RX2-specific inhibitor is administered to or near a site of metastasis. In some embodiments, the P2RX2 inhibitor or P2RX2-specific inhibitor is administered to or near a tumor microenvironment.


In some embodiments of any of the above aspects, the method further includes administering a second therapeutic agent.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor decreases tumor volume, tumor or cancer cell growth, decreases tumor innervation, decreases cancer cell proliferation, decreases cancer cell invasion, decreases cancer cell migration, decreases cancer cell metastasis, decreases tumor innervation, induces cancer cell autophagy, increases cancer cell death, increases time to recurrence, reduces cancer cell P2RX2 expression, or improves survival.


In some embodiments of any of the above aspects, the method further includes measuring one or more of tumor volume, tumor growth, tumor innervation, cancer cell proliferation, cancer cell invasion, cancer cell migration, cancer cell metastasis, cancer cell death, cancer cell autophagy, or P2RX2 expression before administration of the P2RX2 inhibitor or P2RX2-specific inhibitor.


In some embodiments of any of the above aspects, the method further includes measuring one or more of tumor volume, tumor or cancer cell growth, tumor innervation, cancer cell proliferation, cancer cell invasion, cancer cell metastasis, cancer cell death, cancer cell autophagy, or P2RX2 expression after administration of the P2RX2 inhibitor or P2RX2-specific inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor is administered in an amount sufficient to decrease tumor innervation, decrease nerve activity in a tumor, treat the cancer or tumor, cause remission, reduce tumor or cancer cell growth, reduce tumor volume, reduce tumor or cancer cell metastasis, reduce tumor or cancer cell invasion, reduce tumor or cancer cell proliferation, reduce tumor number, reduce tumor or cancer cell migration, reduce tumor P2RX2 expression, increase cancer cell death, induce cancer cell autophagy increase time to recurrence, or improve survival.


In some embodiments of any of the above aspects, the method further includes monitoring tumor or cancer progression (e.g., monitoring one or more of tumor volume, tumor or cancer cell growth, tumor innervation, tumor number, cancer cell proliferation, cancer cell invasion, cancer cell metastasis, cancer cell death, cancer cell autophagy, or P2RX2 expression) of after administration of the P2RX2 inhibitor or P2RX2-specific inhibitor.


In another aspect, the invention provides an anti-cancer therapy containing a P2RX2 inhibitor and a second agent selected from the group consisting of checkpoint inhibitors, chemotherapeutic agents, biologic cancer agents, anti-angiogenic drugs, drugs that target cancer metabolism, antibodies that mark a cancer cell surface for destruction, antibody-drug conjugates, cell therapies, commonly used anti-neoplastic agents, non-drug therapies, neurotransmission blockers, and neuronal growth factor blockers.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is an inhibitory RNA directed to P2RX2. In some embodiments of any of the above aspects, the P2RX2 inhibitor is an inhibitory RNA directed to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2 inhibitory antibody or an antigen binding fragment thereof. In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2-specific inhibitory antibody or an antigen binding fragment thereof. In some embodiments of any of the above aspects, the P2RX2 inhibitory antibody binds to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a small molecule inhibitor listed in Table 1. In some embodiments, the small molecule inhibitor is a P2RX2-specific inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2-specific inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2 function blocker.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2 signaling inhibitor listed in Table 2.


In another aspect, the invention provides a pharmaceutical containing a P2RX2 inhibitor.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2-specific inhibitor. In some embodiments, the P2RX2-specific inhibitor is an inhibitory RNA directed to P2RX2. In some embodiments, the P2RX2-specific inhibitor is a P2RX2-specific antibody or an antigen binding fragment thereof. In some embodiments, the P2RX2-specific inhibitor is a P2RX2-specific small molecule inhibitor listed in Table 1.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a P2RX2 inhibitory antibody or an antigen binding fragment thereof. In some embodiments, the P2RX2 inhibitory antibody is a P2RX2-specific inhibitory antibody or an antigen binding fragment thereof. In some embodiments, the P2RX2 inhibitory antibody binds to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is a small molecule inhibitor listed in Table 1.


In some embodiments of any of the above aspects, the P2RX2 inhibitor is an inhibitory RNA directed to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7.


In some embodiments of any of the above aspects, the P2RX2-specific inhibitory antibody exhibits one or more of the following activities: (a) disrupts cation channel flux; (b) disrupts extracellular ATP binding; (c) disrupts extracellular purinergic nucleotide binding; (d) sterically hinders binding of P2RX2 to a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7; (e) induces antibody-dependent cell killing of the P2RX2-expressing cell; (f) induces phagocytosis of the P2RX2-expressing cell; (g) induces opsonization of the P2RX2-expressing cell; (h) induces downregulation of P2RX2; (i) prevents formation of homotrimers or heterotrimers containing P2RX2; (j) does not have agonistic activity; (k) antagonizes P2RX2; (l) binds to residue V60 or G353 of P2RX2; or (m) binds to or blocks one or more glycosylation sites at residues 133, 194, and 310 of P2RX2.


In some embodiments of any of the above aspects, the P2RX2 inhibitory antibody binds to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7 and exhibits one or more of the following activities: (a) sterically hinders binding of the binding partner to P2RX2; (b) binds to the P2RX2-binding site of the binding partner; (c) induces antibody-dependent cell killing of the binding partner-expressing cell; (d) induces phagocytosis of the binding partner-expressing cell; (e) induces opsonization of the binding partner-expressing cell; (f) induces downregulation of binding partner; (g) prevents formation of multimers (e.g., dimers or trimers) containing the binding partner; (h) does not have agonistic activity; or (i) antagonizes the binding partner.


In some embodiments of the above aspects, the composition further includes a second therapeutic agent.


In some embodiments of any of the above aspects, the composition further includes a pharmaceutically acceptable excipient.


In some embodiments of any of the above aspects, the second therapeutic agent is an anti-cancer therapeutic, a P2RX2 signaling inhibitor, a P2RX2 function blocker, a neurotransmission blocker, or a neuronal growth factor blocker.


In some embodiments of any of the above aspects, the anti-cancer therapeutic is a checkpoint inhibitor, a chemotherapeutic agent, a biologic cancer agent, an anti-angiogenic drug, a drug that targets cancer metabolism, an antibody that marks a cancer cell surface for destruction, an antibody-drug conjugate, a cell therapy, a commonly used anti-neoplastic agent, or a non-drug therapy.


In some embodiments of any of the above aspects, the checkpoint inhibitor is an inhibitory antibody, a fusion protein, an agent that interacts with a checkpoint protein, an agent that interacts with the ligand of a checkpoint protein, an inhibitor of CTLA-4, an inhibitor of PD-1, an inhibitor of PDL1, an inhibitor of PDL2, or an inhibitor of B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAGS, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, or B-7 family ligands.


In some embodiments of any of the above aspects, the biologic cancer agent is an antibody listed in Table 3.


In some embodiments of any of the above aspects, the cancer is a cancer listed in column 1 of Table 4 and the second agent is a corresponding anti-cancer agent listed in column 2 of Table 4.


In some embodiments of any of the above aspects, the neurotransmission blocker is neurotoxin listed in Table 9, an antagonist of a neurotransmitter receptor listed in Table 5 or a neurotransmitter listed in Table 6, or a GABA re-uptake inhibitor, GABA analog, or GABA prodrug listed in Table 8. In some embodiments, the antagonist of a neurotransmitter receptor listed in Table 5 or a neurotransmitter listed in Table 6 is an antagonist listed in Tables 7A-7K.


In some embodiments of any of the above aspects, the neuronal growth factor blocker is an antagonist of a neuronal growth factor listed in Table 10. In some embodiments, the antagonist of a neuronal growth factor listed in Table 10 is an antibody listed in Table 11 or an antagonist listed in Table 12. In some embodiments, the antagonist of a neuronal growth factor listed in Table 10 is selected from the group consisting of etanercept, thalidomide, lenalidomide, pomalidomide, pentoxifylline, bupropion, DOI, disitertide, and trabedersen.


In some embodiments of any of the above aspects, the P2RX2 function blocker is a P2RX2-specific inhibitor. In some embodiments of any of the above aspects, the P2RX2 function blocker is a P2RX2-specific inhibitory antibody or an antigen binding fragment thereof. In some embodiments of any of the above aspects, the P2RX2 function blocker is an inhibitory RNA directed against P2RX2. In some embodiments of any of the above aspects, the P2RX2 function blocker is an inhibitory RNA directed against a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7. In some embodiments of any of the above aspects, the P2RX2 function blocker is P2RX2 inhibitory antibody that binds to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7. In some embodiments of any of the above aspects, the P2RX2 function blocker is a small molecule inhibitor listed in Table 1. In some embodiments, the small molecule inhibitor is a P2RX2-specific inhibitor.


In some embodiments of any of the above aspects, the P2RX2 signaling inhibitor is a small molecule inhibitor that disrupts downstream calcium signaling. In some embodiments, the P2RX2 signaling inhibitor is a small molecule inhibitor listed in Table 2.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor is selected from the group consisting of an antibody, a small molecule, a polypeptide, a DNA molecule, an RNA molecule, a gRNA, and a viral vector. In some embodiments, the antibody is a P2RX2 inhibitory antibody or an antigen binding fragment thereof. In some embodiments, the P2RX2 inhibitory antibody is a P2RX2-specific inhibitory antibody or an antigen binding fragment thereof. In some embodiments, the P2RX2 inhibitory antibody binds to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7. In some embodiments, the RNA molecule is an inhibitory RNA directed to P2RX2. In some embodiments, the RNA molecule is an inhibitory RNA directed to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7. In some embodiments, the small molecule is a small molecule inhibitor listed in Table 1 or a P2RX2 signaling inhibitor listed in Table 2.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor does not cross the blood brain barrier. In some embodiments, the P2RX2 inhibitor or the P2RX2-specific inhibitor has been modified to prevent blood brain barrier crossing by conjugation to a targeting moiety, formulation in a particulate delivery system, addition of a molecular adduct, or through modulation of its size, polarity, flexibility, or lipophilicity.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2-specific inhibitor does not have a direct effect on the central nervous system or gut.


In some embodiments of any of the above aspects, the P2RX2 inhibitor or P2RX2 specific inhibitor decreases tumor volume, decreases tumor or cancer cell growth, decreases tumor innervation, decreases cancer cell proliferation, decreases cancer cell invasion, decreases cancer cell migration, decreases cancer cell metastasis, increases cancer cell death, increases cancer cell autophagy, increases, time to recurrence, or improves survival.


Definitions

As used herein, “administration” refers to providing or giving a subject a therapeutic agent (e.g., a P2RX2 inhibitor), by any effective route. Exemplary routes of administration are described herein below.


As used herein, the term “agonist” refers to an agent (e.g., a small molecule or antibody) that increases receptor activity. An agonist may activate a receptor by directly binding to the receptor, by acting as a cofactor, by modulating receptor conformation (e.g., maintaining a receptor in an open or active state). An agonist may increase receptor activity by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more. An agonist may induce maximal receptor activation or partial activation depending on the concentration of the agonist and its mechanism of action.


As used herein, the term “analog” refers to a protein of similar nucleotide or amino acid composition or sequence to any of the proteins or peptides of the invention, allowing for variations that do not have an adverse effect on the ability of the protein or peptide to carry out its normal function (e.g., bind to a receptor or promote synapse formation). Analogs may be the same length, shorter, or longer than their corresponding protein or polypeptide. Analogs may have about 60% (e.g., about 60%, about 62%, about 64%, about 66%, about 68%, about 70%, about 72%, about 74%, about 76%, about 78%, about 80%, about 82%, about 84%, about 86%, about 88%, about 90%, about 92%, about 94%, about 96%, about 98%, or about 99%) identity to the amino acid sequence of the naturally occurring protein or peptide. An analog can be a naturally occurring protein or polypeptide sequence that is modified by deletion, addition, mutation, or substitution of one or more amino acid residues.


As used herein, the term “antagonist” refers to an agent (e.g., a small molecule or antibody) that reduces or inhibits receptor activity. An antagonist may reduce receptor activity by directly binding to the receptor, by blocking the receptor binding site, by modulating receptor conformation (e.g., maintaining a receptor in a closed or inactive state). An antagonist may reduce receptor activity by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more. An antagonist may also completely block or inhibit receptor activity. Antagonist activity may be concentration-dependent or -independent.


As used herein, the term “antibody” refers to a molecule that specifically binds to, or is immunologically reactive with, a particular antigen and includes at least the variable domain of a heavy chain, and normally includes at least the variable domains of a heavy chain and of a light chain of an immunoglobulin. Antibodies and antigen-binding fragments, variants, or derivatives thereof include, but are not limited to, polyclonal, monoclonal, multispecific, human, humanized, primatized, or chimeric antibodies, heteroconjugate antibodies (e.g., bi- tri- and quad-specific antibodies, diabodies, triabodies, and tetrabodies), single-domain antibodies (sdAb), epitope-binding fragments, e.g., Fab, Fab′ and F(ab′)2, Fd, Fvs, single-chain Fvs (scFv), rIgG, single-chain antibodies, disulfide-linked Fvs (sdFv), fragments including either a VL or VH domain, fragments produced by an Fab expression library, and anti-idiotypic (anti-Id) antibodies. Antibody molecules of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule. Moreover, unless otherwise indicated, the term “monoclonal antibody” (mAb) is meant to include both intact molecules as well as antibody fragments (such as, for example, Fab and F(ab′)2 fragments) that are capable of specifically binding to a target protein. Fab and F(ab′)2 fragments lack the Fc fragment of an intact antibody.


The term “antigen-binding fragment,” as used herein, refers to one or more fragments of an immunoglobulin that retain the ability to specifically bind to a target antigen. The antigen-binding function of an immunoglobulin can be performed by fragments of a full-length antibody. The antibody fragments can be a Fab, F(ab′)2, scFv, SMIP, diabody, a triabody, an affibody, a nanobody, an aptamer, or a domain antibody. Examples of binding fragments encompassed by the term “antigen-binding fragment” of an antibody include, but are not limited to: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL, and CH1 domains; (ii) a F(ab′)2 fragment, a bivalent fragment including two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb (Ward et al., Nature 341:544-546, 1989) including VH and VL domains; (vi) a dAb fragment that consists of a VH domain; (vii) a dAb that consists of a VH or a VL domain; (viii) an isolated complementarity determining region (CDR); and (ix) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv)). These antibody fragments can be obtained using conventional techniques known to those of skill in the art, and the fragments can be screened for utility in the same manner as intact antibodies. Antigen-binding fragments can be produced by recombinant DNA techniques, enzymatic or chemical cleavage of intact immunoglobulins, or, in certain cases, by chemical peptide synthesis procedures known in the art.


As used herein, the term “binding partner” refers to a polypeptide or fragment thereof that binds to a protein of interest (e.g., P2RX2). Binding partners include receptors and other molecules that selectively bind to the ligand of interest. Exemplary P2RX2 binding partners are P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7.


As used herein, the term “cell type” refers to a group of cells sharing a phenotype that is statistically separable based on gene expression data. For instance, cells of a common cell type may share similar structural and/or functional characteristics, such as similar gene activation patterns and antigen presentation profiles. Cells of a common cell type may include those that are isolated from a common tissue (e.g., epithelial tissue, neural tissue, connective tissue, or muscle tissue) and/or those that are isolated from a common organ, tissue system, blood vessel, or other structure and/or region in an organism.


As used herein, a “combination therapy” or “administered in combination” means that two (or more) different agents or treatments are administered to a subject as part of a defined treatment regimen for a particular disease or condition. The treatment regimen defines the doses and periodicity of administration of each agent such that the effects of the separate agents on the subject overlap. In some embodiments, the delivery of the two or more agents is simultaneous or concurrent and the agents may be co-formulated. In other embodiments, the two or more agents are not co-formulated and are administered in a sequential manner as part of a prescribed regimen. In some embodiments, administration of two or more agents or treatments in combination is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination may be administered orally.


As used herein, the terms “effective amount,” “therapeutically effective amount,” and a “sufficient amount” of a composition, antibody, vector construct, viral vector or cell described herein refer to a quantity sufficient to, when administered to a subject, including a mammal (e.g., a human), effect beneficial or desired results, including effects at the cellular level, tissue level, or clinical results, and, as such, an “effective amount” or synonym thereto depends upon the context in which it is being applied. For example, in the context of treating cancer it is an amount of the composition, antibody, vector construct, viral vector or cell sufficient to achieve a treatment response as compared to the response obtained without administration of the composition, antibody, vector construct, viral vector or cell. The amount of a given composition described herein that will correspond to such an amount will vary depending upon various factors, such as the given agent, the pharmaceutical formulation, the route of administration, the type of disease or disorder, the identity of the subject (e.g., age, sex, weight) or host being treated, and the like, but can nevertheless be routinely determined by one skilled in the art. Also, as used herein, a “therapeutically effective amount” of a composition, antibody, vector construct, viral vector or cell of the present disclosure is an amount that results in a beneficial or desired result in a subject as compared to a control. As defined herein, a therapeutically effective amount of a composition, antibody, vector construct, viral vector or cell of the present disclosure may be readily determined by one of ordinary skill by routine methods known in the art. Dosage regimen may be adjusted to provide the optimum therapeutic response.


As used herein, the terms “increasing” and “decreasing” refer to modulating resulting in, respectively, greater or lesser amounts, of function, expression, or activity of a metric relative to a reference. For example, subsequent to administration of a P2RX2 inhibitor in a method described herein, the amount of a marker of a metric (e.g., cancer cell death) as described herein may be increased or decreased in a subject by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 98% or more relative to the amount of the marker prior to administration. Generally, the metric is measured subsequent to administration at a time that the administration has had the recited effect, e.g., at least one week, one month, 3 months, or 6 months, after a treatment regimen has begun.


As used herein, the term “innervated” refers to a tissue (e.g., a tumor) that contains nerves. “Innervation” refers to the process of nerves entering a tissue.


As used herein, “locally” or “local administration” means administration at a particular site of the body intended for a local effect and not a systemic effect. Examples of local administration are epicutaneous, inhalational, intra-articular, intrathecal, intravaginal, intravitreal, intrauterine, intra-lesional administration, lymph node administration, intratumoral administration and administration to a mucous membrane of the subject, wherein the administration is intended to have a local and not a systemic effect.


As used herein, the term “percent (%) sequence identity” refers to the percentage of amino acid (or nucleic acid) residues of a candidate sequence that are identical to the amino acid (or nucleic acid) residues of a reference sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity (e.g., gaps can be introduced in one or both of the candidate and reference sequences for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). Alignment for purposes of determining percent sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software, such as BLAST, ALIGN, or Megalign (DNASTAR) software. Those skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full length of the sequences being compared. For example, a reference sequence aligned for comparison with a candidate sequence may show that the candidate sequence exhibits from 50% to 100% sequence identity across the full length of the candidate sequence or a selected portion of contiguous amino acid (or nucleic acid) residues of the candidate sequence. The length of the candidate sequence aligned for comparison purposes may be, for example, at least 30%, (e.g., 30%, 40, 50%, 60%, 70%, 80%, 90%, or 100%) of the length of the reference sequence. When a position in the candidate sequence is occupied by the same amino acid residue as the corresponding position in the reference sequence, then the molecules are identical at that position.


As used herein, a “pharmaceutical composition” or “pharmaceutical preparation” is a composition or preparation having pharmacological activity or other direct effect in the mitigation, treatment, or prevention of disease, and/or a finished dosage form or formulation thereof and which is indicated for human use.


As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, compositions and/or dosage forms, which are suitable for contact with the tissues of a subject, such as a mammal (e.g., a human) without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.


As used herein, the term “proliferation” refers to an increase in cell numbers through growth and division of cells.


As used herein, the term “reference” refers to a level, expression level, copy number, sample or standard that is used for comparison purposes. For example, a reference sample can be obtained from a healthy individual (e.g., an individual who does not have cancer). A reference level can be the level of expression of one or more reference samples. For example, an average expression (e.g., a mean expression or median expression) among a plurality of individuals (e.g., healthy individuals, or individuals who do not have cancer). In other instances, a reference level can be a predetermined threshold level, e.g., based on functional expression as otherwise determined, e.g., by empirical assays.


As used herein, the term “sample” refers to a specimen (e.g., blood, blood component (e.g., serum or plasma), urine, saliva, amniotic fluid, cerebrospinal fluid, tissue (e.g., placental or dermal), pancreatic fluid, chorionic villus sample, and cells) isolated from a subject.


As used herein, the terms “subject” and “patient” refer to an animal (e.g., a mammal, such as a human). A subject to be treated according to the methods described herein may be one who has been diagnosed with a particular condition, or one at risk of developing such conditions. Diagnosis may be performed by any method or technique known in the art. One skilled in the art will understand that a subject to be treated according to the present disclosure may have been subjected to standard tests or may have been identified, without examination, as one at risk due to the presence of one or more risk factors associated with the disease or condition.


“Treatment” and “treating,” as used herein, refer to the medical management of a subject with the intent to improve, ameliorate, stabilize (i.e., not worsen), prevent or cure a disease, pathological condition, or disorder. This term includes active treatment (treatment directed to improve the disease, pathological condition, or disorder), causal treatment (treatment directed to the cause of the associated disease, pathological condition, or disorder), palliative treatment (treatment designed for the relief of symptoms), preventative treatment (treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder); and supportive treatment (treatment employed to supplement another therapy). Treatment also includes diminishment of the extent of the disease or condition; preventing spread of the disease or condition; delay or slowing the progress of the disease or condition; amelioration or palliation of the disease or condition; and remission (whether partial or total), whether detectable or undetectable. “Ameliorating” or “palliating” a disease or condition means that the extent and/or undesirable clinical manifestations of the disease, disorder, or condition are lessened and/or time course of the progression is slowed or lengthened, as compared to the extent or time course in the absence of treatment. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment. Those in need of treatment include those already with the condition or disorder, as well as those prone to have the condition or disorder or those in which the condition or disorder is to be prevented.


As used herein, the term “overexpressed” refers to a nucleic acid or polypeptide that is expressed or caused to be expressed or produced in a cell at a greater level than is normally expressed in the corresponding wild-type cell. For example, P2RX2 is “overexpressed” in a cancer cell when P2RX2 is present at a higher level in the cancer cell compared to the level in a non-cancerous cell of the same tissue or cell type from the same species or individual. P2RX2 is overexpressed when P2RX2 expression is increased by 1.1-fold or more (e.g., 1.1, 1.2, 1.3, 1.4, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0-fold or more) compared to a reference (e.g., a non-cancerous cell of the same type).


As used herein, the term “cancer” refers to a condition characterized by unregulated or abnormal cell growth. The terms “cancer cell,” “tumor cell,” and “tumor” refer to an abnormal cell, mass, or population of cells that result from excessive division that may be malignant or benign and all pre-cancerous and cancerous cells and tissues.


As used herein, the term “P2RX2-associated cancer” refers to a cancer in which P2RX2 is expressed (e.g., a cancer in which P2RX2 is overexpressed compared to a reference (e.g., a non-cancerous cell of the same type), or a cancer in which P2RX2 is mutated (e.g., a cancer carrying an activating P2RX2 mutation, e.g., a cancer expressing hyperactive P2RX2)). P2RX2-associated cancers can be identified by assessing a cancer cell or tumor sample for P2RX2 gene or protein expression and comparing it to P2RX2 gene or protein expression in a reference cell.


The term “P2RX2 inhibitory antibody” refers to antibodies that are capable of binding to P2RX2 or a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7, and inhibiting or reducing P2RX2 function and/or attenuating one or more signal transduction pathways mediated by P2RX2. For example, P2RX2 inhibitory antibodies may disrupt cation channel flux or extracellular ATP binding, prevent P2RX2 from binding to P2RX3 or other P2RX2 binding partners, or block the formation of homotrimers or heterotrimers containing P2RX2. The term “P2RX2-specific inhibitory antibody” refers to antibodies that bind specifically to P2RX2 (e.g., antibodies that do not bind to other P2RX family members) and inhibit or reduce P2RX2 function and/or attenuate one or more signal transduction pathways mediated by P2RX2. P2RX2 inhibitory antibodies and P2RX2-specific inhibitory antibodies inhibit or reduce P2RX2 function and/or attenuate one or more P2RX2-mediated signal transduction pathways by at least 10% (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more).


As used herein, the term “P2RX2 inhibitor” refers to an agent that inhibits or reduces P2RX2 function or signaling. P2RX2 inhibitors include P2RX2 inhibitory antibodies, small molecules, or inhibitory RNAs that reduce or inhibit P2RX2 expression, P2RX2 binding, P2RX2 function, or signal transduction downstream of P2RX2. P2RX2 inhibitors reduce P2RX2 function or signaling by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more).


As used herein, the term “P2RX2-specific inhibitor” refers to a P2RX2 inhibitor that selectively inhibits or reduces P2RX2 function or signaling without substantially affecting the function or signaling of any other protein (e.g., without substantially affecting the function or signaling of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7). P2RX2-specific inhibitors include P2RX2-specific inhibitory antibodies, inhibitory RNAs directed to P2RX2, and P2RX2-specific small molecule inhibitors. P2RX2-specific inhibitors reduce P2RX2 function or signaling by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more).


As used herein, the term “P2RX2 function blocker” refers to a type of P2RX2 inhibitor that reduces or inhibits P2RX2 function by reducing the expression of P2RX2 or preventing P2RX2 from interacting with one or more of its binding partners (e.g., other P2RX family members). Exemplary P2RX2 function blockers include antibodies that bind to the extracellular domain of P2RX2, inhibitory RNAs directed to P2RX2 or a P2RX2 binding partner, P2RX2 small molecule inhibitors, and antibodies that bind to one or more P2RX2 binding partners (e.g., antibodies that bind to the extracellular domains of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7 and block their interaction with P2RX2). P2RX2 function blockers reduce P2RX2 function by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more).


As used herein, the term “P2RX2 signaling inhibitor” refers to a type of P2RX2 inhibitor that reduces or inhibits the intracellular signaling that is downstream of P2RX2 activation or interaction with a binding partner. Exemplary P2RX2 signaling inhibitors include small molecules that inhibit intracellular calcium signaling. P2RX2 signaling inhibitors reduce downstream signaling by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more).


As used herein, an agent that “does not cross the blood brain barrier” is an agent that does not significantly cross the barrier between the peripheral circulation and the brain and spinal cord. This can also be referred to as a “blood brain barrier impermeable” agent. Agents will have a limited ability to cross the blood brain barrier if they are not lipid soluble or have a molecular weight of over 600 Daltons.


Agents that typically cross the blood brain barrier can be modified to become blood brain barrier impermeable based on chemical modifications that increase the size or alter the hydrophobicity of the agent, packaging modifications that reduce diffusion (e.g., packaging an agent within a microparticle or nanoparticle), and conjugation to biologics that direct the agent away from the blood brain barrier (e.g., conjugation to a pancreas-specific antibody). An agent that does not cross the blood brain barrier is an agent for which 30% or less (e.g., 30%, 25%, 20%, 15%, 10%, 5%, 2% or less) of the administered agent crosses the blood brain barrier.


As used herein, an agent that “does not have a direct effect on the central nervous system (CNS) or gut” is an agent that does not directly alter neurotransmission, neuronal numbers, or neuronal morphology in the CNS or gut when administered according to the methods described herein. This may be assessed by administering the agents to animal models and performing electrophysiological recordings or immunohistochemical analysis. An agent will be considered not to have a direct effect on the CNS or gut if administration according to the methods described herein has an effect on neurotransmission, neuronal numbers, or neuronal morphology in the CNS or gut that is 50% or less (e.g., 50%, 45%, 40%, 35%, 30%, 25%, 20%, 15%, 10%, 5%, or less) of the effect observed if the same agent is administered directly to the CNS or gut.


As used herein, the term “neuronal growth factor blocker” refers to an agent that decreases or inhibits neuronal growth, development, or survival. Neuronal growth factors include proteins that promote neurogenesis, neuronal growth, and neuronal differentiation (e.g., neurotrophic factors NGF, NT3, BDNF, CNTF, and GDNF), proteins that promote neurite outgrowth (e.g., axon or dendrite outgrowth or stabilization), or proteins that promote synapse formation (e.g., synaptogenesis, synapse assembly, synaptic adhesion, synaptic maturation, synaptic refinement, or synaptic stabilization). These processes lead to innervation of tissue, including neural tissue, muscle, and tumors, and the formation of synaptic connections between two or more neurons and between neurons and non-neural cells (e.g., tumor cells). A neuronal growth factor blocker reduces or inhibits one or more of these processes (e.g., through the use of antibodies that block neuronal growth factors or their receptors). Exemplary neuronal growth factors are listed in Table 10. Neuronal growth factor blockers decrease or inhibit neurite outgrowth, innervation, synapse formation, or any of the aforementioned processes by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more).


As used herein, the term “neurotransmission blocker” refers to an agent that decreases or blocks neurotransmission. Neurotransmission blockers can decrease neurotransmission by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more. Exemplary neurotransmitters and neurotransmitter receptors are listed in Tables 5 and 6. Neurotransmission blockers may decrease neurotransmission by decreasing neurotransmitter synthesis or release, increasing neurotransmitter reuptake or degradation, decreasing neurotransmitter receptor activity, decreasing neurotransmitter receptor synthesis or membrane insertion, increasing neurotransmitter degradation, regulating neurotransmitter receptor conformation, and disrupting the pre- or postsynaptic machinery. Neurotransmission blockers include antibodies that bind to or block the function of neurotransmitters, neurotransmitter receptor antagonists, inhibitory RNAs directed to neurotransmitter receptors, and toxins that disrupt synaptic release.







DETAILED DESCRIPTION

Described herein are compositions and methods for the treatment of cancer in a subject (e.g., a mammalian subject, such as a human) by administering purinergic receptor P2X2 (P2RX2) inhibitors. P2RX2 inhibitors include inhibitors specific to P2RX2 (e.g., anti-P2RX2-specific inhibitory antibodies) and non-specific inhibitors that could potentially affect other proteins due to their having shared binding partners or signaling pathways with P2RX2. These methods and compositions provide new mechanistic approaches for treating cancer.


P2RX2


P2RX2 (Entrez Gene ID 22953) is a transmembrane purinergic receptor gated by extracellular ATP. This ligand-gated ion channel is a non-selective cation channel that is involved in a number of neuronal functions, such as synaptic transmission between neurons and from neurons to smooth muscle, neuromuscular junction formation, hearing, and taste perception. P2RX2 forms a trimer, which can be composed of three P2RX2 molecules (a homotrimer) or P2RX2 and two other P2RX receptors (a heterotrimer). P2X receptors are expressed by neurons and glial cells throughout the central and peripheral nervous system.


The present invention relates to the discovery that loss of P2RX2 in pancreatic cancer cell lines prevented tumor growth when P2RX2 knockout cancer cells were implanted in mice. These findings indicate that inhibition of P2RX2 can be used as a therapeutic strategy for treating pancreatic cancer and other cancers. These data also suggest that patients with overexpression of P2RX2 are at increased risk of developing cancer and would benefit from specific treatments, such as treatment with the compositions and methods described herein.


P2RX2 Inhibitors


P2RX2 inhibitors described herein can reduce or inhibit P2RX2 function or signaling in order to treat cancer. P2RX2 inhibitors can be grouped into categories based on their mechanism of action and their effect on P2RX2: 1) P2RX2-specific inhibitors (e.g., inhibitors that only disrupt P2RX2 function or signaling, such as P2RX2-specific inhibitory antibodies, P2RX2-specific small molecule inhibitors, or inhibitory RNA directed to P2RX2), 2) P2RX2 function blockers (e.g., inhibitors that prevent P2RX2 from binding to a binding partner, forming trimers, or carrying out other processes necessary for normal P2RX2 activity, e.g., P2RX2 inhibitory antibodies, such as anti-P2X family member (e.g., P2RX1, 3, 4, 5, 6, or 7) antibodies, and 3) P2RX2 signaling inhibitors (e.g., inhibitors that disrupt downstream signaling pathways or intracellular events that occur after activation of P2RX2).


P2RX2-Specific Inhibitors


In some embodiments, the P2RX2 inhibitor is a P2RX2-specific inhibitor. P2RX2-specific inhibitors selectively reduce or inhibit P2RX2 function, expression, or signaling without directly affecting other proteins. P2RX2-specific inhibitors include P2RX2-specific inhibitory antibodies or antigen binding fragments thereof, inhibitory RNAs directed to P2RX2, and small molecules that specifically bind to and inhibit P2RX2 (e.g., P2RX2-specific small molecule inhibitors listed in Table 1). P2RX2-specific inhibitors can reduce P2RX2 function, expression, or signaling by 5% or more (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99%, or more).


In some embodiments, the P2RX2 antibody is a P2RX2-specific inhibitory antibody or an antigen binding fragment thereof that binds to P2RX2 and reduces or inhibits P2RX2 function. P2RX2-specific inhibitory antibodies include antibodies having one or more of the following functional properties: prevent P2RX2 from binding to a binding partner (e.g., sterically hinder the binding of P2RX2 to a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7); reduce or inhibit cation channel flux; reduce or inhibit extracellular ATP binding; reduce or inhibit extracellular purinergic nucleotide binding; do not have agonistic activity (e.g., do not activate P2RX2); induce antibody-dependent cell killing of the cell expressing P2RX2 (e.g., antibody-dependent cell killing by Natural Killer (NK) cells, monocytes, macrophages, neutrophils, dendritic cells, or eosinophils); induce phagocytosis of the cell expressing P2RX2 (e.g., macrophage phagocytosis of the cancer cell); induce opsonization of the cell expressing P2RX2; prevent the formation of homotrimers or heterotrimers containing P2RX2 (e.g., the antibody is a monovalent antibody or an antibody with a single heavy chain); induce downregulation of P2RX2 on the cell surface (e.g., hyper-crosslink or cluster P2RX2 to induce internalization and degradation, e.g., the antibody is a polyvalent antibody); or antagonize P2RX2. In some embodiments, P2RX2 inhibitory antibodies have one or more of the following properties: bind to residue V60 or G353 of P2RX2, or bind to or block one or more glycosylation sites at residues 133, 194, and 310 of P2RX2. Antibodies having one or more of these functional properties are routinely screened and selected once the desired functional property is identified herein (e.g., by screening of phage display or other antibody libraries).


In some embodiments, the P2RX2-specific inhibitor is an inhibitory RNA directed to P2RX2. In some embodiments, the P2RX2 inhibitor is a small molecule inhibitor (e.g., antagonist) that is selective for P2RX2. P2RX2-specific small molecule inhibitors for use in the compositions and methods described herein are listed in Table 1.


P2RX2 Function Blockers


In some embodiments, the P2RX2 inhibitor is a P2RX2 function blocker. P2RX2 function blockers reduce or inhibit P2RX2 function by reducing P2RX2 expression, preventing P2RX2 from interacting with its binding partners (e.g., P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, or P2RX7), or preventing P2RX2 from becoming activated. P2RX2 function blockers include P2RX2-specific inhibitors that reduce or inhibit P2RX2 function or expression (e.g., P2RX2-specific inhibitory antibodies or antigen binding fragments thereof, inhibitory RNAs directed against P2RX2, small molecule inhibitors that are specific for P2RX2), P2RX2 inhibitory antibodies that bind to P2RX2 binding partners or antigen binding fragments thereof (e.g., antibodies that bind to P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, or P2RX7 and block their interaction with P2RX2), inhibitory RNAs directed to P2RX2 binding partners, and small molecule inhibitors that non-specifically reduce or inhibit the function of P2RX2 (e.g., small molecule inhibitors of the P2X receptor family).


P2RX2 binding partners include P2RX1 (Entrez Gene ID 5023), P2RX3 (Entrez Gene ID 5024), P2RX4 (Entrez Gene ID 5025), P2RX5 (Entrez Gene ID 5026), P2RX6 (Entrez Gene ID 9127), and P2RX7 (Entrez Gene ID 5027).


In some embodiments, P2RX2 inhibitory antibodies that bind to P2RX2 binding partners are antibodies that bind to P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, or P2RX7. In some embodiments, P2RX2 inhibitory antibodies that bind to P2RX2 binding partners have one or more of the following functional properties: sterically hinders the binding partner from binding to P2RX2 (e.g., blocks the interaction between P2RX2 and P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, or P2RX7); binds to the P2RX2 binding site on the binding partner; antagonizes the binding partner (e.g., prevents binding partner signaling or prevents formation of multimers (e.g., dimers or trimers) containing the binding partner or containing the binding partner and P2RX2 (e.g., the antibody is monovalent and can only bind to one binding partner and cannot induce multimerization); induces binding partner downregulation on the cell surface (e.g., hyper-crosslinks the binding partner to induce internalization, e.g., the antibody is a polyvalent antibody); induces antibody-dependent cell killing of the binding partner-expressing cell (e.g., antibody-dependent cell killing by NK cells, monocytes, macrophages, neutrophils, dendritic cells, or eosinophils); induces phagocytosis of the binding partner-expressing cell (e.g., macrophage phagocytosis of the cancer cell); induces opsonization of the binding partner-expressing cell; or does not have agonistic activity (e.g., does not activate the binding partner). Antibodies having one or more of these functional properties are routinely screened and selected once the desired functional property is identified herein (e.g., by screening of phage display or other antibody libraries).


In some embodiments, the P2RX2 function blocker is an inhibitory RNA directed to a P2RX2 binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7. In some embodiments, the P2RX2 function blocker is a small molecule signaling inhibitor listed in Table 1 (e.g. a P2RX2/P2RX3 heterotrimer inhibitor).


P2RX2 Signaling Inhibitors


In some embodiments, the P2RX2 inhibitor is a P2RX2 signaling inhibitor. P2RX2 signaling inhibitors include agents that reduce or inhibit signaling that occurs downstream of P2RX2 activation or binding to a binding partner, such as small molecule inhibitors of intracellular signaling cascades. P2RX2 signaling inhibitors include small molecules that disrupt intracellular calcium signaling (e.g., Ned K, A23187, BAPTA, or Dantrolene). Small molecule P2RX2 signaling inhibitors for use in the methods and compositions described herein are listed in Table 2.









TABLE 1







SMALL MOLECULE INHIBITORS OF P2RX2











Type of












Inhibitor
Inhibitors














P2RX2-specific
CHEMBL494161
CHEMBL119416
CHEMBL604158
CHEMBL1672098


inhibitors (IC50
CHEMBL495204
CHEMBL499580
CHEMBL598857
CHEMBL1671997


<10 μM)
CHEMBL523173
CHEMBL1672107
CHEMBL597820
CHEMBL1671996



CHEMBL492300
CHEMBL523043
CHEMBL597591
CHEMBL1671993



CHEMBL494159
CHEMBL521983
CHEMBL597203
CHEMBL1671992



CHEMBL494353
CHEMBL500550
CHEMBL596982
CHEMBL134193



CHEMBL494160
CHEMBL492299
CHEMBL524284
CHEMBL133576



CHEMBL494158
CHEMBL504607
CHEMBL524064
CHEMBL131271



CHEMBL526307
CHEMBL494176
CHEMBL522725
CHEMBL118007



CHEMBL492934
CHEMBL493547
CHEMBL522053
CHEMBL116926



CHEMBL492933
CHEMBL493546
CHEMBL521709
CHEMBL492729



CHEMBL494582
CHEMBL446310
CHEMBL499428
CHEMBL521820



CHEMBL492907
CHEMBL69727
CHEMBL498038
CHEMBL494940



CHEMBL492703
CHEMBL331358
CHEMBL496229
CHEMBL492789



CHEMBL1672104
CHEMBL494833
CHEMBL496022
CHEMBL69234



CHEMBL495203
CHEMBL509572
CHEMBL495834
CHEMBL401735



CHEMBL1672105
CHEMBL496030
CHEMBL495796
CHEMBL494834



CHEMBL448525
CHEMBL1671995
CHEMBL450832
CHEMBL494832



CHEMBL271672
CHEMBL523000
CHEMBL404659
CHEMBL494772



CHEMBL496401
CHEMBL492968
CHEMBL404450
CHEMBL494181



CHEMBL413145
CHEMBL271688
CHEMBL403051
CHEMBL257495



CHEMBL119180
CHEMBL494581
CHEMBL402239
CHEMBL117766



CHEMBL502618
CHEMBL445413
CHEMBL256864
CHEMBL495195



CHEMBL444469
CHEMBL331250
CHEMBL256688
CHEMBL493740



CHEMBL1672106
CHEMBL492967
CHEMBL256057
CHEMBL492562



CHEMBL493741
CHEMBL492744
CHEMBL1672103
CHEMBL477339



CHEMBL443930
CHEMBL606414
CHEMBL1672102
CHEMBL265502



CHEMBL492935
CHEMBL604300
CHEMBL1672099
CHEMBL522184



CHEMBL492745





P2RX2/P2RX3
CHEMBL3717153
CHEMBL3729278
CHEMBL3732185



heterotrimer
CHEMBL3731399
CHEMBL3730477
CHEMBL3728501
CHEMBL3728833


inhibitors
CHEMBL3728821
CHEMBL3730649
CHEMBL3729987
CHEMBL3727542



CHEMBL3732843
CHEMBL3733102
CHEMBL3729871
CHEMBL3730896



CHEMBL3730913
CHEMBL3729724
CHEMBL3732208
CHEMBL3731357



CHEMBL3731410
CHEMBL3731231
CHEMBL3728453
CHEMBL3731248



CHEMBL3729264
CHEMBL3730174
CHEMBL3732421
CHEMBL3731731



CHEMBL3732909
CHEMBL3731419
CHEMBL3731986
CHEMBL3730650



CHEMBL3727382
CHEMBL3729891
CHEMBL3728126
CHEMBL3728033



CHEMBL3732309
CHEMBL3730205
CHEMBL3732353
CHEMBL3728818



CHEMBL3731936
CHEMBL3730868
CHEMBL3732671
CHEMBL3730941



CHEMBL3731366
CHEMBL3729489
CHEMBL3731191
CHEMBL3727639



CHEMBL3731764
CHEMBL3732409
CHEMBL3727749
CHEMBL3729851



CHEMBL3717395
CHEMBL3729478
CHEMBL3730212
CHEMBL3731921



CHEMBL3728949
CHEMBL3731744
CHEMBL3730748
CHEMBL3729135



CHEMBL3733189
CHEMBL3731305
CHEMBL3728853
CHEMBL3732239



CHEMBL3732959
CHEMBL3732414
CHEMBL3728157
CHEMBL3733326



CHEMBL3730584
CHEMBL3732154
CHEMBL3732341
CHEMBL3729654



CHEMBL3731980
CHEMBL3733209
CHEMBL3731610
CHEMBL3731805



CHEMBL3733173
CHEMBL3732768
CHEMBL3728519
CHEMBL3731729



CHEMBL3728301
CHEMBL3727897
CHEMBL3728605
CHEMBL3730253



CHEMBL3718959
CHEMBL3729611
CHEMBL3732062
CHEMBL3730371



CHEMBL3732382
CHEMBL3727954
CHEMBL3729325
CHEMBL3731465
















TABLE 2







SMALL MOLECULE INHIBITORS OF CALCIUM SIGNALING








Signaling Pathway
Inhibitors





Calcium signaling
Ned K, A23187, BAPTA, Dantrolene,



DHBP dibromide, EGTA, ionomycin, MDL



12330A, MRS 1845, Ned 19 (cis- or trans-),



NPC 15199, ruthenium red, ryanodine,



SKF 96365, (-)-Xestospongin C, YM 58483









Agent Modalities


A P2RX2 inhibitor can be selected from a number of different modalities. A P2RX2 inhibitor can be a nucleic acid molecule (e.g., DNA molecule or RNA molecule, e.g., mRNA or inhibitory RNA molecule (e.g., siRNA, shRNA, or miRNA), or a hybrid DNA-RNA molecule), a small molecule (e.g., a small molecule P2RX2 inhibitor, an inhibitor of a signaling cascade (e.g., calcium signaling), or an epigenetic modifier), or a polypeptide (e.g., an antibody molecule, e.g., an antibody or antigen binding fragment thereof). A P2RX2 inhibitor can also be a viral vector expressing a P2RX2 inhibitor or a cell infected with a viral vector. Any of these modalities can be a P2RX2 inhibitor directed to target (e.g., to reduce or inhibit) P2RX2 function, P2RX2 expression, P2RX2 binding, or P2RX2 signaling.


The nucleic acid molecule, small molecule, peptide, polypeptide, or antibody molecule can be modified. For example, the modification can be a chemical modification, e.g., conjugation to a marker, e.g., fluorescent marker or a radioactive marker. In other examples, the modification can include conjugation to a molecule that enhances the stability or half-life of the P2RX2 inhibitor (e.g., an Fc domain of an antibody or serum albumin, e.g., human serum albumin). The modification can also include conjugation to an antibody to target the agent to a particular cell or tissue. Additionally, the modification can be a chemical modification, packaging modification (e.g., packaging within a nanoparticle or microparticle), or targeting modification to prevent the agent from crossing the blood brain barrier.


Small Molecules


Numerous small molecule P2RX2 inhibitors useful in the methods of the invention are described herein in Tables 1 and 2 and additional small molecule P2RX2 inhibitors useful as therapies for cancer can also be identified through screening based on their ability to reduce or inhibit P2RX2 function or signaling. Small molecules include, but are not limited to, small peptides, peptidomimetics (e.g., peptoids), amino acids, amino acid analogs, synthetic polynucleotides, polynucleotide analogs, nucleotides, nucleotide analogs, organic and inorganic compounds (including heterorganic and organometallic compounds) generally having a molecular weight less than about 5,000 grams per mole, e.g., organic or inorganic compounds having a molecular weight less than about 2,000 grams per mole, e.g., organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, e.g., organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other pharmaceutically acceptable forms of such compounds.


In some embodiments, the P2RX2 inhibitor is a small molecule inhibitor of P2RX2 (e.g., a small molecule inhibitor listed in Table 1), or a P2RX2 binding partner antagonist (e.g., an antagonist of a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7). P2RX2 inhibitors can be used to treat a disorder or condition described herein. A pharmaceutical composition including the P2RX2 inhibitor can be formulated for treatment of a cancer described herein. In some embodiments, a pharmaceutical composition that includes the P2RX2 inhibitor is formulated for local administration, e.g., to the affected site in a subject.


Antibodies


The P2RX2 inhibitor can be an antibody or antigen binding fragment thereof. For example, a P2RX2 inhibitor described herein is an antibody that reduces or blocks the activity and/or function of P2RX2 through binding to P2RX2 or a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7) to block the binding between P2RX2 and a binding partner or to block P2RX2 multimerization.


The making and use of therapeutic antibodies against a target antigen (e.g., P2RX2 or a P2RX2 binding partner) is known in the art. See, for example, the references cited herein above, as well as Zhiqiang An (Editor), Therapeutic Monoclonal Antibodies: From Bench to Clinic. 1st Edition. Wiley 2009, and also Greenfield (Ed.), Antibodies: A Laboratory Manual. (Second edition) Cold Spring Harbor Laboratory Press 2013, for methods of making recombinant antibodies, including antibody engineering, use of degenerate oligonucleotides, 5′-RACE, phage display, and mutagenesis; antibody testing and characterization; antibody pharmacokinetics and pharmacodynamics; antibody purification and storage; and screening and labeling techniques.


Nucleic Acids


Inhibitory RNA


In some embodiments, the P2RX2 inhibitor is an inhibitory RNA molecule, e.g., that acts by way of the RNA interference (RNAi) pathway. An inhibitory RNA molecule can decrease the expression level (e.g., protein level or mRNA level) of P2RX2, a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7), or a molecule required for P2RX2 signaling or function (e.g., a molecule required for downstream calcium signaling). For example, an inhibitory RNA molecule includes a short interfering RNA, short hairpin RNA, and/or a microRNA that targets full-length P2RX2, a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7), or a molecule required for P2RX2 downstream signaling or function. A siRNA is a double-stranded RNA molecule that typically has a length of about 19-25 base pairs. A shRNA is a RNA molecule including a hairpin turn that decreases expression of target genes via RNAi. shRNAs can be delivered to cells in the form of plasmids, e.g., viral or bacterial vectors, e.g., by transfection, electroporation, or transduction). A microRNA is a non-coding RNA molecule that typically has a length of about 22 nucleotides. MiRNAs bind to target sites on mRNA molecules and silence the mRNA, e.g., by causing cleavage of the mRNA, destabilization of the mRNA, or inhibition of translation of the mRNA. In embodiments, the inhibitory RNA molecule decreases the level and/or activity of a negative regulator of function or a positive regulator of function. In other embodiments, the inhibitory RNA molecule decreases the level and/or activity of an inhibitor of a positive regulator of function.


An inhibitory RNA molecule can be modified, e.g., to contain modified nucleotides, e.g., 2′-fluoro, 2′-o-methyl, 2′-deoxy, unlocked nucleic acid, 2′-hydroxy, phosphorothioate, 2′-thiouridine, 4′-thiouridine, 2′-deoxyuridine. Without being bound by theory, it is believed that certain modification can increase nuclease resistance and/or serum stability, or decrease immunogenicity.


In some embodiments, the inhibitory RNA molecule decreases the level and/or activity or function of P2RX2, a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7), or a molecule required for P2RX2 downstream signaling or function. In embodiments, the inhibitory RNA molecule inhibits expression of P2RX2, a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7), or a molecule required for P2RX2 downstream signaling or function (e.g., inhibits translation to protein). In other embodiments, the inhibitor RNA molecule increases degradation of P2RX2, a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7), or a molecule required for P2RX2 downstream signaling or function and/or decreases the stability (i.e., half-life) of P2RX2, a P2RX2 binding partner (e.g., a binding partner selected from the group consisting of P2RX1, P2RX3, P2RX4, P2RX5, P2RX6, and P2RX7), or a molecule required for P2RX2 downstream signaling or function. The inhibitory RNA molecule can be chemically synthesized or transcribed in vitro.


The making and use of inhibitory therapeutic agents based on non-coding RNA such as ribozymes, RNAse P, siRNAs, and miRNAs are also known in the art, for example, as described in Sioud, RNA Therapeutics: Function, Design, and Delivery (Methods in Molecular Biology). Humana Press 2010.


Viral Vectors


Viral vectors can be used to express a neurotoxin from Table 9 for combination therapy with a P2RX2 inhibitor. A viral vector expressing a neurotoxin from Table 9 can be administered to a cell or to a subject (e.g., a human subject or animal model) to decrease or block neurotransmission. Viral vectors can be directly administered (e.g., injected) to a tumor to treat cancer.


Viral genomes provide a rich source of vectors that can be used for the efficient delivery of exogenous genes into a mammalian cell. Viral genomes are particularly useful vectors for gene delivery because the polynucleotides contained within such genomes are typically incorporated into the nuclear genome of a mammalian cell by generalized or specialized transduction. These processes occur as part of the natural viral replication cycle, and do not require added proteins or reagents in order to induce gene integration. Examples of viral vectors include a retrovirus (e.g., Retroviridae family viral vector), adenovirus (e.g., Ad5, Ad26, Ad34, Ad35, and Ad48), parvovirus (e.g., adeno-associated viruses), coronavirus, negative strand RNA viruses such as orthomyxovirus (e.g., influenza virus), rhabdovirus (e.g., rabies and vesicular stomatitis virus), paramyxovirus (e.g., measles and Sendai), positive strand RNA viruses, such as picornavirus and alphavirus, and double stranded DNA viruses including adenovirus, herpesvirus (e.g., Herpes Simplex virus types 1 and 2, Epstein-Barr virus, cytomegalovirus, replication deficient herpes virus), and poxvirus (e.g., vaccinia, modified vaccinia Ankara (MVA), fowlpox and canarypox). Other viruses include Norwalk virus, togavirus, flavivirus, reoviruses, papovavirus, hepadnavirus, human papilloma virus, human foamy virus, and hepatitis virus, for example. Examples of retroviruses include: avian leukosis-sarcoma, avian C-type viruses, mammalian C-type, B-type viruses, D-type viruses, oncoretroviruses, HTLV-BLV group, lentivirus, alpharetrovirus, gammaretrovirus, spumavirus (Coffin, J. M., Retroviridae: The viruses and their replication, Virology (Third Edition) Lippincott-Raven, Philadelphia, 1996). Other examples include murine leukemia viruses, murine sarcoma viruses, mouse mammary tumor virus, bovine leukemia virus, feline leukemia virus, feline sarcoma virus, avian leukemia virus, human T-cell leukemia virus, baboon endogenous virus, Gibbon ape leukemia virus, Mason Pfizer monkey virus, simian immunodeficiency virus, simian sarcoma virus, Rous sarcoma virus and lentiviruses. Other examples of vectors are described, for example, in U.S. Pat. No. 5,801,030, the teachings of which are incorporated herein by reference.


Blood Brain Barrier Permeability


In some embodiments, the P2RX2 inhibitors for use in the present invention are agents that are not capable of crossing, or that do not cross, the blood brain barrier (BBB) of a mammal, e.g., an experimental rodent (e.g., mouse or rat), dog, pig, non-human primate, or a human. The BBB is a highly selective semipermeable membrane barrier that separates the circulating blood from the brain extracellular fluid (e.g., cerebrospinal fluid) in the central nervous system (CNS). The BBB is made up of high-density endothelial cells, which are connected by tight junctions. These cells prevent most molecular compounds in the bloodstream (e.g., large molecules and hydrophilic molecules) from entering the brain. Water, some gases (e.g., oxygen and carbon dioxide), and lipid-soluble molecules (e.g., hydrophobic molecules, such as steroid hormones) can cross the BBB by passive diffusion. Molecules that are needed for neural function, such as glucose and amino acids, are actively transported across the BBB.


A number of approaches can be used to render an agent BBB impermeable. These methods include modifications to increase an agent's size, polarity, or flexibility or reduce its lipophilicity, targeting approaches to direct an agent to another part of the body and away from the brain, and packaging approaches to deliver an agent in a form that does not freely diffuse across the BBB. These approaches can be used to render a BBB permeable P2RX2 inhibitor impermeable, and they can also be used to improve the properties (e.g., cell-specific targeting) of a P2RX2 inhibitor that does not cross the BBB. The methods that can be used to render an agent BBB impermeable are discussed in greater detail herein below.


Formulation of BBB-Permeable Agents for Enhanced Cell Targeting


One approach that can be used to render a P2RX2 inhibitor BBB impermeable is to conjugate the agent to a targeting moiety that directs it somewhere other than the brain. The targeting moiety can be an antibody for a receptor expressed by the target cell (e.g., N-Acetylgalactosamine for liver transport; DGCR2, GBF1, GPR44 or SerpinB10 for pancreas transport; Secretoglobin, family 1A, member 1 for lung transport). The targeting moiety can also be a ligand of any receptor or other molecular identifier expressed on the target cell in the periphery. These targeting moieties can direct the P2RX2 inhibitor of interest to its corresponding target cell, and can also prevent BBB crossing by directing the agent away from the BBB and increasing the size of the P2RX2 inhibitor via conjugation of the targeting moiety.


P2RX2 inhibitors can also be rendered BBB impermeable through formulation in a particulate delivery system (e.g., a nanoparticle, liposome, or microparticle), such that the agent is not freely diffusible in blood and cannot cross the BBB. The particulate formulation used can be chosen based on the desired localization of the P2RX2 inhibitor (e.g., a tumor, lymph node, lymphoid organ, or site of inflammation), as particles of different sizes accumulate in different locations. For example, nanoparticles with a diameter of 45 nm or less enter the lymph node, while 100 nm nanoparticles exhibit poor lymph node trafficking. Some examples of the link between particle size and localization in vivo are described in Reddy et al., J Controlled Release 112:26 2006, and Reddy et al., Nature Biotechnology 25:1159 2007.


P2RX2 inhibitors can be tested after the addition of a targeting moiety or after formulation in a particulate delivery system to determine whether or not they cross the BBB. Models for assessing BBB permeability include in vitro models (e.g., monolayer models, co-culture models, dynamic models, multi-fluidic models, isolated brain microvessels), in vivo models, and computational models as described in He et al., Stroke 45:2514 2014; Bickel, NeuroRx 2:15 2005; and Wang et al., Int J Pharm 288:349 2005. A P2RX2 inhibitor that exhibits BBB impermeability can be used in the methods described herein.


Modification of Existing Compounds to Render them BBB Impermeable


There are multiple parameters that have been empirically derived in the field of medicinal chemistry to predict whether a compound will cross the BBB. The most common numeric value for describing permeability across the BBB is the log BB, defined as the logarithmic ratio of the concentration of a compound in the brain and in the blood. Empirical rules of thumb have been developed to predict BBB permeability, including rules regarding molecular size, polar surface area, sum of oxygen and nitrogen atoms, lipophilicity (e.g., partition coefficient between apolar solvent and water), “lipoaffinity”, molecular flexibility, and number of rotable bonds (summarized in Muehlbacher et al., J Comput Aided Mol Des. 25: 1095 2011; and Geldenhuys et al., Ther Deliv. 6: 961 2015). Some preferred limits on various parameters for BBB permeability are listed in Table 1 of Ghose et al., ACS Chem Neurosci. 3: 50 2012, which is incorporated herein by reference. Based on the parameters shown in the table, one of skill in the art could modify an existing P2RX2 inhibitor to render it BBB impermeable.


One method of modifying a P2RX2 inhibitor to prevent BBB crossing is to add a molecular adduct that does not affect the target binding specificity, kinetics, or thermodynamics of the agent. Molecular adducts that can be used to render an agent BBB impermeable include polyethylene glycol (PEG), a carbohydrate monomer or polymer, a dendrimer, a polypeptide, a charged ion, a hydrophilic group, deuterium, and fluorine. P2RX2 inhibitors can be tested after the addition of one or more molecular adducts or after any other properties are altered to determine whether or not they cross the BBB. Models for assessing BBB permeability include in vitro models (e.g., monolayer models, co-culture models, dynamic models, multi-fluidic models, isolated brain microvessels), in vivo models, and computational models as described in He et al., Stroke 45:2514 2014; Bickel, NeuroRx 2:15 2005; and Wang et al., Int J Pharm 288:349 2005. A P2RX2 inhibitor that exhibits BBB impermeability can be used in the methods described herein.


Screening for or Development of BBB Impermeable Agents


Another option for developing BBB impermeable agents is to find or develop new agents that do not cross the BBB. One method for finding new BBB impermeable agents is to screen for compounds that are BBB impermeable. Compound screening can be performed using in vitro models (e.g., monolayer models, co-culture models, dynamic models, multi-fluidic models, isolated brain microvessels), in vivo models, and computational models, as described in He et al., Stroke 45:2514 2014; Bickel, NeuroRx 2:15 2005; Wang et al., Int J Pharm 288:349 2005, and Czupalla et al., Methods Mol Biol 1135:415 2014. For example, the ability of a molecule to cross the blood brain barrier can be determined in vitro using a transwell BBB assay in which microvascular endothelial cells and pericytes are co-cultured separated by a thin macroporous membrane, see e.g., Naik et al., J Pharm Sci 101:1337 2012 and Hanada et al., Int J Mol Sci 15:1812 2014; or in vivo by tracking the brain uptake of the target molecule by histology or radio-detection. Compounds would be deemed appropriate for use as P2RX2 inhibitors in the methods described herein if they do not display BBB permeability in the aforementioned models.


Cancer


The methods described herein can be used to treat cancer in a subject by administering to the subject an effective amount of a P2RX2 inhibitor, e.g., a P2RX2 inhibitor described herein. The method may include administering locally (e.g., intratumorally) to the subject a P2RX2 inhibitor described herein in a dose (e.g., effective amount) and for a time sufficient to treat the cancer. For example, the stroma associated with the tumor, e.g., fibroblasts, is disrupted such that an essential function, e.g., the production of matrix metalloproteases, is altered to inhibit tumor survival or promote tumor control.


In some embodiments, the P2RX2 inhibitor inhibits proliferation or disrupts the function of non-neural cells associated with the cancer, e.g., the method includes administering to the subject an effective amount of a P2RX2 inhibitor for a time sufficient to inhibit proliferation or disrupt the function of non-neural cells associated with the cancer. Non-neural cells associated with the cancer include malignant cancer cells, malignant cancer cells in necrotic and hypoxic areas, adipocytes, pericytes, endothelial cells, cancer associated fibroblasts, fibroblasts, mesenchymal stem cells, red blood cells, or extracellular matrix. The proliferation of non-neural cells associated with the cancer may be decreased in the subject at least 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more, compared to before the administration. The proliferation of non-neural cells associated with the cancer can be decreased in the subject between 5-20%, between 5-50%, between 10-50%, between 20-80%, between 20-70%.


The P2RX2 inhibitor can treat cancer by increasing cancer cell death in a subject (e.g., a human subject or animal model) or in a cancer cell culture (e.g., a culture generated from a patient tumor sample, a cancer cell line, or a repository of patient samples). A P2RX2 inhibitor can increase cancer cell death by at least 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more compared to before administration to a subject or cancer cell culture. A P2RX2 inhibitor can increase cancer cell death in a subject or cancer cell culture between 5-20%, between 5-50%, between 10-50%, between 20-80%, between 20-70%.


The P2RX2 inhibitor can also act to inhibit cancer cell growth, proliferation, metastasis, migration, or invasion, e.g., the method includes administering to the subject (e.g., a human subject or animal model) or a cancer cell culture (e.g., a culture generated from a patient tumor sample, a cancer cell line, or a repository of patient samples) a P2RX2 inhibitor in an amount (e.g., an effective amount) and for a time sufficient to inhibit cancer cell growth, proliferation, metastasis, migration, or invasion. Cancer cell growth, proliferation, metastasis, migration, or invasion can be decreased in the subject or cancer cell culture at least 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more, compared to before the administration Cancer cell growth, proliferation, metastasis, migration, or invasion can be decreased in the subject or cancer cell culture between 5-20%, between 5-50%, between 10-50%, between 20-80%, between 20-70%.


The P2RX2 inhibitor can inhibit cancer cell invasion or metastasis along a nerve, e.g., the method includes administering to the subject (e.g., a human subject or animal model) a P2RX2 inhibitor in an amount (e.g., an effective amount) and for a time sufficient to inhibit cancer cell invasion or metastasis along a nerve. The P2RX2 inhibitor can decrease cancer cell invasion or metastasis along a nerve in the subject at least 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more, compared to before the administration. The P2RX2 inhibitor can decrease cancer cell invasion or metastasis along a nerve in the subject between 5-20%, between 5-50%, between 10-50%, between 20-80%, between 20-70%.


The P2RX2 inhibitor can also reduce the number of nerve fibers in the affected tissue or reduce the activity of peripheral nerve fibers in the affected tissue. For example, the method includes administering to the subject (e.g., a human subject or animal model) a P2RX2 inhibitor in an amount (e.g., an effective amount) and for a time sufficient to reduce the number of nerve fibers in the affected tissue or reduce the activity of peripheral nerve fibers in the affected tissue. The affected tissue can be a tumor or a tumor micro-environment. The number of nerve fibers in the affected tissue or the activity of peripheral nerve fibers in the affected tissue can be decreased in the subject at least 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more, compared to before the administration. The number of nerve fibers in the affected tissue or the activity of peripheral nerve fibers in the affected tissue can be decreased in the subject between 5-20%, between 5-50%, between 10-50%, between 20-80%, between 20-70%.


The nerve fibers that are modulated can be part of the peripheral nervous system, e.g., a somatic nerve, an autonomic nerve, a sensory nerve, a cranial nerve, an optic nerve, an olfactory nerve, a sympathetic nerve, a parasympathetic nerve, a chemoreceptor, a photoreceptor, a mechanoreceptor, a thermoreceptor, a nociceptor, an efferent nerve fiber, or an afferent nerve fiber.


Cancer Types


In the methods described herein, the cancer or neoplasm may be any solid or liquid cancer and includes benign or malignant tumors, and hyperplasias, including gastrointestinal cancer (such as non-metastatic or metastatic colorectal cancer, pancreatic cancer, gastric cancer, esophageal cancer, hepatocellular cancer, cholangiocellular cancer, oral cancer, lip cancer); urogenital cancer (such as hormone sensitive or hormone refractory prostate cancer, renal cell cancer, bladder cancer, penile cancer); gynecological cancer (such as ovarian cancer, cervical cancer, endometrial cancer); lung cancer (such as small-cell lung cancer and non-small-cell lung cancer); head and neck cancer (e.g., head and neck squamous cell cancer); CNS cancer including malignant glioma, astrocytomas, retinoblastomas and brain metastases; malignant mesothelioma; non-metastatic or metastatic breast cancer (e.g., hormone refractory metastatic breast cancer); skin cancer (such as malignant melanoma, basal and squamous cell skin cancers, Merkel Cell Carcinoma, lymphoma of the skin, Kaposi Sarcoma); thyroid cancer; bone and soft tissue sarcoma; and hematologic neoplasias (such as multiple myeloma, acute myelogenous leukemia, chronic myelogenous leukemia, myelodysplastic syndrome, acute lymphoblastic leukemia, Hodgkin's lymphoma).


Additional cancers that can be treated according to the methods described herein include breast cancer, lung cancer, stomach cancer, colon cancer, liver cancer, renal cancer, colorectal cancer, prostate cancer, pancreatic cancer, cervical cancer, anal cancer, vulvar cancer, penile cancer, vaginal cancer, testicular cancer, pelvic cancer, thyroid cancer, uterine cancer, rectal cancer, brain cancer, head and neck cancer, esophageal cancer, bronchus cancer, gallbladder cancer, ovarian cancer, bladder cancer, oral cancer, oropharyngeal cancer, larynx cancer, biliary tract cancer, skin cancer, a cancer of the central nervous system, a cancer of the respiratory system, and a cancer of the urinary system. Examples of breast cancers include, but are not limited to, triple-negative breast cancer, triple-positive breast cancer, HER2-negative breast cancer, HER2-positive breast cancer, estrogen receptor-positive breast cancer, estrogen receptor-negative breast cancer, progesterone receptor-positive breast cancer, progesterone receptor-negative breast cancer, ductal carcinoma in situ (DCIS), invasive ductal carcinoma, invasive lobular carcinoma, inflammatory breast cancer, Paget disease of the nipple, and phyllodes tumor.


Other cancers that can be treated according to the methods described herein include leukemia (e.g., B-cell leukemia, T-cell leukemia, acute myeloid leukemia (AML), chronic myeloid leukemia (CML), acute lymphocytic (lymphoblastic) leukemia (ALL), chronic lymphocytic leukemia (CLL), and erythroleukemia), sarcoma (e.g., angiosarcoma, chondrosarcoma, Ewing's sarcoma, fibrosarcoma, gastrointestinal stromal tumor, leiomyosarcoma, liposarcoma, malignant peripheral nerve sheath tumor, malignant fibrous cytoma, osteosarcoma, pleomorphic sarcoma, rhabdomyosarcoma, synovial sarcoma, vascular sarcoma, Kaposi's sarcoma, dermatofibrosarcoma, epithelioid sarcoma, leyomyosarcoma, and neurofibrosarcoma), carcinoma (e.g., basal cell carcinoma, large cell carcinoma, small cell carcinoma, non-small cell lung carcinoma, renal carcinoma, hepatocarcinoma, gastric carcinoma, choriocarcinoma, adenocarcinoma, hepatocellular carcinoma, giant (or oat) cell carcinoma, squamous cell carcinoma, adenosquamous carcinoma, anaplastmic carcinoma, adrenocortical carcinoma, cholangiocarcinoma, Merkel cell carcinoma, ductal carcinoma in situ (DCIS), and invasive ductal carcinoma), blastoma (e.g., hepatoblastoma, medulloblastoma, nephroblastoma, neuroblastoma, pancreatoblastoma, pleuropulmonary blastoma, retinoblastoma, and glioblastoma multiforme), lymphoma (e.g., Hodgkin's lymphoma, non-Hodgkin's lymphoma, and Burkitt lymphoma), myeloma (e.g., multiple myeloma, plasmacytoma, localized myeloma, and extramedullary myeloma), melanoma (e.g., superficial spreading melanoma, nodular melanoma, lentigno maligna melanoma, acral lentiginous melanoma, and amelanotic melanoma), neuroma (e.g., ganglioneuroma, Pacinian neuroma, and acoustic neuroma), glioma (e.g., astrocytoma, oligoastrocytoma, ependymoma, brainstem glioma, optic nerve glioma, and oligoastrocytoma), pheochromocytoma, meningioma, malignant mesothelioma, and virally induced cancer.


In some embodiments, the cancer is a paraneoplastic cancer (e.g., a cancer that causes a paraneoplastic syndrome). Paraneoplastic syndromes are rare disorders that are triggered by an altered immune system response to a neoplasm, and are mediated by humoral factors such as hormones, cytokines, or auto-antibodies produced by the tumor. Symptoms of paraneoplastic syndrome may be endocrine, neuromuscular, or musculoskeletal, cardiovascular, cutaneous, hematologic, gastrointestinal, renal, or neurological. Paraneoplastic syndromes commonly present with lung, breast, and ovarian cancer and cancer of the lymphatic system (e.g., lymphoma). Paraneoplastic neurological disorders are disorders that affect the central or peripheral nervous system, and can include symptoms such as ataxia (difficulty with walking and balance), dizziness, nystagmus (rapid uncontrolled eye movements), difficulty swallowing, loss of muscle tone, loss of fine motor coordination, slurred speech memory loss, vision problems, sleep disturbances, dementia, seizures, or sensory loss in the limbs. Breast, ovarian, and lung cancers are most commonly associated with paraneoplastic neurological disorders. Other common types of paraneoplastic syndromes include paraneoplastic cerebellar degeneration, paraneoplastic pemphigus, paraneoplastic autonomic neuropathy, paraneoplastic encephalomyelitis, and cancer-associated autoimmune retinopathy.


Endocrine paraneoplastic syndromes include Cushing syndrome (caused by ectopic ACTH), which is most commonly caused by small cell lung cancer, pancreatic carcinoma, neural tumors, or thymoma; SIADH (caused by antidiuretic hormone), which is most commonly caused by small cell lung cancer and CNS malignancies; hypercalcemia (caused by PTHrp, TGFα, TNF, or IL-1), which is most commonly caused by lung cancer, breast carcinoma, renal and bladder carcinoma, multiple myeloma, adult T cell leukemia/lymphoma, ovarian carcinoma, and squamous cell carcinoma (e.g., lung, head, neck, or esophagus carcinoma); hyperglycemia (caused by insulin insulin-like substance, or “big” IGF-II), which is most commonly caused by fibrosarcoma, mesenchymal sarcomas, insulinoma, and hepatocellular carcinoma; carcinoid syndrome (caused by serotonin or bradykinin), which is most commonly caused by bronchial adenoma, pancreatic carcinoma, and gastric carcinoma; and hyperaldosteronism (caused by aldosterone), which is most commonly caused by adrenal adenoma/Conn's syndrome, non-Hodgkin's lymphoma, ovarian carcinoma, and pulmonary cancer.


Neurological paraneoplastic syndromes include Lambert-Eaton myasthenic syndrome (LEMS), which is most commonly caused by small cell lung cancer; paraneoplastic cerebellar degeneration, which is most commonly caused by lung cancer, ovarian cancer, breast carcinoma, and Hodgkin's lymphoma; encephalomyelitis; limbic encephalitis, which is most commonly caused by small cell lung carcinoma; myasthenia gravis, which is most commonly caused by thymoma; brainstem encephalitis; opsoclonus myoclonus ataxia (caused by autoimmune reaction against Nova-1), which is most commonly caused by breast carcinoma, ovarian carcinoma, small cell lung carcinoma, and neuroblastoma; anti-NMDA receptor encephalitis (caused by autoimmune reaction against NMDAR subunits), which is most commonly caused by teratoma; and polymyositis, which is most commonly caused by lung cancer, bladder cancer, and non-Hodgkin's lymphoma. Mucotaneous paraneoplastic syndromes include acanthosis nigricans, which is most commonly caused by gastric carcinoma, lung carcinoma, and uterine carcinoma; dermatomyositis, which is most commonly caused by bronchogenic carcinoma, breast carcinoma, ovarian cancer, pancreatic cancer, stomach cancer, colorectal cancer, and Non-Hodgkin's lymphoma; Leser-Trelat sign; necrolytic migratory erythema, which is most commonly caused by glucoganoma; Sweet's syndrome; florid cutaneous papillomatosis; pyoderma gangrenosum; and acquired generalized hypertrichosis.


Hematological syndromes include granulocytosis (caused by G-CSF); polycythemia (caused by erythropoietin), which is commonly caused by renal carcinoma, cerebellar hemangioma, and heptatocellular carcinoma; Trousseau sign (caused by mucins), which is commonly caused by pancreatic carcinoma and bronchogenic carcinoma; nonbacterial thrombotic endocarditis, which is caused by advanced cancers; and anemia, which is most commonly caused by thymic neoplasms. Other paraneoplastic syndromes include membranous glomerular nephritis; neoplastic fever; Staffer syndrome, which is caused by renal cell carcinoma; and tumor-induced osteomalacia (caused by FGF23), which is caused by hemangiopericytoma and phosphaturic mesenchymal tumor.


In some embodiments, a subject is identified as having cancer after presenting with symptoms of a paraneoplastic syndrome. A common symptom of paraneoplastic syndrome is fever. Auto-antibodies directed against nervous system proteins are also frequently observed in patients with paraneoplastic syndromes, including anti-Hu, anti-Yo, anti-Ri, anti-amphiphysin, anti-CV2, anti-Ma2, anti-recoverin, anti-transducin, anti-carbonic anhydrase II, anti-arrestin, anti-GCAP1, anti-GCAP2, anti-HSP27, anti-Rab6A, and anti-PNR. Other symptoms that can be used to identify a patient with paraneoplastic cancer include ataxia, dizziness, nystagmus, difficulty swallowing, loss of muscle tone, loss of fine motor coordination, slurred speech memory loss, vision loss, sleep disturbances, dementia, seizures, dysgeusia, cachexia, anemia, itching, or sensory loss in the limbs. In some embodiments, a patient presents with symptoms of paraneoplastic syndrome and is then identified as having cancer based on imaging tests (e.g., CT, MRI, or PET scans).


The cancer may be highly innervated, metastatic, non-metastatic cancer, or benign (e.g., a benign tumor). The cancer may be a primary tumor or a metastasized tumor.


In some embodiments, the cancer is a P2RX2-associated cancer (e.g., a cancer in which P2RX2 is expressed, amplified, and/or overexpressed).


Subjects who can be treated with the methods disclosed herein include subjects who have had one or more tumors resected, received chemotherapy or other pharmacological treatment for the cancer, received radiation therapy, and/or received other therapy for the cancer. Subjects who have not previously been treated for cancer can also be treated with the methods disclosed herein.


Combination Therapies


A P2RX2 inhibitor described herein can be administered in combination with a second therapeutic agent for treatment of cancer. In some embodiments, the second therapeutic agent is selected based on tumor type, tumor tissue of origin, tumor stage, or mutations in genes expressed by the tumor.


Checkpoint Inhibitors


One type of agent that can be administered in combination with a P2RX2 inhibitor described herein is a checkpoint inhibitor. Checkpoint inhibitors can be broken down into at least 4 major categories: i) agents such as antibodies that block an inhibitory pathway directly on T cells or NK cells (e.g., PD-1 targeting antibodies such as nivolumab and pembrolizumab, antibodies targeting TIM-3, and antibodies targeting LAG-3, 2B4, CD160, A2aR, BTLA, CGEN-15049, or KIR), ii) agents such as antibodies that activate stimulatory pathways directly on T cells or NK cells (e.g., antibodies targeting OX40, GITR, or 4-1 BB), iii) agents such as antibodies that block a suppressive pathway on immune cells or rely on antibody-dependent cellular cytotoxicity to deplete suppressive populations of immune cells (e.g., CTLA-4 targeting antibodies such as ipilimumab, antibodies targeting VISTA, and antibodies targeting PD-L2, Gr1, or Ly6G), and iv) agents such as antibodies that block a suppressive pathway directly on cancer cells or that rely on antibody-dependent cellular cytotoxicity to enhance cytotoxicity to cancer cells (e.g., rituximab, antibodies targeting PD-L1, and antibodies targeting B7-H3, B7-H4, Gal-9, or MUC1). Such agents described herein can be designed and produced, e.g., by conventional methods known in the art (e.g., Templeton, Gene and Cell Therapy, 2015; Green and Sambrook, Molecular Cloning, 2012).


Chemotherapy


A second type of therapeutic agent that can be administered in combination with a P2RX2 inhibitor described herein is a chemotherapeutic agent (e.g., a cytotoxic agent or other chemical compound useful in the treatment of cancer). These include alkylating agents, antimetabolites, folic acid analogs, pyrimidine analogs, purine analogs and related inhibitors, vinca alkaloids, epipodopyyllotoxins, antibiotics, L-asparaginase, topoisomerase inhibitors, interferons, platinum coordination complexes, anthracenedione substituted urea, methyl hydrazine derivatives, adrenocortical suppressant, adrenocorticosteroides, progestins, estrogens, antiestrogen, androgens, antiandrogen, and gonadotropin-releasing hormone analog. Also included is 5-fluorouracil (5-FU), leucovorin (LV), irenotecan, oxaliplatin, capecitabine, paclitaxel and doxetaxel. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethiylenethiophosphoramide and trimethylolomelamine; acetogenins (especially bullatacin and bullatacinone); a camptothecin (including the synthetic analogue topotecan); bryostatin; callystatin; CC-1065 (including its adozelesin, carzelesin and bizelesin synthetic analogues); cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin; duocarmycin (including the synthetic analogues, KW-2189 and CB1-TM1); eleutherobin; pancratistatin; a sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, and ranimnustine; antibiotics such as the enediyne antibiotics (e.g., calicheamicin, especially calicheamicin gammaII and calicheamicin omegaII; dynemicin, including dynemicin A; bisphosphonates, such as clodronate; an esperamicin; as well as neocarzinostatin chromophore and related chromoprotein enediyne antiobiotic chromophores), aclacinomysins, actinomycin, authramycin, azaserine, bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycinis, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-doxorubicin and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins such as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine; androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; an epothilone; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidainine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanmol; nitraerine; pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-ethylhydrazide; procarbazine; razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; trichothecenes (especially T-2 toxin, verracurin A, roridin A and anguidine); urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxoids, e.g., paclitaxel; chloranbucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum coordination complexes such as cisplatin, oxaliplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin; xeloda; ibandronate; irinotecan (e.g., CPT-11); topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the above. Two or more chemotherapeutic agents can be used in a cocktail to be administered in combination with the first therapeutic agent described herein. Suitable dosing regimens of combination chemotherapies are known in the art.


Biologic Cancer Agents


Another type of therapeutic agent that can be administered in combination with a P2RX2 inhibitor described herein is a therapeutic agent that is a biologic such a cytokine (e.g., interferon or an interleukin (e.g., IL-2)) used in cancer treatment. In other embodiments the biologic is an anti-angiogenic agent, such as an anti-VEGF agent, e.g., bevacizumab. In some embodiments the biologic is an immunoglobulin-based biologic, e.g., a monoclonal antibody (e.g., a humanized antibody, a fully human antibody, an Fc fusion protein or a functional fragment thereof) that agonizes a target to stimulate an anti-cancer response, or antagonizes an antigen important for cancer. Such agents include Rituximab; Daclizumab; Basiliximab; Palivizumab; Infliximab; Trastuzumab; Gemtuzumab ozogamicin; Alemtuzumab; Ibritumomab tiuxetan; Adalimumab; Omalizumab; Tositumomab-I-131; Efalizumab; Cetuximab; Bevacizumab; Natalizumab; Tocilizumab; Panitumumab; Ranibizumab; Eculizumab; Certolizumab pegol; Golimumab; Canakinumab; Ustekinumab; Ofatumumab; Denosumab; Motavizumab; Raxibacumab; Belimumab; Ipilimumab; Brentuximab Vedotin; Pertuzumab; Ado-trastuzumab emtansine; and Obinutuzumab. Also included are antibody-drug conjugates. Examples of biologic cancer agents that can be used in combination with P2RX2 inhibitors described herein are shown in Table 3 below.









TABLE 3







APPROVED CANCER ANTIBODIES










Antibody
Company
Antigen
Indication





ado-trastuzumab
Genentech
HER2
Metastatic breast cancer


emtansine





alemtuzumab
Genzyme
CD52
B-cell chronic lymphocytic leukemia


atezolizumab
Genentech
PD-L1
Urothelial carcinoma





Metastatic non-small cell lung cancer


avelumab
EMD Serono
PD-L1
Metastatic Merkel cell carcinoma


bevacizumab
Genentech
VEGF
Metastatic colorectal cancer


blinatumomab
Amgen
CD19
Precursor B-cell acute lymphoblastic leukemia


brentuximab
Seattle Genetics
CD30
Hodgkin lymphoma


vedotin


Anaplastic large-cell lymphoma


cetuximab
ImClone Systems
EGFR
Metastatic colorectal carcinoma


daratumumab
Janssen Biotech
CD38
Multiple myeloma


dinutuximab
United Therapeutics
GD2
Pediatric high-risk neuroblastoma


durvalumab
AstraZeneca
PD-L1
Urothelial carcinoma


elotuzumab
Bristol-Myers
SLAMF7
Multiple myeloma



Squibb




ibritumomab
Spectrum
CD20
Relapsed or refractory low-grade, follicular, or


tiuxetan
Pharmaceuticals

transformed B-cell non-Hodgkin's lymphoma


ipilimumab
Bristol-Myers
CTLA-4
Metastatic melanoma



Squibb




necitumumab
Eli Lilly
EGFR
Metastatic squamous non-small cell lung





carcinoma


nivolumab
Bristol-Myers
PD-1
Metastatic melanoma



Squibb

Metastatic squamous non-small cell lung





carcinoma


obinutuzumab
Genentech
CD20
Chronic lymphocytic leukemia


ofatumumab
Glaxo Grp
CD20
Chronic lymphocytic leukemia


olaratumab
Eli Lilly
PDGFRA
Soft tissue sarcoma


panitumumab
Amgen
EGFR
Metastatic colorectal cancer


pembrolizumab
Merck
PD-1
Metastatic melanoma


pertuzumab
Genentech
HER2
Metastatic breast cancer


ramucirumab
Eli Lilly
VEGFR2
Gastric cancer


rituximab
Genentech
CD20
B-cell non-Hodgkin's lymphoma


trastuzumab
Genentech
HER2
Metastatic breast cancer









Cancer-Specific Agents


In some embodiments, the therapeutic agents administered with the P2RX2 inhibitors described herein are cancer-specific. Cancer-specific agents are agents that have been shown to be particularly effective against certain types of cancer. Cancer-specific agents that can be administered with the P2RX2 inhibitors described herein are listed in Table 4 below.









TABLE 4







CANCER-SPECIFIC AGENTS








Cancer type
Agents





Pancreatic cancer
Chemotherapeutics (Paclitaxel Albumin-stabilized Nanoparticle Formulation,



Erlotinib Hydrochloride, Everolimus, Fluorouracil Injection, Gemcitabine



Hydrochloride, Irinotecan Hydrochloride Liposome, Mitomycin C, Sunitinib Malate,



Folfirinox, Gemcitabine-Cisplatin, Gemcitabine-Oxaliplatin, Off, Lanreotide



Acetate, Abraxane, Gemcitabine, Irinotecan, 5-FU, Oxaliplatin)


Melanoma
Checkpoint inhibitors (pembro, ipi, nivolumab, durvalumab), BRaf inhibitors



(vemurafenib, debrafenib), MEK inhibitors, CDK4 inhibitors (ribociclib)


Renal cell carcinoma
Checkpoint inhibitors (pembro, ipi, nivolumab, durvalumab), mTOR inhibitors



(everolimus), bevacizumab


Lung cancer
Checkpoint inhibitors (pembro, ipi, nivolumab, durvalumab), EGFR inhibitors



(erlotinib, gefitinib, cetuximab)


Esophageal cancer
Chemotherapeutic agents (5FU, docetaxel), trastuzumab


Ovarian cancer
Chemotherapeutics (taxanes, cisplatin)


Uterine cancer
Chemotherapeutics (taxanes, cisplatin)


Head and Neck
Checkpoint inhibitors (pembro, ipi, nivolumab, durvalumab), EGFR inhibitors


cancer
(erlotinib, gefitinib, cetuximab)


Mesothelioma
Chemotherapeutics (pemetrexed, cisplatin)









Non-Drug Therapies


Another type of agent that can be administered in combination with a P2RX2 inhibitor is a therapeutic agent that is a non-drug treatment. For example, the second therapeutic agent is radiation therapy, cryotherapy, hyperthermia and/or surgical excision of tumor tissue.


Neurotransmission Blockers


In some embodiments, the P2RX2 inhibitor is administered in combination with a neurotransmission blocker (e.g., an agent that decreases neurotransmission). A neurotransmission blocker can be used to reduce or inhibit neural activity in a cancer or tumor that is innervated by nerves or to decrease the number of nerves in the tumor. For example, in some embodiments, the neurotransmission blocker is an antagonist of a neurotransmitter receptor listed in Table 5. Exemplary antagonists are listed in Tables 7A-7K. Neurotransmission blockers also include agents that decrease neurotransmitter synthesis or release (e.g., agents that decrease the activity of a biosynthetic protein encoded by a gene in Table 5 via inhibition or downregulation, or agents that decrease the activity of a synaptic or vesicular protein via blocking, disrupting, downregulating, or antagonizing the protein), increase neurotransmitter reuptake or degradation (e.g., agents that agonize, open, or stabilize transporters that remove neurotransmitter from the synaptic cleft), decrease neurotransmitter receptor activity (e.g., agents that decrease the activity of a signaling protein encoded by a gene in Table 5 via blocking or antagonizing the protein, or agents that block, antagonize, or downregulate a neurotransmitter receptor listed in Table 5), decrease neurotransmitter receptor synthesis or membrane insertion, increase neurotransmitter degradation, regulate neurotransmitter receptor conformation (e.g., agents that bind to a receptor and keep it in a “closed” or “inactive” conformation), and disrupt the pre- or postsynaptic machinery (e.g., agents that block or disrupt a structural protein, or agents that block, disrupt, downregulate, or antagonize a synaptic or vesicular protein). In some embodiments, the neurotransmitter receptor is a channel (e.g., a ligand or voltage gated ion channel), the activity of which can be decreased by blockade, antagonism, or inverse agonism of the channel. Neurotransmission blockers further include agents that sequester, block, antagonize, or degrade a neurotransmitter listed in Tables 5 or 6. Neurotransmission blockers include antibodies that bind to or block the function of neurotransmitters, neurotransmitter receptor antagonists, and toxins that disrupt synaptic release. Neurotransmission modulators can decrease neurotransmission by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98% or more. Neurotransmission blockers can be administered in any of the modalities described herein (e.g., antibody, small molecule, nucleic acid, polypeptide, or viral vector).









TABLE 5







NEUROTRANSMITTER GENES & PATHWAYS














Accession
Entrez


Gene
Pathway
Type
Number
Gene ID














ABAT
Neurotransmitter
Biosynthesis
P80404
18


ACHE
Neurotransmitter
Biosynthesis
P22303
43


ADORA2A
Neurotransmitter
Receptor
P29274
135


ADORA2B
Neurotransmitter
Receptor
P29275
136


Adra1a
Adrenergic/
Receptor
P35348
148



Neurotransmitter





Adra1b
Adrenergic/
Receptor
P35368
147



Neurotransmitter





Adra1d
Adrenergic/
Receptor
P25100
146



Neurotransmitter





Adra2a
Adrenergic/
Receptor
P08913
150



Neurotransmitter





Adra2b
Adrenergic/
Receptor
P18089
151



Neurotransmitter





Adra2c
Adrenergic/
Receptor
P18825
152



Neurotransmitter





Adrb1
Adrenergic/
Receptor
P08588
153



Neurotransmitter





Adrb2
Adrenergic/
Receptor
P07550
154



Neurotransmitter





Adrb3
Adrenergic/
Receptor
P13945
155



Neurotransmitter





Adrbk1
Adrenergic
Kinase
P25098
156


Adrbk2
Adrenergic
Kinase
P35626
157


BACE1
Neurotransmitter
Biosynthesis
P56817
23621


BCHE
Neurotransmitter
Biosynthesis
P06276
590


BRS3
Neuromodulator
Receptor
P32247
P32247


C6orf89
Neuromodulator
Receptor
Q6UWU4
221477


CHAT
Neurotransmitter
Biosynthesis
P28329
1103


CHRFAM7A
Neurotransmitter
Receptor
Q494W8
89832


Chrm1
Cholinergic/
Receptor
P11229
1128



Neurotransmitter





Chrm2
Cholinergic/
Receptor
P08172
1129



Neurotransmitter





Chrm3
Cholinergic/
Receptor
P20309
1131



Neurotransmitter





Chrm4
Cholinergic/
Receptor
P08173
1132



Neurotransmitter





Chrm5
Cholinergic/
Receptor
P08912
1133



Neurotransmitter





Chrna1
Cholinergic/
Receptor
P02708
1134



Neurotransmitter





Chrna10
Cholinergic/
Receptor
Q9GZZ6
57053



Neurotransmitter





Chrna2
Cholinergic/
Receptor
Q15822
1135



Neurotransmitter





Chrna3
Cholinergic/
Receptor
P32297
1136



Neurotransmitter





Chrna4
Cholinergic/
Receptor
P43681
1137



Neurotransmitter





Chrna5
Cholinergic/
Receptor
P30532
1138



Neurotransmitter





Chrna6
Cholinergic/
Receptor
Q15825
8973



Neurotransmitter





Chrna7
Cholinergic/
Receptor
P36544
1139



Neurotransmitter





Chrna9
Cholinergic/
Receptor
Q9UGM1
55584



Neurotransmitter





Chrnb1
Cholinergic/
Receptor
P11230
1140



Neurotransmitter





Chrnb2
Cholinergic/
Receptor
P17787
1141



Neurotransmitter





Chrnb3
Cholinergic/
Receptor
Q05901
1142



Neurotransmitter





Chrnb4
Cholinergic/
Receptor
P30926
1143



Neurotransmitter





Chrnd
Cholinergic/
Receptor
Q07001
1144



Neurotransmitter





Chrne
Cholinergic/
Receptor
Q04844
1145



Neurotransmitter





Chrng
Cholinergic/
Receptor
P07510
1146



Neurotransmitter





CNR1
Cannabinoid/
Receptor
P21554
1268



Neurotransmitter





CNR2
Cannabinoid/
Receptor
P34972
1269



Neurotransmitter





CNRIP1
Neurotransmitter
Receptor
Q96F85
25927


COMT
Neurotransmitter
Biosynthesis
P21964
1312


CPA4
Neurotransmitter
Biosynthesis
Q9UI42
51200


CPE
Neuropeptide/
Biosynthesis
P16870
1363



Neurotransmitter





CREM
Neurotransmitter
Signaling
Q03060
1390


DAGLA
Neurotransmitter
Biosynthesis
Q9Y4D2
747



(Cannabinoid)





DAGLB
Neurotransmitter
Biosynthesis
Q8NCG7
221955



(Cannabinoid)





DBH
Neurotransmitter
Biosynthesis
P09172
1621


DDC
Neurotransmitter
Biosynthesis
P20711
1644


DGKI
Neurotransmitter
Biosynthesis
O75912
9162


DOPO
Dopaminergic
Receptor
P09172
1621


DPP4
Neurotransmitter
Biosynthesis
P27487
1803


Drd1
Dopaminergic/
Receptor
P21728
1812



Neurotransmitter





Drd2
Dopaminergic/
Receptor
P14416
1813



Neurotransmitter





Drd3
Dopaminergic/
Receptor
P35462
1814



Neurotransmitter





Drd4
Dopaminergic/
Receptor
P21917
1815



Neurotransmitter





Drd5
Dopaminergic/
Receptor
P21918
1816



Neurotransmitter





ECEL1
Neurotransmitter
Biosynthesis
O95672
9427


FAAH
Neurotransmitter
Biosynthesis
O00519
2166


FNTA
Neurotransmitter
Signaling
P49354
2339


GABARAP
Neurotransmitter
Receptor
O95166
11337


GABARAPL1
Amine
Receptor
Q9H0R8
23710



Neuromodulator





GABARAPL2
Amine
Receptor
P60520
11345



Neuromodulator





GABBR1
Neurotransmitter
Receptor
Q9UBS5
2550


GABBR2
Amine
Receptor
O75899
9568



Neuromodulator





GABRA1
Neurotransmitter
Receptor
P14867
2554


GABRA2
Neurotransmitter
Receptor
P47869
2555


GABRA3
Neurotransmitter
Receptor
P34903
2556


GABRA4
Neurotransmitter
Receptor
P48169
2557


GABRA5
Neurotransmitter
Receptor
P31644
2558


GABRA6
Neurotransmitter
Receptor
Q16445
2559


GABRB1
Neurotransmitter
Receptor
P18505
2560


GABRB2
Neurotransmitter
Receptor
P47870
2561


GABRB3
Neurotransmitter
Receptor
P28472
2562


GABRD
Neurotransmitter
Receptor
O14764
2563


GABRE
Neurotransmitter
Receptor
P78334
2564


GABRG1
Neurotransmitter
Receptor
Q8N1C3
2565


GABRG2
Neurotransmitter
Receptor
P18507
2566


GABRG3
Neurotransmitter
Receptor
Q99928
2567


GABRP
Neurotransmitter
Receptor
O00591
2568


GABRQ
Neurotransmitter
Receptor
Q9UN88
55879


GABRR1
Neurotransmitter
Receptor
P24046
2569


GABRR2
Neurotransmitter
Receptor
P28476
2570


GABRR3
Neurotransmitter
Receptor
A8MPY1
200959


GAD1
Neurotransmitter
Biosynthesis
Q99259
2571


GAD2
Neurotransmitter
Biosynthesis
Q05329
2572


GCHFR
Neurotransmitter
Biosynthesis
P30047
2644


GLRA1
Neurotransmitter
Receptor
P23415
2741


GLRA2
Neurotransmitter
Receptor
P23416
2742


GLRA3
Neurotransmitter
Receptor
O75311
8001


GLRA4
Neurotransmitter
Receptor
Q5JXX5
441509


GLRB
Neurotransmitter
Receptor
P48167
2743


GLS
Neurotransmitter
Biosynthesis
O94925
2744


GLS2
Neurotransmitter
Biosynthesis
Q9UI32
27165


GluA1 (GluR1)
Amine
Receptor
P42261
2890



Neuromodulator





GluK1 (GluR5)
Amine
Receptor
P39086
2897



Neuromodulator





GLUL
Neurotransmitter
Biosynthesis
P15104
2752


GluN1(NR1)
Amine
Receptor
Q05586
2902



Neuromodulator





GNMT
Neurotransmitter
Biosynthesis
Q14749
27232


GPER1
Neurotransmitter
Receptor
Q99527
2852


GPR1
Neurotransmitter
Receptor
P46091
2825


GPR139
Neurotransmitter
Receptor
Q6DWJ6
124274


GPR143
Neurotransmitter
Receptor
P51810
4935


GPR149
Neurotransmitter
Receptor
Q86SP6
344758


GPR18
Neurotransmitter
Receptor
Q14330
2841


GPR21
Neurotransmitter
Receptor
Q99679
2844


GPR26
Neurotransmitter
Receptor
Q8NDV2
2849


GPR3
Neurotransmitter
Receptor
P46089
2827


GPR35
Neurotransmitter
Receptor
Q9HC97
2859


GPR52
Neurotransmitter
Receptor
Q9Y2T5
9293


GPR55
Neurotransmitter
Receptor
Q9Y2T6
9290


GPR78
Neurotransmitter
Receptor
Q96P69
27201


GPR83
Neurotransmitter
Receptor
Q9NYM4
10888


GPR84
Neurotransmitter
Receptor
Q9NQS5
53831


GPRASP1
Neurotransmitter
Receptor
Q5JY77
9737


GPR50
Amine
Receptor
Q13585
9248



Neuromodulator





GRIA1
Neurotransmitter
Receptor
P42261
2890


GRIA2
Neurotransmitter
Receptor
P42262
2891


GRIA3
Neurotransmitter
Receptor
P42263
2892


GRIA4
Neurotransmitter
Receptor
P48058
2893


GRID1
Neurotransmitter
Receptor
Q9ULK0
2894


GRID2
Neurotransmitter
Receptor
O43424
2895


GRIK1
Neurotransmitter
Receptor
P39086
2897


GRIK2
Neurotransmitter
Receptor
Q13002
2898


GRIK3
Neurotransmitter
Receptor
Q13003
2899


GRIK4
Neurotransmitter
Receptor
Q16099
2900


GRIK5
Neurotransmitter
Receptor
Q16478
2901


GRIN1
Neurotransmitter
Receptor
Q05586
2902


GRIN2A
Neurotransmitter
Receptor
Q12879
2903


GRIN2B
Neurotransmitter
Receptor
Q13224
2904


GRIN2C
Neurotransmitter
Receptor
Q14957
2905


GRIN2D
Neurotransmitter
Receptor
Q15399
2906


GRIN3A
Neurotransmitter
Receptor
Q8TCU5
116443


GRIN3B
Neurotransmitter
Receptor
O60391
116444


GRK2
Neurotransmitter
Receptor
P25098
156


GRK3
Neurotransmitter
Receptor
P35626
157


GRM1
Neurotransmitter
Receptor
Q13255
2911


GRM2
Neurotransmitter
Receptor
Q14416
2912


GRM3
Neurotransmitter
Receptor
Q14832
2913


GRM4
Neurotransmitter
Receptor
Q14833
2914


GRM5
Neurotransmitter
Receptor
P41594
2915


GRM6
Neurotransmitter
Receptor
O15303
2916


GRM7
Neurotransmitter
Receptor
Q14831
2917


GRM8
Neurotransmitter
Receptor
O00222
2918


HNMT
Neurotransmitter
Biosynthesis
P50135
3176


HOMER1
Neurotransmitter
Receptor
Q86YM7
9456


HRH1
Neurotransmitter
Receptor
P35367
3269


HRH2
Neurotransmitter
Receptor
P25021
3274


HRH3
Neurotransmitter
Receptor
Q9Y5N1
11255


HRH4
Neurotransmitter
Receptor
Q9H3N8
59340


Htr1a
Neurotransmitter
Receptor
P08908
3350


Htr1b
Neurotransmitter
Receptor
P28222
3351


Htr1c
Neurotransmitter
Receptor
P28335



Htr1d
Neurotransmitter
Receptor
P28221
3352


Htr1e
Neurotransmitter
Receptor
P28566
3354


Htr1f
Neurotransmitter
Receptor
P30939
3355


Htr2a
Neurotransmitter
Receptor
P28223
3356


Htr2b
Neurotransmitter
Receptor
P41595
3357


Htr2c
Neurotransmitter
Receptor
P28335
3358


Htr3a
Neurotransmitter
Receptor
P46098
3359


Htr3b
Neurotransmitter
Receptor
O95264
9177


Htr3c
Neurotransmitter
Receptor
Q8WXA8
170572


Htr3d
Neurotransmitter
Receptor
Q70Z44
200909


HTR3E
Neurotransmitter
Receptor
A5X5Y0
285242


Htr4
Neurotransmitter
Receptor
Q13639
3360


Htr5a
Neurotransmitter
Receptor
P47898
3361


Htr5b
Neurotransmitter
Receptor
P35365
79247


HTR5BP
Neurotransmitter
Receptor

645694


Htr6
Neurotransmitter
Receptor
P50406
3362


Htr7
Neurotransmitter
Receptor
P32305
3363


ITPR1
Neurotransmitter
Signaling
Q14643
3708


ITPR2
Neurotransmitter
Signaling
Q14571
3709


ITPR3
Neurotransmitter
Signaling
Q14573
3710


LYNX1
Neurotransmitter
Receptor
Q9BZG9
66004


MAOA
Neurotransmitter
Biosynthesis
P21397
4128


MAOB
Neurotransmitter
Biosynthesis
P27338
4129


NAMPT
Neurotransmitter
Biosynthesis
P43490
10135


NISCH
Neurotransmitter
Receptor
Q9Y2I1
11188


NOS1
Neurotransmitter
Biosynthesis
P29475
4842


NPTN
Neurotransmitter
Receptor
Q9Y639
27020


P2RX1
Neurotransmitter
Receptor
P51575
5023


P2RX2
Neurotransmitter
Receptor
Q9UBL9
22953


P2RX3
Neurotransmitter
Receptor
P56373
5024


P2RX4
Neurotransmitter
Receptor
Q99571
5025


P2RX5
Neurotransmitter
Receptor
Q93086
5026


P2RX6
Neurotransmitter
Receptor
O15547
9127


P2RX7
Neurotransmitter
Receptor
Q99572
5027


P2RY11
Neurotransmitter
Receptor
Q96G91
5032


PAH
Neurotransmitter
Biosynthesis
P00439
5053


PC
Neurotransmitter
Biosynthesis
P11498
5091


PDE1B
Neurotransmitter
Signaling
Q01064
5153


PDE4A
Neurotransmitter
Signaling
P27815
5141


PDE4D
Neurotransmitter
Signaling
Q08499
5144


PHOX2A
Neurotransmitter
Biosynthesis
O14813
401


PHOX2B
Neurotransmitter
Biosynthesis
Q99453
8929


PIK3CA
Neurotransmitter
Signaling
P42336
5290


PIK3CB
Neurotransmitter
Signaling
P42338
5291


PIK3CG
Neurotransmitter
Signaling
P48736
5294


PLCB1
Neurotransmitter
Signaling
Q9NQ66
23236


PLCB2
Neurotransmitter
Signaling
Q00722
5330


PLCB3
Neurotransmitter
Signaling
Q01970
5331


PLCB4
Neurotransmitter
Signaling
Q15147
5332


PLCD1
Neurotransmitter
Signaling
P51178
5333


PLCE1
Neurotransmitter
Signaling
Q9P212
51196


PLCG1
Neurotransmitter
Signaling
P19174
5335


PLCL1
Neurotransmitter
Signaling
Q15111
5334


PLCL2
Neurotransmitter
Signaling
Q9UPR0
23228


PPP1CB
Neurotransmitter
Signaling
P62140
5500


PPP100
Neurotransmitter
Signaling
P36873
5501


PRIMA1
Neurotransmitter
Biosynthesis
Q86XR5
145270


PRKACG
Neurotransmitter
Signaling
P22612
5568


PRKAR2B
Neurotransmitter
Signaling
P31323
5577


PRKCG
Neurotransmitter
Signaling
P05129
5582


PRKX
Neurotransmitter
Signaling
P51817
5613


RIC3
Neurotransmitter
Receptor
Q7Z5B4
79608


SHANK3
Neurotransmitter
Signaling
Q9BYB0
85358


SLC6A1
Amine
Transferase
P30531
6529



Neuromodulator





SLC6A13
Amine
Transferase
Q9NSD5
6540



Neuromodulator





Slc6a4
Serotonin
Transporter
P31645
6532


SNX13
Neurotransmitter
Signaling
Q9Y5W8
23161


TAAR1
Amine
Receptor
Q96RJ0
134864



Neuromodulator





TAAR2
Amine
Receptor
Q9P1P5
9287



Neuromodulator





TAAR5
Neurotransmitter
Receptor
O14804
9038


TH
Neurotransmitter
Biosynthesis
P07101
7054


TPH1
Neurotransmitter
Biosynthesis
P17752
7166


TPH2
Neurotransmitter
Biosynthesis
Q8IWU9
121278


TRHDE
Neurotransmitter
Biosynthesis
Q9UKU6
29953
















TABLE 6







NEUROTRANSMITTERS









Ligand
Pathway
Type





2-Arachidonoylglycerol
Endocannabinoid
Ligand


2-Arachidonyl glyceryl ether
Endocannabinoid
Ligand


3-methoxytyramine
Amines
Ligand


Acetylcholine
Amino Acids
Ligand


Adenosine
Purine
Ligand


Adenosine triphosphate
Purine
Ligand


Agmatine
Amino Acids
Ligand


Anandamide
Endocannabinoid
Ligand


Aspartate
Amino Acids
Ligand


Carbon monoxide
Gas
Ligand


D-serine
Amino Acids
Ligand


Dopamine
Monoamines
Ligand


Dynorphin
Opioids
Ligand


Endorphin
Opioids
Ligand


Enkephalin
Opioids
Ligand


Epinephrine
Monoamines
Ligand


Gamma-aminobutyric acid
Amino Acids
Ligand


Glutamate
Amino Acids
Ligand


Glycine
Amino Acids
Ligand


Histamine
Monoamines
Ligand


N-Acetylaspartylglutamate
Neuropeptides
Ligand


N-Arachidonoyl dopamine
Endocannabinoid
Ligand


N-methylphenethylamine
Amines
Ligand


N-methyltryptamine
Amines
Ligand


Nitric oxide
Gas
Ligand


Norepinephrine
Monoamines
Ligand


Octopamine
Amines
Ligand


Phenethylamine
Amines
Ligand


Serotonin
Monoamines
Ligand


Synephrine
Amines
Ligand


Tryptamine
Amines
Ligand


Tyramine
Amines
Ligand


Virodhamine
Endocannabinoid
Ligand
















TABLE 7A







AGONISTS AND ANTAGONIST AGENTS









Gene
Agonist
Antagonist





Adrb2
NCX 950
Alprenolol


Accession Number:
Bitolterol
Carvedilol


P07550
Isoetarine
Desipramine



Norepinephrine
Nadolol



Phenylpropanolamine
Levobunolol



Dipivefrin
Metipranolol



Epinephrine
Bevantolol



Orciprenaline
Oxprenolol



Dobutamine
Nebivolol



Ritodrine
Asenapine



Terbutaline
Bupranolol



Salmeterol
Penbutolol



Formoterol
Celiprolol



Salbutamol
Pindolol



Isoprenaline
Acebutolol



Arbutamine
Bopindolol



Arformoterol




Fenoterol




Pirbuterol




Ephedra




Procaterol




Clenbuterol




Bambuterol




Indacaterol




Droxidopa




Olodaterol




Vilanterol




Pseudoephedrine




Cabergoline




Mirtazepine



Adra1d
Midodrine
Dapiprazole


Accession Number:
Norepinephrine
Amitriptyline


P25100
Clonidine
Alfuzosin



Oxymetazoline
Promazine



Pergolide
Prazosin



Bromocriptine
Imipramine



Droxidopa
Nortriptyline



Xylometazoline
Doxazosin



Ergotamine
Nicardipine



Cirazoline
Dronedarone



Cabergoline
Tamsulosin



Methoxamine
Propiomazine



Epinephrine
Phenoxybenzamine




Carvedilol




Doxepin




Terazosin




Quetiapine




Methotrimeprazine




Silodosin


Adrb1
Isoetarine
Esmolol


Accession Number:
Norepinephrine
Betaxolol


P08588
Phenylpropanolamine
Metoprolol



Epinephrine
Atenolol



Dobutamine
Timolol



Salbutamol
Sotalol



Isoprenaline
Propranolol



Arbutamine
Labetalol



Fenoterol
Bisoprolol



Pirbuterol
Alprenolol



Ephedra
Amiodarone



Clenbuterol
Carvedilol



Droxidopa
Nadolol



Pseudoephedrine
Levobunolol



Carteolol
Metipranolol



Cabergoline
Bevantolol



Mirtazapine
Practolol



Loxapine
Oxprenolol



Vortioxetine
Celiprolol



Desipramine
Nebivolol




Asenapine




Bupranolol




Penbutolol




Pindolol




Acebutolol




Bopindolol




Cartelol


Adrb3
SR 58611
Bopindolol


Accession Number:
Norepinephrine
Propranolol


P13945
Epinephrine
Bupranolol



Isoprenaline




Arbutamine




Fenoterol




Ephedra




Clenbuterol




Droxidopa




Mirabegron



Adrbk1
ATP
Alprenolol


Accession Number:
Carbachol
Heparin


P25098
Dopamine




Isoproterenol




Morphine




DAMGO




histamine




Acetylcholine




Etorphine




NMDA




Dopamine



Adrbk2
Isoproterenol
Propranolol


Accession Number:
DAMGO



P26819
ATP



Chrm3
cgmp
MT3


Accession Number:
ATP
Hexocyclium


P20309
Cevimeline
Himbacine



arecoline
Biperiden



oxotremorine-M
lithocholylcholine



NNC 11-1314
AFDX384



xanomeline
4-DAMP



oxotremorine
hexahydrodifenidol



pentylthio-TZTP
VU0255035



arecaidine propargyl ester
N-methyl scopolamine



NNC 11-1607
Darifenacin



furmethide
Thiethylperazine



NNC 11-1585
methoctramine



Acetylcholine
silahexocyclium



methylfurmethide
Strychnine



Bethanechol
MT7



Carbachol
Heparin



Succinylcholine
Olanzapine



ALKS 27
Pirenzepine



itopride
Clidinium



methacholine
Ipratropium



Meperidine
Propantheline



Cinnarizine
Dicyclomine



Trimipramine
Darifenacin




Tiotropium




Atropine




Scopolamine




Amitriptyline




Doxepin




Lidocaine




Nortriptyline




Tropicamide




Metixene




Homatropine Methylbromide




Solifenacin




Glycopyrrolate




Propiomazine




Diphemanil Methylsulfate




Promethazine




Diphenidol




Pancuronium




Ziprasidone




Quetiapine




Imipramine




Clozapine




Cyproheptadine




Aripiprazole




Nicardipine




Amoxapine




Loxapine




Promazine




Oxyphencyclimine




Anisotropine Methylbromide




Tridihexethyl




Chlorpromazine




Ketamine




Cyclosporin A




Paroxetine




Benzquinamide




Tolterodine




Oxybutynin




Alcuronium




WIN 62, 577




Tramadol




Chlorprothixene




Aclidiniurn




Methotrimeprazine




Umeclidinium




Cryptenamine




Mepenzolate




Maprotiline




Brompheniramine




Isopropamide




Trihexyphenidyl




Ipratropium bromide




Hyoscyamine




Procyclidine




Pipecuronium




Fesoterodine




Disopyramide




Desipramine




Mivacuriurn


Chrna3
Nicotine
A-867744


Accession Number:
Varenicline
NS1738


P32297
Acetylcholine
Hexamethonium



Ethanol
Mecamylamine



Cytisine
Dextromethorphan



Levamisole
Pentolinium



Galantamine
Levomethadyl Acetate




Bupropion


Chrna6
Nicotine
Hexamethonium


Accession Number:
Cytisine
Mecamylamine


Q15825
Varenicline




Galantamine



Chrna9
Nicotine
Hexamethonium


Accession Number:
Galantamine
Mecamylamine


Q9UGM1
Ethanol
Tetraethylammonium




Muscarine



ATG003
Strychnine



Lobeline




RPI-78M



Chrnb1
Galantamine



Accession Number:




P11230




Chrnb4
Nicotine
Atropine


Accession Number:
Varenicline
Oxybutynin


P30926
PNU-120596
Pentolinium



Ethanol
Dextromethorphan



Galantamine



Chrng
Galantamine



Accession Number:




P07510




Adcyap1
Nicotine
Atropine


Accession Number:
CGMP
PPADS


P18509
Apomorphine
Onapristone



Suramin
Muscarine



Nifedipine
Haloperidol



ATP
Astressin



Dihydrotestosterone
Melatonin



Maxadilan
Scopolamine



Dexamethasone
Tetrodotoxin



Acetylcholine
Apamin



Histamine
Hexamethonium



Carbachol
Indomethacin



NMDA
Propranolol



Dopamine
Bumetanide



Isoproterenol
Progesterone



Salbutamol
Charybdotoxin



Morphine
Prazosin



Clonidine




Nimodipine




2,6-Diamino-Hexanoic Acid Amide



CYSLTR1
Salbutamol
Montelukast


Accession Number:
Dexamethasone
Zafirlukast


Q9Y271
Arachidonic acid
Cinalukast



Histamine
Pranlukast




Nedocromil




Theophylline




Indomethacin




Zileuton




Iralukast




Pobilukast




Sulukast




Verlukast


LTB4R
LTB
U75302


Accession Number:
ATP
CP105696


Q15722
Dexamethasone
CP-195543



cholesterol
Etalocib



20-hydroxy-LTB<
SC-41930



12R-HETE
LY255283



arachidonic acid
Zafirlukast




ONO-4057




RO5101576




BILL 260


PENK
Dopamine
Naltrexone


Accession Number:
kainate
Naloxone


P01210
NMDA
Progesterone



DAMGO




Morphine



Htr2c
Apomorphine
Melatonin


Accession Number:
Bifeprunox
SB 224289


P28335
Tramadol
LY334362



AL-37350A
FR260010



5-MeO-DMT
Sulpiride



BW723C86
Thiethylperazine



CGS-12066
cyamemazine



DOI
Mesulergine



5-CT
SB 221284



YM348
Zotepine



LSD
Metergoline



xanomeline
methiothepin



WAY-163909
Spiperone



Dopamine
SB 215505



LY344864
Tiospirone



VER-3323
SB 228357



TFMPP
Pizotifen



8-OH-DPAT
SB 206553



MK-212
SB 204741



NMDA
SDZ SER-082



org 12962
Ritanserin



5-MeOT
SB 242084



RU 24969
S33084



Acetylcholine
Roxindole



QUINPIROLE
RS-127445



quipazine
Terguride



tryptamine
EGIS-7625



Ro 60-0175
SB 243213



Oxymetazoline
RS-102221



Ergotamine
Olanzapine



Cabergoline
Aripiprazole



Lorcaserin
Agomelatine



Pergolide
Ziprasidone



Methylergonovine
Quetiapine



Renzapride
Sarpogrelate



Pramipexole
Perphenazine



GR-127935
Thioridazine



BRL-15572
Sertindole



ipsapirone
Loxapine



SB 216641
Methysergide



SL65.0155
Risperidone



S 16924
Asenapine



Bromocriptine
Mianserin



Lisuride
Clozapine



Tegaserod
Trifluoperazine



Epicept NP-1
Trazodone



dapoxetine
Doxepin



Dexfenfluramine
Nortriptyline



3,4-
Chlorprothixene



Methylenedioxymethamphetamine




Ropinirole
Minaprine



Maprotiline
Propiomazine



Desipramine
Mirtazapine




Amoxapine




Yohimbine




Cyproheptadine




Imipramine




Amitriptyline




Promazine




Chlorpromazine




Ketamine




Propranolol




Fluoxetine




Ketanserin




Mesulergine




AC-90179




Ergoloid mesylate 2




Methotrimeprazine




Paliperidone




Clomipramine




Trimipramine




Captodiame




Nefazodone


GABA Receptor
Bamaluzole
bicuculline


Accession Numbers
GABA
Metrazol


(Q9UBS5, O95166,
Gabamide
Flumazenil


O75899, P28472, P18507,
GABOB
Thiothixine


P47870, P47869, O14764)
Gaboxadol
Bupropion



Ibotenic acid
Caffeine



Isoguvacine




Isonipecotic acid




Muscimol




Phenibut




Picamilon




Progabide




Quisqualamine




SL 75102




Thiomuscimol




Alcohols (e.g., ethanol, isopropanol)




Avermectins (e.g., ivermectin)




Barbiturates (e.g., phenobarbital)




Benzodiazepines




Bromides (e.g., potassium bromide




Carbamates (e.g., meprobamate,




carisoprodol)




Chloralose




Chlormezanone




Clomethiazole




Dihydroergolines (e.g., ergoloid




(dihydroergotoxine))




Etazepine




Etifoxine




Imidazoles (e.g., etomidate)




Kavalactones (found in kava)




Loreclezole




Neuroactive steroids (e.g.,




allopregnanolone, ganaxolone)




Nonbenzodiazepines (e.g.,




zaleplon, zolpidem, zopiclone,




eszopiclone)




Petrichloral




Phenols (e.g., propofol)




Piperidinediones (e.g., glutethimide,




methyprylon)




Propanidid




Pyrazolopyridines (e.g., etazolate)




Quinazolinones (e.g.,




methaqualone)




Skullcap constituents




Stiripentol




Sulfonylalkanes (e.g.,




sulfonmethane, tetronal, trional)




Valerian constituents (e.g., valeric




acid, valerenic acid)




Volatiles/gases (e.g., chloral




hydrate, chloroform, diethyl ether,




sevoflurane)



Glutamate Receptor
3,5-dihydroxyphenylglycine
APICA


Accession Number:
eglumegad
EGLU


(P42261, P39086,
Biphenylindanone A
LY-341, 495


P39086, Q13585, P42261,
DCG-IV



P42262, P42263, P48058,
L-AP4



P39086, Q13002,




Q13003, Q13003,




Q16478, Q12879,




Q14957, Q13224,




Q14957, Q15399,




Q8TCU5, O60391)




CNR1/CNR2
N-Arachidonoylethanolamine
SR 141716A


Accession Number:
2-Arachidonoyl-glycerol
LY-320135


(P21554, P34972)
2-Arachidonoyl-glycerylether
AM251



N-Arachidonoyl-dopamine
AM281



O-Arachidonoyl-ethanolamine
SR 144528



N-Arachidonoylethanolamine
AM630



2-Arachidonoyl-glycerol




2-Arachidonoyl-glycerylether




N-Arachidonoyl-dopamine




O-Arachidonoyl-ethanolamine




Δ-9-THC




CP-55, 940




R(+)-WIN 55, 212-2




HU-210




Levonantradol




Nabilone




Methanandamide




ACEA




O-1812




Δ9-THC




CP-55, 940




R(+)-WIN 55, 212-2




HU-210




Levonantradol




Nabilone




Methanandamide




JWH-015




JWH-133
















TABLE 7B







ADRENERGIC AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Non-selective
adrenaline (epinephrine),
carvedilol, arotinolol, and labetalol



noradrenaline (norepinephrine),




isoprenaline (isoproterenol),




dopamine, caffeine, nicotine,




tyramine, methylphenidate, ephedrine




and pseudophedrine.



α1 selective (ADRA1A,
phenylephrine, methoxamine,
acepromazine, alfuzosin, doxazosin,


ADRA1B, ADRA1D)
midodrine, cirazoline,
labetalol, phenoxybenzamine,



xylometazoline, metaraminol
KW3902, phentolamine, prazosin,



chloroehtylclonidine, oxymetazoline
tamsulosin, terazosin, tolazoline,




trazodone, amitriptyline, silodosin,




clomipramine, doxepin, trimipramine,




typical and atypical antipsychotics, and




antihistamines, such as hyroxyzine


α2 selective (ADRA2A,
α-methyl dopa, clonidine,
phentolamine, phenoxybenzamine,


ADRA2B, ADRA2C)
brimonidine, agmatine,
yohimbine, idazoxan, atipamezole,



dexmedetomidine,
mirtazapine, tolazoline, trazodone, and



medetomidine, romifidine
typical and atypical antipsychotics



chloroethylclonidine,




detomidine, lofexidine, xylazine,




tizanidine, guanfacine, and amitraz



β1 selective (ADRB1)
Dobutamine
metroprolol, atenolol, acebutolol,




bisoprolol, betaxolol, levobetaxolol,




esmolol, celiprolol, carteolol, landiolol,




oxprenolol, propanolol, practolol,




penbutolol, timolol, labetalol, nebivolol,




levobunolol, nadolol, pindolol, sotalol,




metipranolol, tertatolol, vortioxene


β2 selective (ADRB2)
salbutamol, albuterol, bitolterol
butaxamine, acebutolol, timolol,



mesylate, levabuterol, ritodrine,
propanolol, levobunolol, carteolol,



metaproterenol, terbutaline,
labetalol, pindolol, oxprenolol, nadolol,



salmeterol, formoterol, and pirbuterol
metipranolol, penbutolol, tertatolol,




sotalol


β3 selective (ADRB3)
L-796568, amibegron, solabegron,
SR 59230A, arotinolol



mirabegron
















TABLE 7C







DOPAMINE AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Non-selective
pramipexole, ropinirole, rotigotine,
haloperidol, paliperidone, clozapine,



apomorphine,
risperidone, olanzapine, quetiapine,



propylnorapomorphine,
ziprasidone, metoclopramide,



bromocriptine, cabergoline,
droperidol, domperidone, amoxapine,



ciladopa, dihydrexidine,
clomipramine, trimipramine, choline,



dinapsoline, doxamthrine,
melatonin, acepromazine, amisulpride,



epicriptine, lisuride, pergolide,
asenapine, azaperone, benperidol,



piribedil, quinagolide, roxindole,
bromopride, butaclamol,



dopamine
chlorpromazine, clebopride,




chlorprothixene, clopenthixol,




clocapramine, eticlopride, flupenthixol,




fluphenazine, fluspirilene, hydroxyzine,




itopride, iodobenzamide,




levomepromazine, levosulpiride,




loxapine, mesoridazine, metopimazine,




mosapramine, nafadotride,




nemonapride, penfluridol, perazine,




perphenazine, pimozide,




prochlorperazine, promazine,




pipotiazine, raclopride, remoxipride,




spiperone, spiroxatrine, stepholidine,




sulpiride, sultopride,




tetrahydropalmatine, thiethylperazine,




thioridazine, thiothixene, tiapride,




trifluoperazine, trifluperidol,




triflupromazine, thioproperazine,




taractan, zotepine, zuclopenthixol,




ziprasidone, ANP-010, NGD-94-4


D1 (DRD1)
Fenoldopam, A-86929,
SCH-23,390, SKF-83,959, Ecopipam,



dihydrexidine, dinapsoline,
Clebopride, Flupenthixol,



dinoxyline, doxanthrine, SKF-
Zuclopenthixol, Taractan, PSYRX-101,



81297, SKF-82958, SKF-38393, G-
LuAF-35700, GLC-756, ADX10061,



BR-APB, dopexamine
Zicronapine


D2 (DRD2)
Cabergoline, pergolide,
Chloroethylnorapomorphine,



quinelorane, sumanirole, talipexole,
desmethoxyfallypride, domperidone,



piribedil, quinpirole, quinelorane,
eticlopride, fallypride, hydroxyzine,



dinoxyline, dopexamine
itopride, L-741,626, SV 293, yohimbine,




raclopride, sulpiride, paliperidone,




penfluridol, quetiapine, lurasidone,




risperidone, olanzapine, blonanserin,




perphenazine, metoclopramide,




trifluoperazine, clebopride, levosulpiride,




flupenthixol, haloperidol, thioridazine,




alizapride, amisulpride, asenapine,




bromopride, bromperidol, clozapine,




fluphenazine, perphanazine, loxapine,




nemonapride, pericyazine,




pipamperone, prochlorperazine,




thioproperazine, thiethylperazine,




tiapride, ziprasidone, zuclopenthixol,




taractan, fluanisone, melperone,




molindone, remoxipride, sultopride,




ALKS 3831, APD-403, ONC201,




pridopidine, DSP-1200, NG-101, TAK-




906, ADN-1184, ADN-2013, AG-0098,




DDD-016, IRL-626, KP303, ONC-206,




PF-4363467, PGW-5, CG-209, ABT-




925, AC90222, ACP-005, ADN-2157,




CB030006, CLR-136, Egis-11150,




Iloperidone, JNJ-37822681, DLP-115,




AZ-001, S-33138, SLV-314, Y-931,




YKP1358, YK-P1447, APD405, CP-




903397, ocaperidone, zicronapine,




TPN-902


D3 (DRD3)
Piribedil, quinpirole, captodiame,
Domperidone, FAUC 365, nafadotride,



compound R, R-16, FAUC 54,
raclopride, PNU-99,194, SB-277011-A,



FAUC 73, PD-128,907, PF-
sulpiride, risperidone, YQA14, U99194,



219,061, PF-592,379, CJ-1037,
SR 21502, levosulpiride, amisulpride,



FAUC 460, FAUC 346, cariprazine
nemonapride, ziprasidone, taractan,




sultopride, APD-403, F17464, ONC201,




NG-101, TAK-906, ONC-206, PF-




4363467, ABT-127, ABT-614, GSK-




598809, GSK-618334, S-14297, S-




33138, YKP1358, YK-P1447


D4 (DRD4)
WAY-100635, A-412,997, ABT-724,
A-381393, FAUC 213, L-745,870, L-



ABT-670, FAUC 316, PD-168, 077,
570,667, ML-398, fananserin, clozapine,



CP-226,269
PNB-05, SPI-376, SPI-392, Lu-35-138,




NG D-94-1


D5 (DRD5)
Dihydrexidine, rotigotine, SKF-
SCH 23390



83,959, fenoldopam,



Partial
aplindore, brexpiprazole,




aripiprazole, CY-208,243,




pardoprunox, phencyclidine, and




salvinorin A
















TABLE 7D







GABA AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





GABAA
barbiturates (e.g., allobarbital,
bicuculline, gabazine, hydrastine,



amobarbital, aprobarbital, alphenal,
pitrazepin, sinomenine, tutin,



barbital, brallobarbital,
thiocolchicoside, metrazol, securinine,



phenobarbital, secobarbital,
gabazine



thiopental), bamaluzole, GABA,




GABOB, gaboxadol, ibotenic acid,




isoguvacine, isonipecotic acid,




muscimol, phenibut, picamilon,




progabide, quisqualamine, SL




75102, thiomuscimol, positive




allosteric modulators (PAMs) (e.g.,




alcohols, such as ethanol and




isopropanol; avermectins, such as




ivermectin; benzodiazepines, such




as diazepam, alprazolam,




chlordiazepoxide, clonazepam,




flunitrazepam, lorazepam,




midazolam, oxazepam, prazepam,




brotizolam, triazolam, estazolam,




lormetazepam, nitrazepam,




temazepam, flurazepam,




clorazepate halazepam, prazepam,




nimetazapem, adinazolam, and




climazolam; bromides, such as




potassium bromide; carbamates,




such as meprobamate and




carisoprodol; chloralose;




chlormezanone; chlomethiazole;




dihydroergolines, such as ergoloid;




etazepine; etifoxine; imidazoles,




such as etomidate;




imidazopyridines, such as alpidem




and necopdiem; kavalactones;




loreclezole; neuroactive steroids,




such as allogregnanolone,




pregnanolone,




dihydrodeoxycorticosterone,




tetrahydrodeoxycortisosterone,




androstenol, androsterone,




etiocholanolone, 3α-androstanediol,




5α, 5β, or 3α-dihydroprogesterone,




and ganaxolone;




nonbenzodiazepines, such as




zalepon, zolpidem, zopiclone, and




eszopiclone; petrichloral; phenols,




such as propofol; piperidinediones,




such as glutethimide and




methyprylon; propanidid;




pyrazolopyridines, such as




etazolate; pyrazolopyrimidines,




such as divaplon and fasiplon;




cyclopyrrolones, sush as pagoclone




and suproclone; β-cabolines, such




as abecarnil and geodecarnil;




quinazolinones, such as




methaqualone; Scutellaria




constituents; stiripentol;




sulfonylalkanes, such as




sulfonomethane, teronal, and




trional; Valerian constituents, such




as valeric acid and valerenic acid;




and gases, such as chloral hydrate,




chloroform, homotaurine, diethyl




ether, and sevoflurane.



GABAB
1,4-butanediol, baclofen, GABA,
CGP-35348, homotaurine, phaclofen,



Gabamide,GABOB, gamma-
saclofen, and SCH-50911



butyrolactone, gamma-




hydroxybutyric acid, gamma-




hyrdoxyvaleric acid, gamma-




valerolactone, isovaline,




lesogaberan, phenibut, picamilon,




progabide, homotaurine, SL-75102,




tolgabide



GABAA-ρ
CACA, CAMP, GABA, GABOB, N4-
gabazine, gaboxadol, isonipecotic acid,



chloroacetylcytosine arabinoside,
SKF-97,541, and (1,2,5,6-



picamilon, progabide, tolgabide,
Tetrahydropyridin-4-yl)methylphosphinic



and neuroactive steroids, such as
acid



allopregnanolone, THDOC, and




alphaxol one
















TABLE 7E







MUSCARINC AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Chrm1
AF102B, AF150(S), AF267B,
atropine, dicycloverine, hyoscyamine,



acetylcholine, carbachol, cevimeline,
ipratropium, mamba toxin muscarinic



muscarine, oxotremorine, pilocarpine,
toxin 7 (MT7), olanzapine, oxybutynin,



vedaclidine, 77-LH-28-1, CDD-0097,
pirenzepine, telenzepine, and



McN-A-343, L689,660, and
tolterodine



xanomeline



Chrm2
acetylcholine, methacholine, iper-8-
atropine, dicycloverine, hyoscyamine,



naph, berbine, and (2S,2′R,3′S,5′R)-
otenzepad, AQRA-741, AFDX-384,



1-methyl-2-(2-methyl-1,3-oxathiolan-
thorazine, diphenhydramine,



5-yl)pyrrolidine 3-sulfoxide methyl
dimenhydrinate, ipratropium,



iodide
oxybutynin, pirenzepine,




methoctramine, tripitramine,




gallamine, and tolterodine


Chrm3
acetylcholine, bethanechol,
atropine, dicycloverine, hyoscyamine,



carbachol, L689, 660, oxotremorine,
alcidium bromide, 4-DAMP,



pilocarpine, aceclidine, arecoline,
darifenacin, DAU-5884, HL-031,120,



and cevimeline
ipratropium, J-104,129, oxybutynin,




tiotropium, zamifenacin, and




tolterodine


Chrm4
acetylcholine, carbachol, and
AFDX-384, dicycloverine, himbacine,



oxotremorine), and Chrm5 agonists
mamba toxin 3, PD-102,807,



(e.g., acetylcholine, milameline,
PD-0298029, and tropicamide



sabcomeline



Chrm5
acetylcholine, milameline,
VU-0488130, xanomeline



sabcomeline



Non-selective

scopolamine, hydroxyzine,




doxylamine, dicyclomine, flavoxate,




cyclopentolate, atropine methonitrate,




trihexyphenidyl/benzhexol,




solifenacin, benzatropine,




mebeverine, and procyclidine
















TABLE 7F







NICOTINIC AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Chrna receptors
choline, acetylcholine, carbachol,
turbocurarine, bupropion,



methacholine, nicotine, varenicline
mecamylamine, 18-



tartrate, galantamine hydrobromide,
methozycoronaridine,



suxamethonium chloride
hexamethonium, trimethaphan,



(succinylcholine chloride),
atraciurium, doxacurium, mivacurium,



epibatidine, iobeline,
pancuronium, vecuronium,



decamethonium, isopronicline/TC-
succinylcholine, dextromethorphan,



1734/AZD3480 (TC-1734), AZD1446
neramexane, dextrophan, and 3-



(TC-6683), TC-5619, TC-5214, MEM
methoxymorphinan



3454 (RG3487), ABT-894, ABT-560,




EVP-6124, EVP-4473, PNU-282987,




AR-R17779, SSR 189711, JN403,




ABBF, PHA-543613, SEN12333,




GTS-21/DMXB-A, AZD0328, A-




582941, ABT-418, 5-iodo-A-85380,




SIB-1765F, ABT-089, and ABT-594
















TABLE 7G







SEROTONIN AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





5-HT1A
azapirones, such as alnespirone,
pindolol, tertatolol, alprenolol, AV-



binosperone, buspirone,
965, BMY-7,378, cyanopindolol,



enilospirone, etapirone, geprione,
dotarizine, flopropione, GR-46,611,



ipsaprione, revospirone, zalospirone,
iodocyanopindolol, isamoltane,



perospirone, tiosperone,
lecozotan, mefway, methiothepin,



umespirone, and tandospirone; 8-
methysergide, MPPF, NAN-190,



OH-DPAT, befiradol, F-15,599,
oxprenolol, pindobind, propanolol,



lesopitron, MKC-242, LY-283,284,
risperidone, robalzotan, SB-649,915,



osemozotan, repinotan U-92,016-A,
SDZ-216,525, spiperone, spiramide,



RU-24969, 2C-B, 2C-E, 2C-T-2,
spiroxatrine, UH-301, WAY-100,135,



aripiprazole, asenapine, bacoside,
WAY-100,635, and xylamidine



befiradol, brexpiprazole, bufotenin,




cannabidiol, and fibanserin



5-HT1B
triptans, such as sumatriptan,
methiothepin, yohimbine,



rizatriptan, eletriptan, donitripatn,
metergoline, aripiprazole, isamoltane,



almotriptan, frovatriptan, avitriptan,
AR-A000002, SB-216,641, SB-



zolmitriptan, and naratriptan;
224,289, GR-127,935, SB-236,057



ergotamine, 5-




carboxamidotryptamine, CGS-




12066A, CP-93,129, CP-94,253,




CP-122,288, CP-135,807, RU-24969,




vortioxetine, ziprasidone, and




asenapine



5-HT1D
triptans, such as sumatriptan,
ziprasidone, methiothepin,



rizatriptan, and naratriptan;
yohimbine, metergoline, ergotamine,



ergotamine, 5-(nonyloxy)tryptaime,
BRL-15572, vortioxetine,



5-(t-butyl)-N-methyltryptamine,
GR-127,935, LY-310,762, LY-367,642,



CP-286,601, PNU-109,291, PNU-
LY-456,219, and LY-456,220



142,633, GR-46611, L-694,247,




L-772,405, CP-122,288,




and CP-135,807



5-HT1E
BRL-54443, eletriptan



5-HT1F
LY-334,370, 5-n-butyryloxy-DMT,




BRL-54443, eletriptan, LY-344,864,




naratriptan, and lasmiditan



5-HT2A
25I-NBOH, 25I-NBOMe, (R)-DOI,
cyproheptadine, methysergide,



TCB-2, mexamine, O-4310, PHA-
quetiapine, nefazodone, olanzapine,



57378, OSU-6162, 25CN-NBOH,
asenapine, pizotifen, LY-367,265,



juncosamine, efavirenz, mefloquine,
AMDA, hydroxyzine, 5-MeO-NBpBrT,



lisuride, and 2C-B
and niaprazine


5-HT2B
fenfluramine, pergolide, cabergoline,
agomelatine, aripiprazole,



mefloquine, BW-723086, Ro60-
sarpogrelate, lisuride, tegaserod,



0175, VER-3323, 6-APB,
metadoxine, RS-127,445, SDZ SER-



guanfacine, norfenfluramine, 5-MeO-
082, EGIS-7625, PRX-08066, SB-



DMT, DMT, mCPP, aminorex,
200,646, SB-204,741, SB-206,553,



chlorphentermine, MEM, MDA, LSD,
SB-215,505, SB-228,357, LY-



psilocin, MDMA
266,097, and LY-272,015


5-HT2C
lorcaserin, lisuride, A-372,159, AL-
agomelatine, CPC, eltoprazine,



38022A, CP-809,101, fenfluramine,
etoperidone, fluoxetine, FR-260,010,



mesulergine, MK-212,
LU AA24530, methysergide,



naphthyllisopropylamine,
nefazodone, norfluoxetine,



norfenfluramine, ORG-12,962, ORG-
O-desmethyltramadol, RS-102,221,



37,684, oxaflozane, PNU-22395,
SB-200,646, SB-221,284, SB-242,084,



PNU-181731, lysergamides,
SDZ SER-082, tramadol, and



phenethylamines, piperazines,
trazodone



tryptamines, Ro60-0175,




vabicaserin, WAY-629,




WAY-161,503,




WAY-163,909, and YM-348



5-HT2A/2C

ketanserin, risperidone, trazodone,




mirtazapine, clozapine


5-HT3
2-methyl-5-HT, alpha-
dolasetron, granisetron, ondansetron,



methyltryptamine, bufotenin,
palonosetron, tropisetron, alosetron,



chlorophenylbiguanide, ethanol,
cilanosetron, mirtazapine, AS-8112,



ibogaine, phenylbiguanide,
bantopride, metroclopramide,



quipazine, RS-56812, SR-57227,
renzapride, zacopride, mianserin,



varenicline, and YM-31636
vortioxetine, clozapine, olanzapine,




quetiapine, menthol, thujone,




lamotigrine, and 3-tropanyl




indole-3-carboxylate


5-HT4
cisapride, tegaserod, prucalopride,
piboserod, GR-113,808, GR-



BIMU-8, CJ-033,466, ML-10302,
125,487, RS-39604, SB-203,186,



mosapride, renzapride, RS-67506,
SB-204,070, and chamomile



RS-67333, SL65.1055, zacopride,




metoclopramide, and sulpride



5-HT5A
valeronic acid
ASP-5736, AS-2030680,




AS-2674723, latrepiridine,




risperidone,




and SB-699,551


5-HT6
EMDT, WAY-181,187, WAY-
ALX-1161, AVN-211, BVT-5182,



208,466, N-(inden-5-
BVT-74316, cerlapiridine, EGIS-



yl)imidazothiazole-
12233, idalopiridine, interpridine,



5-sulfonamide,
latrepiridine, MS-245, PRX-07034,



E-6837, E-6801, and
SB-258,585, SB-271,046, SB-



EMD-386,088
357,134, SB-339,885, Ro 04-6790,




Ro-4368554, sertindole, olanzapine,




asenapine, clozapine, rosa rugosa




extract, and WAY-255315


5-HT7
AS-19, 5-CT, 5-MeOT,
amisulpride, amitriptyline,



8-OH-DAPT,
amoxapine, clomipramine, clozapine,



aripiprazole, E-55888,
DR-4485, fluphenazine, fluperlapine,



E-57431, LP-12,
ICI 169,369, imipramine, ketanserine,



LP-44, MSD-5a, RA-7, and
JNJ-18038683, loxapine, lurasidone,



N,N-Dimethyltryptamine
LY-215,840, maprotiline,




methysergide, mesulergine,




mianserin, olanzepine, pimozide,




ritanserin, SB-258,719, SB-258,741,




SB-269,970, SB-656,104-A, SB-




691,673, sertindole, spiperone,




tenilapine, TFMPP, vortioxetine,




trifluoperazine, ziprasidone,




and zotepine


Non-selective

chlorpromazine, cyproheptadine,


5-HT

pizotifen, oxetorone, spiperone,


antagonists

ritanserin, parachlorophenylalanine,




metergoline, propranolol, mianserin,




carbinoxamine, methdilazine,




promethazine, pizotifen, oxatomide,




feverfew, fenclonin, and reserpine
















TABLE 7H







GLUATAMATE RECEPTOR AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Ionotropic
AMPA, glutamic acid,
AP5, AP7, CPPene, selfotel,


(GRIA-14,
ibotenic acid,
HU-211, Huperzine A,


GRIK1-5,
kainic acid,
gabapentin,


and
NMDA,
remacemide, amantadine,


GRIN1-3B)
quisqualic acid
atomoxetine, AZD6765,




agmatine, chloroform,




dextrallorphan,




dextromethorphan,




dextrorphan,




diphenidine,




dizocilpine (MK-801),




ethanol, eticyclidine,




gacyclidine,




ibogaine, ifenprodil,




ketamine, kynurenic




acid, memantine,




magnesium, methoxetamine,




nitromemantine, nitrous




oxide, PD-137889,




perampanel, phencyclidine,




rolicyclidine, tenocyclidine,




methoxydine, tiletamine,




neramexane, eliprodil,




etoxadrol,




dexoxadrol, WMS-2539,




NEFA, delucemine,




8A-PDHQ, aptiganel,




rhynchophylline


Metabotropic
L-AP4, ACPD, L-QA,
AIDA, fenobam, MPEP,


(GRM1-8)
CHPG, LY-379,268,
LY-367,385, EGLU,



LY-354,740, ACPT,
CPPG, MAP4, MSOP,



VU0155041
LY-341,495


Glycine

rapastinel, NRX-1074, 7-


antagonists

chlorokynurenic acid, 4-




chlorokynurenine, 5,7-




dichlorokynurenic acid,




kynurenic acid, TK-40,




1-aminocyclopropanecarboxylic




acid (ACPC),




L-phenylalanine, and xenon
















TABLE 7I







HISTAMINE AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Non-selective
histamine dihydrochloride, HTMT




dimaleate, 2-pyridylethlyamine




dihydrochloride



H1

acrivastine, azelastine, astemizole,




bilastine, bromodiphenhydramine,




brompheniramine, buclizine,




carbinoxamine, cetirizine, cetirizine




dihydrochloride, clemastine fumarate,




clemizole hydrochloride,




chlorodiphenhydramine,




chlorphenamine, chlorpromazine,




clemastine, cyclizine, cyproheptadine,




dexbrompheniramine,




dexchlorpheniramine, dimenhydrinate,




dimethindene maleate, dimetindene,




diphenhydramine, diphenhydramine




hydrochloride, doxepin hydrochloride,




doxylamine, ebastine, embramine,




fexofenadine, fexofenadine




hydrochloride, hydroxyzine, ketotifen




fumarate, loratadine, meclizine,




meclizine dihydrochloride,




mepyramine maleate, mirtazapine,




olopatadine, olopatadine




hydrochloride, orphenadrine,




phenindamine, pheniramine,




phenyltoloxamine, promethazine,




quetiapine, rupatadine, terfenadine,




tripelennamine, zotepine, trans-




triprolidine hydrochloride, and




triprolidine


H1 inverse

cetirizine, levocetirizine,


agonists

desloratadine, and pyrilamine


H2
betazole, impromidine, dimaprit
aminopotentidine, cimetidine,



dihydrochloride, and amthamine
famotidine, ICI 162,846, lafutidine,



dihyrdobromide
nizatidine, ranitidine, ranitidine




hyrdochloride, roxatidine, zolantadine




dimaleate, and toitidine


H3
imetit dihydropbromide, immepip
clobenpropit, clobenpropit



dihyrdrobromide, immethridine
dihydrobromide, A 3314440



dihydrobromide,
dihyrdochloride, BF 2649



α-Methylhistamine
hydrochloride, carcinine



dihydrobromide,
ditrifluoroacetate, ABT-239, ciprofaxin,



N-methylhistamine
conessine, GT 2016, A-349,821,



dihydrochloride,
impentamine dihydrobromide,



proxyfan oxalate,
iodophenpropit dihydrobromide, JNJ



and betahistine
10181457 dihydrochloride, JNJ




5207852 dihydrochloride, ROS 234




dioxalate, SEN 12333, VUF 5681




dihydrobromide, and thioperamide


H4
imetit dihydropbromide, immepip
thioperamide, JNJ 7777120, A 943931



dihyrdrobromide, 4-methylhistamine
dihydrochloride, A 987306, JNJ



dihydrochloride, clobenpropit
10191584 maleate, and VUF-6002



dihydrobromide, VUF 10460, and




VUF 8430 dihydrobromide
















TABLE 7J







CANNABINOID AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





Cannabinoid receptor
Anandamide, N-Arachidonoyl



(non-selective)
dopamine, 2-Arachidonoylglycerol




(2-AG), 2-Arachidonyl glyceryl ether,




Δ-9-Tetrahydrocannabinol, EGCG,




Yangonin, AM-1221, AM-1235, AM-




2232, UR-144, JWH-007, JWH-015,




JWH-018, ACEA, ACPA, arvanil, CP




47497, DEA, leelamine,




methanandamide, NADA, noladin




ether, oleamide, CB 65, GP-1a, GP-




2a, GW 405833, HU 308, JWH-133,




L-759,633, L-759,656, LEI 101, MDA




19, and SER 601



CB1 receptor
ACEA, ACPA, RVD-Hpα, (R)-(+)-
rimonabant, cannabidiol, Δ9-



methanandamide
tetrahydrocannabivarin (THCV),




taranabant, otenabant, surinabant,




rosonabant, SLV-319, AVE1625,




V24343, AM 251, AM 281, AM 6545,




hemopressin, LY 320135, MJ 15, CP




945598, NIDA 41020, PF 514273, SLV




319, SR 1141716A, and TC-C 14G


CB2 receptor
CB 65, GP 1a, GP 2a, GW 405833,
cannabidiol, Δ9-tetrahydrocannabivarin



HU 308, JWH 133, L-759,656, L-
(THCV), AM 630, COR 170, JTE 907,



759,633, SER 601, LEI 101
and SR 144528
















TABLE 7K







PURINERGIC RECEPTOR AGONISTS AND ANTAGONISTS









Receptor
Agonist
Antagonist





ADORA1 (P1
Adenosine, N6-
Caffeine, theophylline, 8-Cyclopentyl-


adenosine receptor)
Cyclopentyladenosine, N6-3-
1,3-dimethylxanthine (CPX), 8-



methoxy1-4-hydroxybenzyl adenine
Cyclopenty1-1,3-dipropylxanthine



riboside (B2), CCPA, tecadenoson,
(DPCPX), 8-Pheny1-1,3-



selodenoson, Certain
dipropylxanthine, bamifylline, BG-9719,



Benzodiazepines and Barbiturates,
BG09928, FK-453, FK838, rolofylline,



2′-MeCCPA, GR 79236, and SDZ
N-0861, and PSB 36



WAG 994



ADORA2A (P1
Adenosine, N6-3-methoxyl-4-
Caffeine, theophylline, istradefylline,


adenosine receptor)
hydroxybenzyl adenine riboside (B2),
SCH-58261, SCH-442,416, ATL-444,



YT-146, DPMA, UK-423,097,
MSX-3, preladenant, SCH-412,348,



limonene, NECA, CV-3146,
VER-6623, VER-6947, VER-7835,



binodenoson, ATL-146e, CGS-
vipadenant, and ZM-241,385



21680, and Regadenoson



ADORA2B (P1
Adenosine, 5′-N-
Caffeine, theophylline, CVT-6883, ATL-


adenosine receptor)
ethylcarboxamidoadenosine, BAY
801, compound 38, MRS-1706, MRS-



60-6583, LUF-5835, NECA, (S)-
1754, OSIP-339,391, PSB-603, PSB-



PHPNECA, and LUF-5845
0788, and PSB-1115


ADORA3 (P1
Adenosine, 2-(1-HexynyI)-N-
Caffeine, theophylline, MRS-1191,


adenosine receptor)
methyladenosine, CF-101 (IB-
MRS-1220, MRS-1334, MRS-1523,



MECA), CF-102, 2-Cl-IB-MECA, CP-
MRS-3777, MRE3008F20,



532,903, inosine, LUF-6000, and
MRE3005F20, OT-7999, SSR161421,



MRS-3558
KF-26777, PSB-10, PSB-11, and VUF-




5574


P2Y receptor
ATP, ADP, UTP, UDP, UDP-
clopidogrel, elinogrel, prasugrel,



glucose, 2-methylthioladenosine 5′
ticlopidine, ticagrelor, AR-C 118925XX,



diphosphate (2-MeSADP),
AR-C 66096, AR-C 69931, AZD 1283,



lysophosphatidic acid, PSB 1114,
MRS 2179, MRS 2211, MRS 2279,



PSB 0474, NF 546, MRS 2365, MRS
MRS 2500, MRS 2578, NF 157, NF



2690, MRS 2693, MRS 2768, MRS
340, PPADS, PPTN hydrochloride,



2905, MRS 2957, MRS 4062, and
PSD 0739, SAR 216471, and suramin



denufosol (P2Y2 agonist)



P2X receptor
ATP
A 438079, A 740003, A 804598, A




839977, AZ 10606120, AZ 11645373,




5-BDBD, BX 430, Evans Blue, JNJ




47965567, KN-62, NF 023, NF 110, NF




157, NF 279, NF 449, PPADS, iso-




PPADS, PPNDS, Ro 0437626, Ro 51,




RO-3, TC-P 262, suramin, TNP-ATP,




and P2X7 antagonists NF279,




calmidazolium, and KN-62
















TABLE 8







NEUROTRANSMISSION MODULATORS








Type
Modulators





Norepinephrine reuptake
amedalin, atomoxetine, CP-39,332, daledalin,


inhibitors (increase
edivoxetine, esreboxetine,


adrenergic
lortalamine, nisoxetine, reboxetine, talopram,


neurotransmission)
talsupram, tandamine, viloxazine,



bupropion, ciclazindol, manifaxine,



maprotiline, radafaxine, tapentadol, teniloxazine,



protriptyline, nortriptyline, and desipramine


Norepineprhine-dopamine
amineptine, bupropion, desoxypipradrol,


reuptake inhibitors
dexmethylphenidate, difemetorex,


(increase
diphenylprolinol, ethylphenidate, fencamfamine,


adrenergic and dopamine
fencamine, lefetamine, methylenedioxy-


neurotransmission)
pyrovalerone, methylphenidate, nomifensine,



O-2172, oxolinic acid, pipradrol, prolintane,



pyrovalerone, tametraline, and WY-46824


Serotonin-
mazindol, nefazodone, sibutramine, venlafaxine,


norepinephrine-
esketamine, duloxetine, ketamine, phencyclidine,


dopamine reuptake
tripelennamine, mepiprazole, amitifadine,


inhibitors (SNDRIs)
AN788, ansofaxine, centanafadine, atomoxetine,


and serotonin-
desvenlafaxine, milnacipran, levomilnacipran,


norepinephrine
dasotraline, Lu AA34893, Lu AA37096,


reuptake
NS-2360, tedatioxetine, tesofensine, bicifadine,


inhibitors (SNRIs)
BMS-866,949, brasofensine, diclofensine, DOV-


(increase adrengergic,
216,303, EXP-561, liafensine, NS-2359, RG-


dopamine, and
7166, SEP-227,162, SEP-228,425, SEP-228,432,


serotonin
naphyrone, 3,3-Diphenylcyclobutanamine, 3,4-


neurotransmission)
Dichlorotametraline, D-161, desmethylsertraline,



DMNPC, DOV-102,677, fezolamine,



GSK1360707F, indatraline, JNJ-7925476, JZ-IV-



10, JZAD-IV-22, LR-5182, methylnaphthidate,



MI-4,PRC200-SS, PRC050, PRCO25, SKF-83,



959, TP1, phenyltropanes (e.g., WF-23,



dichloropane, and RTI-55), Ginkgo biloba



extract, St John's Wort, hyperforin,



adhyperforin, and uliginosin B


Dopamine reuptake
Dopamine reuptake inhbiitors (e.g., altropane,


inhibitors
amfonelic acid, amineptine, BTCP, 3C-PEP,


(increase dopamine
DBL-583, difluoropine, GBR-12783, GBR-


neurotransmission)
12935, GBR-13069, GBR-13098, GYKI-52895,



lometopane, methylphenidate, ethylphenidate,



modafinil, armodafinil, RTI-229, vanoxerine,



adrafinil, benztropine, bupropion, fluorenol,



medifoxamine, metaphit, rimcazole, venlafaxine,




Chaenomeles speciosa, and oroxylin A), dopa-




mine releasing agents (e.g., p-Tyramine),



dextroamphetamine, lisdexamfetamine,



dexmethylphenidate, and cathinone


Dopamine prodrugs
Levopoda, docarpamine


(increase dopamine



neurotransmission)



GABA reuptake inhibitors
CL-996, deramciclane, gabaculine, guvacine,


(increase GABA
nipecotic acid, NNC-711, NNC 05-2090, SKF-


neurotransmission)
89976A, SNAP-5114, tiagabine, and hyperforin


GABA analogs
gabapentin, butyric acid, valproic acid,


(increase GABA
valpromide, valnoctamide, 3-hydroxybutanal,


neurotransmission)
GHB, sodium, oxybate, aceburic acid, GBL,



GHBAL, GHV, GVL, GHC, GCL, HOCPCA,



UMB68, pregabalin, tolibut, phaclofen, sacolfen,



arecaidine, gaboxadol, isonipecotic acid, 3-



Methyl-GABA, AABA, BABA, DAVA, GAVA,



Glutamic acid, hopantenic acid, piracetam, and



vigabatrin


GABA prodrugs
L-Glutamine, N-Isonicotinoyl-GABA,


(increase GABA
picamilon, progabide, tolgabide


neurotransmission)



Acetylcholinesterase
carbamates, physostigmine, neostigmine,


inhibitors (increase
pyridostigmine, ambenonium, demecarium,


nicotinic and
rivastigmine, phenanthrene derivatives,


muscarinic
galantamine, caffeine, rosmarinic acid, alpha-


neurotransmission)
pinene, piperidines, donepezil, tacrine,



edrophonium, Huperzine A, ladostigil,



ungeremine, lactucopicrin, dyflos, echothiophate,



parathion, and quasi-irreversible



acetylcholinesterase inhibitors


Serotonin reuptake
alaproclate, cericlamine, citalopram, dapoxetine,


inhibitors (increase
escitalopram, femoxetine, fluoxetine, fluvoxa-


serotonin
mine, ifoxetine, indalpine, omiloxetine, panura-


neurotransmission)
mine, paroxetine, pirandamine, RTI-353, sertra-



line, zimelidine, desmethylcitalopram,



didesmethylcitalopram, seproxetine ((S)-



norfluoxetine), desvenlafaxine, cianopramine,



litoxetine, lubazodone, SB-649,915, trazodone,



vilazodone, vortioxetine, dextromethorphan,



dextropropoxyphene, dimenhydrinate,



diphenhydramine, mepyramine (pyrilamine),



mifepristone, delucemine, mesembrenone,



mesembrine, roxindole, duloxetine,



levomilnacipran, milnacipran, dapoxetine,



sibutramine, chlorpheniramine,



dextropmethorphan, and methadone


Serotonin releasing agents
chlorphentermine, cloforex, dexfenfluramine,


(increase serotonin
etolorex, fenfluramine, flucetorex, indeloxazine,


neurotransmission)
levofenfluramine, tramadol, carbamazepine,



amiflamine (FLA-336), viqualine (PK-5078), 2-



Methyl-3,4-methylenedioxyamphetamine (2-



Methyl-MDA), 3-Methoxy-4-methylampheta-



mine (MMA), 3-Methyl-4,5-methylenedioxyam-



phetamine (5-Methyl-MDA), 3,4-Ethylenedioxy-



N-methylamphetamine (EDMA), 4-Methoxyam-



phetamine (PMA), 4-Methoxy-N-ethylampheta-



mine (PMEA), 4-Methoxy-N-methylampheta-



mine (PMMA), 4-Methylthioamphetamine (4-



MTA), 5-(2-Aminopropyl)-2,3-dihydrobenzo-



furan (5-APDB), 5-Indanyl-2-aminopropane



(IAP), 5-Methoxy-6-methylaminoindane



(MMAI), 5-Trifluoromethyl-2-aminoindane



(TAI), 5,6-Methylenedioxy-2-aminoindane



(MDAI), 5,6-Methylenedioxy-N-methyl-2-



aminoindane (MDMAI), 6-Chloro-2-



aminotetralin (6-CAT), 6-Tetralinyl-2-aminopro-



pane (TAP), 6,7-Methylenedioxy-2-aminotetralin



(MDAT), 6,7-Methylenedioxy-N-methyl-2-



aminotetralin (MDMAT), N-Ethyl-5-trifluoro-



methyl-2-aminoindane (ETAI), N-Methyl-5-



indanyl-2-aminopropane, aminorex, MDMA,



MDEA, MDA, MBDB, and tryptamines,



such as DMT, αMT, 5MeO-NMT, NMT,



NETP, Dimethyl-Serotonin, 5MeO-



NET, αET and αMT


Excitatory amino acid
didydrokanic acid, WAY-213,613, L-trans-2,4-


reuptake inhibitors
PDC, amphetamine, and L-Theanine


(increase Glutamate



receptor



neurotransmission)



Glycine reuptake
bitopertin, Org 24598, Org 25935, ALX-5407,


inhibitors (increase
sacrosine, Org 25543, and N-


Glutamate receptor
arachidonylglycerine


neurotransmission)



Histidine decarboxylase
Tritoqualine, catechin


inhibitors (decrease



histamine



neurotransmission)



Endocannabinoid
AM404, fatty acid amide hydrolase inhibitors


enhancers
(e.g., AM374, ARN2508, BIA 10-2472, BMS-


(increase cannabinoid
469908, CAY-10402, JNJ-245, JNJ-1661010,


neurotransmission)
JNJ-28833155, JNJ-40413269, JNJ-42119779,



JNJ-42165279, MK-3168, MK-4409, MM-



433593, OL-92, OL-135, PF-622, PF-750, PF-



3845, PF-04457845, PF-04862853, RN-450,



SA-47, SA-73, SSR-411298, ST-4068, TK-25,



URB524, URB597, URB694, URB937, VER-



156084, and V-158866


Monoacylglycerol lipase
N-arachidonoyl maleimide, JZL184


inhibitors (increase



cannabinoid



neurotransmission)



Endocannabinoid
SB-FI-26


transporter



inhibitors(increase



cannabinoid



neurotransmission)



Endocannabinoid
AM404, AM1172, LY-2183240, O-2093,


reuptake
OMDM-2, UCM-707, VDM-11, guineensine,


inhibitors (increase
ETI-T-24_B_I, WOBE437, and RX-055


cannabinoid



neurotransmission)



Adenosine uptake
cilostazol, dilazep, and dipyramidole


inhibitors



(increase purinergic



neurotransmission)



Nucleoside transporter
8MDP, Decynium 22, 5-iodotubercidin,


inhibitors
NBMPR, and TC-T 6000


(increase purinergic



neurotransmission)









In some embodiments, the neurotransmission blocker is a neurotoxin listed in Table 9, or a functional fragment or variant thereof. Neurotoxins include, without limitation, convulsants, nerve agents, parasympathomimetics, and uranyl compounds. Neurotoxins may be bacterial in origin, or fungal in origin, or plant in origin, or derived from a venom or other natural product. Neurotoxins may be synthetic or engineered molecules, derived de novo or from a natural product. Suitable neurotoxins include but are not limited to botulinum toxin and conotoxin. Exemplary neurotoxins are listed in Table 9.









TABLE 9





NEUROTOXINS


NEUROTOXINS
















2,4,5-Trihydroxyamphetamine
Grayanotoxin


2,4,5-Trihydroxymethamphetamine
Hainantoxin


3,4-Dichloroamphetamine
Halcurin


5,7-Dihydroxytryptamine
Hefutoxin


5-Iodowillardiine
Helothermine


Ablomin
Heteroscodratoxin-1


Aconitine
Histrionicotoxin


Aconitum
Homoquinolinic acid


Aconitum anthora
Hongotoxin


AETX
Huwentoxin


Agelenin
Ibotenic acid


Agitoxin
Ikitoxin


Aldrin
inhibitor cystine knot


Alpha-Methyldopamine
Jingzhaotoxin


Alpha-neurotoxin
Kainic acid


Altitoxin
Kaliseptine


Anatoxin-a
Kappa-bungarotoxin


Androctonus australis hector
Kodaikanal mercury poisoning


insect toxin



Anisatin
Kurtoxin


Anthopleurin
Latrotoxin


Antillatoxin
Lq2


Anuroctoxin
Maitotoxin


Apamin
Margatoxin


Arum italicum
Maurotoxin


Arum maculatum
Mercury (element)


Babycurus toxin 1
Methanol


Batrachotoxin
Methiocarb


BDS-1
MPP+


Bestoxin
MPTP


Beta-Methylamino-L-alanine
Nemertelline


BgK
Neosaxitoxin


Birtoxin
Nicotine


BmKAEP
N-Methylconiine


BmTx3
Oenanthotoxin


BotIT2
Oxalyldiaminopropionic acid


BotIT6
Oxidopamine


Botulinum toxin
Oxotoxin


Brevetoxin
Pahutoxin


Bukatoxin
Palytoxin


Butantoxin
Pandinotoxin


Calcicludine
Para-Bromoamphetamine


Calciseptine
Para-Chloroamphetamine


Calitoxin
Para-Chloromethamphetamine


Caramboxin
Para-Iodoamphetamine


Carbon disulfide
Penitrem A


CgNa toxin
Phaiodotoxin


Charybdotoxin
Phenol


Cicutoxin
Phoneutria nigriventer toxin-3


Ciguatoxin
Phrixotoxin


Cll1
Polyacrylamide


Clostridium botulinum
Poneratoxin


Conantokins
Psalmotoxin


Conhydrine
Pumiliotoxin


Coniine
Quinolinic acid


Conotoxin
Raventoxin


Contryphan
Resiniferatoxin


CssII
Samandarin


CSTX
Saxitoxin


Curare
Scyllatoxin


Cyanide poisoning
Sea anemone neurotoxin


Cylindrospermopsin
Slotoxin


Cypermethrin
SNX-482


Delta atracotoxin
Stichodactyla toxin


Dendrotoxin
Taicatoxin


Dieldrin
Taipoxin


Diisopropyl fluorophosphates
Tamapin


Dimethylmercury
Tertiapin


Discrepin
Tetanospasmin


Domoic acid
Tetraethylammonium


Dortoxin
Tetramethylenedisulfotetramine


DSP-4
Tetrodotoxin


Ergtoxin
Tityustoxin


Falcarinol
Tricresyl phosphate


Fenpropathrin
TsIV


Gabaculine
Vanillotoxin


Ginkgotoxin
Veratridine


Grammotoxin









Antibodies


Neurotransmission modulators also include antibodies that bind to neurotransmitters or neurotransmitter receptors listed in Tables 5 and 6 and decrease neurotransmission. These antibodies include blocking and neutralizing antibodies. Antibodies to neurotransmitters or neurotransmitter receptors listed in Tables 5 and 6 can be generated by those of skill in the art using well established and routine methods.


Neuronal Growth Factor Blocker


In some embodiments, the P2RX2 inhibitor is administered with a neuronal growth factor blocker (e.g., an agent that decreases neurogenic/axonogenic signals, e.g., an antagonist of a neurotrophic factor, neuronal growth factor, or neuronal growth factor receptor). For example, the neuronal growth factor blocker is an antagonist of a neuronal growth factor or neuronal growth factor receptor listed in Table 10. A neuronal growth factor blocker may decrease neurogenesis, neuronal growth, neuronal differentiation, neurite outgrowth, synapse formation, synaptic maturation, synaptic refinement, or synaptic stabilization. Neuronal growth factor blockers decrease tissue innervation (e.g., innervation of a tumor) and the formation of synaptic connections between two or more neurons and between neurons and non-neural cells. A neuronal growth factor blocker may block one or more of these processes (e.g., through the use of antibodies that block neuronal growth factors or their receptors or inhibitory RNAs directed to neuronal growth factors or their receptors). Neuronal growth factor blockers can decrease one of the above-mentioned processes by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, 200%, 500% or more.


In some embodiments, the neuronal growth factor blocker decreases neurogenic/axonogenic signals, e.g., the method includes administering to the subject or contacting a cell with a neuronal growth factor blocker in an amount and for a time sufficient to decrease neurogenesis, axonogenesis, or innervation. For example, the neuronal growth factor blocker that leads to a decrease in neurogenesis or axonogenesis is a blocking or neutralizing antibody against a neurotrophic factor. Relevant neurotrophic factors include NGF, BDNF, ProNGF, Sortilin, TGFβ and TGFβ family ligands and receptors (e.g., TGFβR1, TGFβR2, TGFβ1, TGFβ2 TGFβ4), GFRα family ligands and receptors (e.g., GFRα1, GFRα2, GFRα3, GFRα4, GDNF), CNTF, LIF, neurturin, artemin, persephin, neurotrophin, chemokines, cytokines, and others listed in Table 10. Receptors for these factors can also be targeted, as well as downstream signaling pathways including Jak-Stat inducers, and cell cycle and MAPK signaling pathways. In some embodiments, the neuronal growth factor blocker decreases neurogenesis, axonogenesis or any of the processes mentioned above by sequestering, blocking, antagonizing, degrading, or downregulating a neuronal growth factor or a neuronal growth factor receptor listed in Table 10. In some embodiments, the neuronal growth factor blocker decreases neurogenesis, axonogenesis or any of the processes mentioned above by blocking or antagonizing a signaling protein that is downstream of a neuronal growth factor. In some embodiments, the neuronal growth factor blocker decreases neurogenesis, axonogenesis or any of the processes mentioned above by blocking, disrupting, or antagonizing a synaptic or structural protein. Neurogenesis, axonogenesis, neuronal growth, neuronal differentiation, neurite outgrowth, synapse formation, synaptic maturation, synaptic refinement, synaptic stabilization, or tissue innervation can be decreased in the subject at least 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, 70%, 80% or more, compared to before the administration. Neurogenesis, axonogenesis, neuronal growth, neuronal differentiation, neurite outgrowth, synapse formation, synaptic maturation, synaptic refinement, synaptic stabilization, or tissue innervation can be decreased in the subject between 5-20%, between 5-50%, between 10-50%, between 20-80%, between 20-70%. Neuronal growth factor blockers can be administered in any of the modalities described herein (e.g., antibody, small molecule, nucleic acid, polypeptide, or viral vector).


In some embodiments, the neuronal growth factor blocker decreases the number of nerves in an affected tissue. For example, the subject has cancer (e.g., the subject has a highly innervated tumor). For example, the neuronal growth factor blocker is administered in an amount and for a time sufficient to decrease neurogenesis/axonogenesis.


Neuronal growth factor blockers include antibodies that bind to neuronal growth factors or neuronal growth factor receptors and decrease their signaling (e.g., blocking antibodies). Exemplary neuronal growth factor blocking antibodies are listed below in Table 11. Antibodies to neuronal growth factors listed in Table 10 can also be generated by those of skill in the art using well established and routine methods.









TABLE 10







NEURONAL GROWTH FACTORS














Accession
Entrez



Gene
Type
Number
Gene ID
















ARTN
Ligand
Q5T4W7
9048



BDNF
Ligand
P23560
627



BDNF-AS
Ligand

497258



BEX1
Signaling
Q9HBH7
55859



BEX3
Signaling
Q00994
27018



CD34
Receptor
P28906
947



CDNF
Ligand
Q49AH0
441549



CNTF
Ligand
P26441
1270



CNTFR
Receptor
P26992
1271



CRLF1
Receptor
O75462
9244



CSPG5
Ligand
O95196
10675



DCLK1
Signaling
O15075
9201



DISC1
Signaling
Q9NRI5
27185



DNAJC5
Signaling
Q9H3Z4
80331



DPYSL2
Signaling
Q16555
1808



DVL1
Signaling
O14640
1855



EFNA5
Ligand
P52803
1946



EGR3
Signaling
Q06889
1960



ENO2
Signaling
P09104
2026



EphA1
Receptor
P21709
2041



EphA10
Receptor
Q5JZY3
284656



EphA2
Receptor
P29317
1969



EphA3
Receptor
P29320
2042



EphA4
Receptor
P29317
2043



EphA5
Receptor
P54756
2044



EphA6
Receptor
Q9UF33
285220



EphA7
Receptor
Q15375
2045



EphA8
Receptor
P29322
2046



EphB1
Receptor
P54762
2047



EphB2
Receptor
P29323
2048



EphB3
Receptor
P54753
2049



EphB4
Receptor
P54760
2050



EphB6
Receptor
O15197
2051



ETBR2
Receptor
O60883
9283



FSTL4
Receptor
Q6MZW2
23105



GDNF
Ligand
P39905
2668



GFRA1
Receptor
P56159
2674



GFRA2
Receptor
O00451
2675



GFRA3
Receptor
O60609
2676



GFRA4
Receptor
Q9GZZ7
64096



GPR37
Receptor
O15354
2861



GPRIN1
Signaling
Q7Z2K8
114787



GPRIN2
Signaling
O60269
9721



GPRIN3
Signaling
Q6ZVF9
285513



GRB2
Signaling
P62993
2885



GZF1
Signaling
Q9H116
64412



IFNA1
Ligand
P01562
3439



IGF1
Ligand
P05019
3479



IGF2
Ligand
P01344
3481



IL11RA
Receptor
Q14626
3590



IL1B
Ligand
P01584
3553



IL3
Ligand
P08700
3562



IL4
Ligand
P05112
3565



IL6
Ligand
P05231
3569



IL6R
Receptor
P08887
3570



IL6ST
Signaling
P40189
3572



INS
Ligand
P01308
3630



L1CAM
Signaling
P32004
3897



LIF
Ligand
P15018
3976



LIFR
Receptor
P42702
3977



MAGED1
Signaling
Q9Y5V3
9500



MANF
Ligand
P55145
7873



NDNF
Ligand
Q8TB73
79625



NENF
Ligand
Q9UMX5
29937



NENFP1
Ligand

106480294



NENFP2
Ligand

100129880



NENFP3
Ligand

106481703



NGF
Ligand
P01138
4803



NGFR
Receptor
P08138
4804



NRG1
Ligand
Q02297
3084



NRP1
Receptor
O14786
8829



NRTN
Ligand
Q99748
902



NTF3
Ligand
P20783
4908



NTF4
Ligand
P34130
4909



NTRK1
Receptor
P04629
4914



NTRK2
Receptor
Q16620
4915



NTRK3
Receptor
Q16288
4916



PDPK1
Signaling
Q15530
5170



PEDF
Ligand
P36955
5176



PLEKHH3
Signaling
Q7Z736
79990



PSAP
Ligand
P07602
5660



PSEN1
Signaling
P49768
5663



PSPN
Ligand
O70300
5623



PTN
Ligand
P21246
5764



RELN
Ligand
P78509
5649



RET
Signaling
P07949
5979



ROR1
Receptor
Q01973
4919



ROR2
Receptor
Q01974
4920



RPS6KA3
Signaling
P51812
6197



SDC3
Receptor
O75056
9672



SEMA3E
Ligand
O15041
9723



SERPINE2
Ligand
P07093
5270



SERPINF1
Ligand
P36955
5176



SHC1
Signaling
P51812
6464



SNTG1
Biosynthesis
P07602
54212



S0RCS1
Receptor
O75056
114815



SORCS2
Receptor
O15041
57537



SORCS3
Receptor
P07093
22986



SORT1
Receptor
Q99523
6272



SULF1
Signaling
Q8IWU6
23213



SULF2
Signaling
Q8IWU5
55959



TGFB1
Ligand
P01137
7040



TGFB2
Ligand
P61812
7042



TGFB3
Ligand
P10600
7043



TMEM158
Receptor
Q8WZ71
25907



TNF
Ligand
P01375
7124



TPM3
Receptor
P06753
7170



VEGFA
Ligand
P15692
7422



VEGFB
Ligand
P49765
7423



VGF
Ligand
O15240
7425



XCR1
Receptor
P46094
2829



ZN274
Signaling
Q96GC6
10782

















TABLE 11







NEURONAL GROWTH FACTOR ANTIBODIES









Neuronal




Growth




Factor
Antibody
Company





BDNF
3868 (agonist antibody)
Pfizer


BDNF
29D7 (agonist antibody)
Pfizer


EphA3
KB004
KaloBios Pharmaceuticals,




Inc.


IFNA1
Faralimomab
Creative Biolabs


IFNA1
Sifalimumab (MEDI-545)
MedImmune


IFNA1
Rontalizumab
Genentech


IGF
Figitumumab (CP-751,871) -
Pfizer



an IGR-1R MAb



IGF
SCH717454 (Robatumamab,
Merck



inhibits IGF initiated




phosphorylation)



IGF
Cixutumumab (IGF-1R
Eli Lilly



antibody)



IGF
Teprotumumab (IGF-1R
Genmab/Roche



blocking antibody)



IGF-2
Dusigitumab
MedImmune/AstraZeneca


IGF-2
DX-2647
Dyax/Shire


IGF
Xentuzumab
Boehringer Ingelheim/Eli




Lilly


IGF
Dalotuzumab (IGFR1
Merck & Co.



blocking antibody)



IGF
Figitumumab (IGFR1
Pfizer



blocking antibody)



IGF
Ganitumab (IGFR1
Amgen



blocking antibody)



IGF
Robatumumab (IGFR1
Roche/Schering-Plough



blocking antibody)



IL1B
Canakinumab
Novartis


IL1B
APX002
Apexigen


IL1B
Gevokizumab
XOMA


IL4
Pascolizumab
GlaxoSmithKline


IL4
Dupilumab
Regeneraon/Sanofi


IL6
Siltuximab
Janssen Biotech, Inc.


IL6
Olokizumab
UCB/R-Pharm


IL6
Elsilimomab
Orphan Pharma International


IL6
Sirukumab
Centocor


IL6
Clazakizumab
Bristol Myers Squib/Alder




Biopharmaceuticals


IL6
Gerilimzumab (ARGX-109)
arGEN-X/RuiYi


IL6
FE301
Ferring Pharmaceuticals


IL6
FM101
Femta Pharmaceuticals


IL-6R
Sarilumab (directed against
Regeneron/Sanofi



IL6R)



IL-6R
Tocilizumab
Hoffmann-La Roche/Chugai


IL-6R
Sapelizumab
Chugai


IL-6R
Vobarilizumab
Ablynx


L1CAM
AB417
Creative biolabs


L1CAM
L1-9.3
Creative biolabs


L1CAM
L1-14.10
Biolegend


NGF
Tanezumab
Pfizer


NGF
Fulranumab (JNJ-42160443),
Amgen


NGF
MNAC13 (anti-TrkA, the
Creative Biolabs



NGF receptor)



NGF
mAb 911
Rinat/Pfizer


NGF
Fasinumab
Regeneron/Teva


NRG1
538.24
Hoffman-La Roche


NRP1
Vesencumab
Genentech/Roche


ROR1
Cirmtuzumab
Oncternal Therapeutics


SAP
GSK2398852
GlaxoSmithKline


TGFβ
Fresolimumab (pan-TGFβ
Genzyme/Aventis



antibody)



TGFβ
IMC-TR1 (LY3022859) (MAb
Eli Lilly



against TGFβRII)



TGFβ
TβM1 (anti-TGFβ1 MAb)
Eli Lilly


TGFβ2
Lerdelimumab (CAT-152)
Genzyme


TGFβ1
Metelimumab
Genzyme


TGFβ1
LY2382770
Eli Lilly


TGFβ
PF-03446962 (MAb against
Pfizer



TGFβRI)



TNF
Infliximab
Janssen Biotech, Inc.


TNF
Adalimumab
AbbVie Inc.


TNF
Certolizumab pegol
UCB


TNF
Golimumab
Janssen Biotech, Inc.


TNF
Afelimomab



TNF
Placulumab
Teva Pharmaceutical




Industries, Inc.


TNF
Nerelimomab
Chiron/Celltech


TNF
Ozoralizumab
Pfizer/Ablynx


VEGFA
Bevacizumab
Genentech


VEGFA
Ranibizumab
Genentech


VEGF
Alacizumab pegol (anti-
UCB



VEGFR2)



VEGFA
Brolucizumab
Novartis


VEGF
Icrucumab (anti-VEGFR1)
Eli Lilly


VEGF
Ramucirumab (anti-VEGFR2)
Eli Lilly









Neuronal growth factor blockers also include agents that antagonize neuronal growth factors and neuronal growth factor receptors. For example, neuronal growth factor blockers include TNF inhibitors (e.g., etanercept, thalidomide, lenalidomide, pomalidomide, pentoxifylline, bupropion, and DOI), TGFβ1 inhibitors, (e.g., disitertide (P144)), and TGFβ2 inhibitors (e.g., trabedersen (AP12009)). Exemplary neuronal growth factor antagonists are listed in Table 12.









TABLE 12







NEURONAL GROWTH FACTOR AGONISTS AND ANTAGONISTS










Agonist
Antagonist





TrkA
NGF, amitriptyline, and
ALE-0540



gambogic amide, gambogic acid



TrkB
BDNF, NT3, NT4, 3,7-
ANA-12, cyclotraxin B, and



Dihydroxyflavone, 3,7,8,2-
gossypetin



Tetrahydroxyflavone, 4′-




Dimethylamino-7,8-




dihydroxyflavone, 7,3′-




Dihydroxyflavone, 7,8-




Dihydroxyflavone, 7,8,2′-




Trihydroxyflavone, 7,8,3′-




Trihydroxyflavone, Amitriptyline,




Deoxygedunin, Diosmetin, HIOC,




LM22A-4, N-Acetylserotonin,




Norwogonin (5,7,8-THF), R7,




LM22A4, and TDP6



Pan-Trk receptor

entrectinib (RXDX-101), AG 879,




GNF 5837, GW 441756, and PF




06273340


GFRα1R
GDNF and XIB4035



VEGF receptor

AEE 788, AG 879, AP 24534,




axitinib, DMH4, GSK 1363089, Ki




8751, RAF 265, SU 4312, SU




5402, SU 5416, SU 6668,




sunitinib, toceranib, vatalanib, XL




184, ZM 306416, and ZM 323881


TGFβRI

galunisertib (LY2157299), TEW-




7197, SB-431542, A 83-01, D




4476, GW 788388, LY 364947, R




268712, RepSox, SB 505124, SB




525334, and SD 208









In any of the combination therapy approaches described herein, the first and second therapeutic agent (e.g., a P2RX2 inhibitor described herein and the additional therapeutic agent) are administered simultaneously or sequentially, in either order. The first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours up to 24 hours or up to 1-7, 1-14, 1-21 or 1-30 days before or after the second therapeutic agent.


Diagnosis and Prognosis of P2RX2-Associated Cancer


The methods described herein include methods of diagnosing or identifying patients with P2RX2-associated cancer. Subjects who can be diagnosed or identified as having P2RX2-associated cancer are subjects who have cancer (e.g., subjects identified as having cancer), or subjects suspected of having cancer. Subjects can be diagnosed or identified as having P2RX2-associated cancer based on screening of patient cancer samples (e.g., tumor biopsies). P2RX2 expression can be assessed in a cancer sample isolated from a subject using standard techniques known in the art, such as immunohistochemistry, western blot analysis, quantitative RT-PCR, RNA sequencing, fluorescent in situ hybridization, cDNA microarray, and droplet digital PCR. P2RX2 expression can be assessed by comparing measurements obtained from subject cancer samples to measurements of P2RX2 expression obtained from a reference sample (e.g., a non-cancerous cell of the same type or a cell that does not express P2RX2). Reference samples can be obtained from healthy subjects (e.g., subjects without cancer), or they can be obtained from databases in which average measurements of P2RX2 expression are cataloged for a variety of types of healthy (e.g., non-cancerous) cells from many subjects.


Subjects are diagnosed or identified as having P2RX2-associated cancer if P2RX2 expression is elevated in the cancer sample compared to the reference sample. An increase of P2RX2 expression of 1.1-fold or more (e.g., 1.1, 1.2, 1.3, 1.4, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0-fold or more) in the cancer sample compared to the reference indicates that the subject has P2RX2-associated cancer. Subjects can also be diagnosed or identified as having P2RX2-associated cancer (e.g., a cancer in which P2RX2 is functional) by contacting a cancer cell or tumor sample (e.g., biopsy) isolated from the subject with a P2RX2 agonist (e.g., ATP) and evaluating intracellular calcium using a calcium sensitive dye. An increase in intracellular calcium by 10% or more (e.g., 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99%, or more) indicates that the tumor or cancer cell expresses functional P2RX2 (e.g., is a P2RX2-associated cancer). The tumor sample or cancer cell can then optionally be contacted with a P2RX2 inhibitor or P2RX2-specific inhibitor to determine whether the increase in intracellular calcium by a P2RX2 agonist (e.g., ATP) is reduced or blocked. Subjects diagnosed or identified as having P2RX2-associated cancer can be treated with the methods and compositions described herein (e.g., P2RX2 inhibitors). Subjects can also be selected for treatment with the methods and compositions described herein if the cancer sample from the subject is found to express P2RX2.


The methods described herein also include methods of predicting patient response (e.g., the response of cancer in a subject) to P2RX2 inhibitors in order to determine whether P2RX2 inhibitors can be used for cancer treatment. In some embodiments, a cancer sample (e.g., a tumor biopsy or cancer cell) is isolated from a subject and contacted with one or more P2RX2 inhibitors or P2RX2-specific inhibitors (e.g., cancer samples are cultured and contacted with one or more inhibitors in vitro). The response of the cancer sample to the one or more P2RX2 inhibitors or P2RX2-specific inhibitors is evaluated to predict response to treatment. Responses that are evaluated include cancer cell or tumor growth, cancer cell or tumor proliferation, cancer cell or tumor migration, cancer cell or tumor metastasis, cancer cell or tumor invasion, cancer cell or tumor death, cancer cell or tumor autophagy, or cancer cell or tumor P2RX2 expression. A decrease of at least 5% or more (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99%, or more) in cancer cell or tumor growth, cancer cell or tumor proliferation, cancer cell or tumor migration, cancer cell or tumor metastasis, cancer cell or tumor invasion, or cancer cell or tumor P2RX2 expression in treated cells compared to untreated or control-treated cells, or an increase of at least 5% or more (e.g., 5%, 10%, 15%, 20%, 25%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 99%, or more) in cancer cell or tumor death or cancer cell or tumor autophagy in treated cells compared to untreated or control-treated cells indicates that the cancer would respond to treatment with a P2RX2 inhibitor.


The methods used above to diagnose or identify a subject with P2RX2-associated cancer can also be used to predict patient response (e.g., the response of cancer in a subject) to treatment with a P2RX2 inhibitor. If the expression of P2RX2 is elevated in a cancer sample compared to a reference (e.g., 1.1, 1.2, 1.3, 1.4, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 6.0, 7.0, 8.0, 9.0, 10.0-fold or more higher in the cancer sample compared to the reference), the subject can be predicted to respond to treatment with a P2RX2 inhibitor. Subjects predicted to respond to treatment with a P2RX2 inhibitor or P2RX2-specific inhibitor can be treated using the methods and compositions described herein (e.g., P2RX2 inhibitors).


Methods of Treatment


Administration


An effective amount of a P2RX2 inhibitor described herein for treatment of cancer can be administered to a subject by standard methods. For example, the agent can be administered by any of a number of different routes including, e.g., intravenous, intradermal, subcutaneous, percutaneous injection, oral, transdermal (topical), or transmucosal. The P2RX2 inhibitor can be administered orally or administered by injection, e.g., intramuscularly, or intravenously. The most suitable route for administration in any given case will depend on the particular agent administered, the patient, the particular disease or condition being treated, pharmaceutical formulation methods, administration methods (e.g., administration time and administration route), the patients age, body weight, sex, severity of the diseases being treated, the patient's diet, and the patient's excretion rate. The agent can be encapsulated or injected, e.g., in a viscous form, for delivery to a chosen site, e.g., a tumor site. The agent can be provided in a matrix capable of delivering the agent to the chosen site. Matrices can provide slow release of the agent and provide proper presentation and appropriate environment for cellular infiltration. Matrices can be formed of materials presently in use for other implanted medical applications. The choice of matrix material is based on any one or more of: biocompatibility, biodegradability, mechanical properties, and cosmetic appearance and interface properties. One example is a collagen matrix.


The agent (e.g., P2RX2 inhibitor, e.g., polypeptide, small molecule, nucleic acid, or antibody) can be incorporated into pharmaceutical compositions suitable for administration to a subject, e.g., a human. Such compositions typically include the agent and a pharmaceutically acceptable carrier. As used herein the term “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances are known. Except insofar as any conventional media or agent is incompatible with the active compound, such media can be used in the compositions of the invention. Supplementary active compounds can also be incorporated into the compositions.


A pharmaceutical composition can be formulated to be compatible with its intended route of administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.


Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable carriers include physiological saline, bacteriostatic water, or phosphate buffered saline (PBS). In all cases, the composition must be sterile and should be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a P2RX2 inhibitor described herein) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.


Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.


The active compounds can be prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art.


Nucleic acid molecule agents described herein can be administered directly (e.g., therapeutic mRNAs) or inserted into vectors used as gene therapy vectors. Gene therapy vectors can be delivered to a subject by, for example, intravenous injection, local administration (see U.S. Pat. No. 5,328,470) or by stereotactic injection (see, e.g., Chen et al., PNAS 91:3054 1994). The pharmaceutical preparation of the gene therapy vector can include the gene therapy vector in an acceptable diluent, or can include a slow release matrix in which the gene delivery vehicle is embedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.


The pharmaceutical compositions can be included in a container, pack, or dispenser together with instructions for administration.


Methods of formulating pharmaceutical agents are known in the art, e.g., Niazi, Handbook of Pharmaceutical Manufacturing Formulations (Second Edition), CRC Press 2009, describes formulation development for liquid, sterile, compressed, semi-compressed and OTC forms. Transdermal and mucosal delivery, lymphatic system delivery, nanoparticles, controlled drug release systems, theranostics, protein and peptide drugs, and biologics delivery are described in Wang et al., Drug Delivery: Principles and Applications (Second Edition), Wiley 2016; formulation and delivery of peptide and protein agent is described, e.g., in Banga, Therapeutic Peptides and Proteins: Formulation, Processing, and Delivery Systems (Third Edition), CRC Press 2015.


Local Administration


The P2RX2 inhibitors described herein can be administered locally, e.g., to the site of cancer in the subject. Examples of local administration include epicutaneous, inhalational, intra-articular, intrathecal, intravaginal, intravitreal, intrauterine, intra-lesional administration, lymph node administration, intratumoral administration and administration to a mucous membrane of the subject, wherein the administration is intended to have a local and not a systemic effect. As an example, for the treatment of a cancer described herein, the P2RX2 inhibitor may be administered locally (e.g., intratumorally) in a compound-impregnated substrate such as a wafer, microcassette, or resorbable sponge placed in direct contact with the affected tissue. Alternatively, the P2RX2 inhibitor is infused into the brain or cerebrospinal fluid using standard methods. As yet another example, a pulmonary cancer described herein may be treated, for example, by administering the P2RX2 inhibitor locally by inhalation, e.g., in the form of an aerosol spray from a pressured container or dispenser which contains a suitable propellant, e.g., a gas such as carbon dioxide or a nebulizer. A P2RX2 inhibitor for use in the methods described herein can be administered at the site of a tumor, e.g., intratumorally. In certain embodiments, the agent is administered to a mucous membrane of the subject.


Combination Therapy


The P2RX2 inhibitors described herein may be administered in combination with one or more additional therapies (e.g., 1, 2, 3 or more additional therapeutic agents). The two or more agents can be administered at the same time (e.g., administration of all agents occurs within 15 minutes, 10 minutes, 5 minutes, 2 minutes or less). The agents can also be administered simultaneously via co-formulation. The two or more agents can also be administered sequentially, such that the action of the two or more agents overlaps and their combined effect is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with one agent or treatment delivered alone or in the absence of the other. The effect of the two or more treatments can be partially additive, wholly additive, or greater than additive (e.g., synergistic). Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, local routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination may be administered by intravenous injection while a second therapeutic agent of the combination can be administered locally in a compound-impregnated microcassette. The first therapeutic agent may be administered immediately, up to 1 hour, up to 2 hours, up to 3 hours, up to 4 hours, up to 5 hours, up to 6 hours, up to 7 hours, up to, 8 hours, up to 9 hours, up to 10 hours, up to 11 hours, up to 12 hours, up to 13 hours, 14 hours, up to hours 16, up to 17 hours, up 18 hours, up to 19 hours up to 20 hours, up to 21 hours, up to 22 hours, up to 23 hours up to 24 hours or up to 1-7, 1-14, 1-21 or 1-30 days before or after the second therapeutic agent.


For use in treating cancer, the second agent may be a checkpoint inhibitor, a chemotherapeutic drug, a biologic drug, a non-drug therapy, a neurotransmission blocker, or a neuronal growth factor blocker. In one embodiment, the inhibitor of checkpoint is an inhibitory antibody (e.g., a monospecific antibody such as a monoclonal antibody). The antibody may be, e.g., humanized or fully human. In other embodiments, the inhibitor of checkpoint is a fusion protein, e.g., an Fc-receptor fusion protein. In some embodiments, the inhibitor of checkpoint is an agent, such as an antibody, that interacts with a checkpoint protein. In other embodiments, the inhibitor of checkpoint is an agent, such as an antibody, that interacts with the ligand of a checkpoint protein. In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of CTLA-4 (e.g., an anti-CTLA4 antibody such as ipilimumab or tremelimumab). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of PD-1 (e.g., nivolumab; pembrolizumab; pidilizumab/CT-011). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of PDL1 (e.g., MPDL3280A/RG7446; MEDI4736; MSB0010718C; BMS 936559). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or Fc fusion or small molecule inhibitor) of PDL2 (e.g., a PDL2/Ig fusion protein such as AMP 224). In one embodiment, the inhibitor of checkpoint is an inhibitor (e.g., an inhibitory antibody or small molecule inhibitor) of B7-H3 (e.g., MGA271), B7-H4, BTLA, HVEM, TIM3, GAL9, LAGS, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands, or a combination thereof. The second agent may also be an anti-angiogenic drug, e.g., an anti-VEGF antibody, or the second agent may be an oncolytic agent e.g., a chemotherapy, a drug that targets cancer metabolism, an antibody that marks a cancer cell surface for destruction, e.g., rituximab or trastuzumab, an antibody-drug conjugate, e.g., trastuzumab emtansine, a cell therapy, or other commonly-used anti-neoplastic agent.


Dosing


Subjects that can be treated as described herein are subjects with cancer or at risk of developing cancer. The cancer may be a primary tumor or a metastasized tumor. In some embodiments, the cancer is a P2RX2-associated cancer. Subjects who can be treated with the methods disclosed herein include subjects who have had one or more tumors resected, received chemotherapy or other pharmacological treatment for the cancer, received radiation therapy, and/or received other therapy for the cancer. Subjects who have never previously been treated for cancer can also be treated using the methods described herein.


In some embodiments, the agent is administered in an amount and for a time effective to result in one of (or more, e.g., 2 or more, 3 or more, 4 or more of): (a) reduced tumor size, (b) reduced rate of tumor growth, (c) increased tumor cell death (d) reduced tumor progression, (e) reduced number of metastases, (f) reduced rate of metastasis, (g) reduced tumor migration, (h) reduced tumor invasion, (i) reduced tumor volume, (j) decreased tumor recurrence, (k) increased survival of subject, (l) increased progression free survival of subject.


The methods described herein may include a step of selecting a treatment for a patient. The method includes (a) identifying (e.g., diagnosing) a patient who has cancer or is at risk of developing cancer, and (b) selecting a P2RX2 inhibitor, e.g., a P2RX2 inhibitor described herein, to treat the condition in the patient. In some embodiments, the method includes administering the selected treatment to the subject. In some embodiments, a patient is identified as having cancer based on imaging (e.g., MRI, CT, or PET scan), biopsy, or blood sample (e.g., detection of blood antigen markers, circulating tumor DNA (e.g., by PCR). In some embodiments, a patient is identified as having cancer after presenting with one or more symptoms of a paraneoplastic syndrome (e.g., fever, auto-antibodies directed against nervous system proteins, ataxia, dizziness, nystagmus, difficulty swallowing, loss of muscle tone, loss of fine motor coordination, slurred speech memory loss, vision loss, sleep disturbances, dementia, seizures, dysgeusia, cachexia, anemia, itching, or sensory loss in the limbs). In some embodiments, a patient presents with symptoms of paraneoplastic syndrome and is then identified as having cancer based on imaging (e.g., CT, MRI, or PET scans).


The method may also include (a) identifying (e.g., diagnosing) a patient who has a neoplasm, (b) optionally evaluating the neoplasm for innervation, and (c) selecting a P2RX2 inhibitor (e.g., a P2RX2 inhibitor described herein) to treat the patient if the neoplasm is highly innervated (e.g., if the level of innervation is at least 10% higher (e.g., at least 20%, 30%, 40%, 50%, 60%, 70%, 80% higher) than the level of innervation in control tissue, e.g., non-cancerous tissue of the same subject). Innervation may be measured by staining tissue sections for neural markers e.g., immuno-histochemical staining for tyrosine hydroxylase, vesicular acetylcholine transporter; NGF-Inducible Large External glycoprotein, choline acetyltransferase, parvalbumin, neurofilament protein, Synapsin, synaptophysin, NeuN, NSE, MAP2, Beta III tubulin, 160 kD Neurofilament medium/200 kD Neurofilament Heavy, NSE, PSD93/PSD95, Doublecortin (DCX), c-fos, PSA-NCAM, NeuroD or Beta2, Tau, Calbindin-D28k, Calretinin, Neurofilament Protein (NFP), Glial fibrillary acidic protein (GFAP), S100β, Vimentin and CNPase; or by staining tissue sections with cell-identifying stains, e.g., H&E stain, Nissl Stain, Cresyl violet, Neutral red, Thionine and Toluidine blue, Luxol Fast blue stain, Weigert's Chromium hematoxylin method, Page's iron-eriochrome cyanine R, Dextran Conjugates (Fluorescein, Tetramethylrhodamine, Texas Red, Rhodamine Green), Hydrazides & Biocytins, Isolectin GS-IB4 conjugates, Golgi silver stain, or myelin stain; or by imaging the nervous system, e.g., by MRI, CT, PET, EEG, EMG, Myelogram, or magnetoencephalography. In some embodiments, the neoplasm is selected from: head and neck squamous cell carcinoma, adenoid cystic carcinoma, lymphoma, rhabdomyosarcoma, biliary tract cancer, gastric cancer, pancreatic cancer, prostate cancer, lung cancer, breast cancer, skin cancer (e.g., melanoma), renal cell carcinoma, or colorectal cancer. In some embodiments, the cancer is a cancer listed in Table 4. In some embodiments, the neoplasm is derived from a secretory tissue, glandular tissue, or endocrine or hormonal tissue.


In one embodiment, the method includes (a) identifying (e.g., diagnosing) a patient who has a neoplasm, (b) optionally evaluating the neoplasm for perineural invasion, and (c) selecting a P2RX2 inhibitor to treat the patient if the neoplasm exhibits perineural invasion. In some embodiments, the neoplasm is selected from: head and neck squamous cell carcinoma, adenoid cystic carcinoma, lymphoma, rhabdomyosarcoma, biliary tract cancer, gastric cancer, pancreatic cancer, and prostate cancer.


In one embodiment, the method includes (a) identifying (e.g., diagnosing) a patient who has a neoplasm, (b) optionally evaluating the subject for metastasis to brain or spinal cord, and (c) selecting a P2RX2 inhibitor to treat the patient if the neoplasm exhibits metastasis to brain or spinal cord. In some embodiments, the neoplasm is a lung cancer, breast cancer, skin cancer (e.g., melanoma), lymphoma, renal cell carcinoma, GI tract cancer, prostate cancer, or colorectal cancer.


In one embodiment, the method includes (a) identifying (e.g., diagnosing) a patient who has cancer, (b) optionally evaluating the subject for P2RX2 expression (e.g., overexpression), and (c) selecting a P2RX2 inhibitor to treat the patient if the cancer exhibits P2RX2 expression (e.g., overexpression, e.g., if the patient has P2RX2-associated cancer). In some embodiments, the neoplasm is a melanoma, small cell lung cancer, non-small cell lung cancer, gastric cancer, colorectal cancer, head and neck cancer, ovarian cancer, testicular cancer, thymoma, uterine cancer, kidney cancer, acute myeloid leukemia, diffuse large B-cell lymphoma, prostate cancer, breast cancer, or hepatocellular carcinoma. P2RX2 amplification and/or expression can be measured in a cancer sample collected from a subject using standard techniques known in the art, such as immunohistochemistry, western blot analysis, quantitative RT-PCR, RNA sequencing, fluorescent in situ hybridization, cDNA microarray, and droplet digital PCR. A cancer sample can be evaluated for increased expression and/or amplification of P2RX2 by comparison to a reference sample (e.g., a non-cancerous cell of the same type).


In some embodiments, the method includes administering the selected treatment to the subject.


The method may also include a step of assessing the subject for a parameter of cancer progression or remission, e.g., assessing the subject for one or more (e.g., 2 or more, 3 or more, 4 or more) of: primary tumor size (e.g., by imaging), number of metastases (e.g., by imaging or biopsy), cell death in situ (e.g., by biopsy), blood antigen markers (e.g., by ELISA), circulating tumor DNA (e.g., by PCR), or function of the affected organ (e.g., by a test of circulating enzymes for liver, albuminuria for kidney, lung capacity for lung, etc.).


In some embodiments, the tumor is treated with a P2RX2 inhibitor and a second therapeutic agent. The second therapeutic agent can be selected based on tumor type, tumor tissue of origin, tumor stage, tumor innervation, or mutations in genes expressed by the tumor.


In certain embodiments, a P2RX2 inhibitor administered according to the methods described herein does not have a direct effect on the central nervous system (CNS) or gut. Any effect on the CNS or gut is reduced compared to the effect observed if the P2RX2 inhibitor is administered directly to the CNS or gut. In some embodiments, direct effects on the CNS or gut are avoided by modifying the P2RX2 inhibitor not to cross the BBB, as described herein above, or administering the agent locally to a subject.


Subjects with cancer or at risk of developing cancer are treated with an effective amount of a P2RX2 inhibitor. The methods described herein also include contacting a tumor or cancer cell with an effective amount of a P2RX2 inhibitor. In some embodiments, an effective amount of a P2RX2 inhibitor is an amount sufficient to decrease tumor innervation or nerve activity in a tumor. In some embodiments, an effective amount of a P2RX2 inhibitor is an amount sufficient to treat the cancer or tumor, cause remission, reduce tumor growth, reduce tumor volume, reduce tumor metastasis, reduce tumor invasion, reduce tumor proliferation, reduce tumor migration, or reduce tumor number, reduce P2RX2 expression, reduce P2RX2 copy number, increase cancer cell death, increase time to recurrence, or improve survival.


The P2RX2 inhibitors described herein are administered in an amount (e.g., an effective amount) and for a time sufficient to effect one of the outcomes described above. The P2RX2 inhibitor may be administered once or more than once. The P2RX2 inhibitor may be administered once daily, twice daily, three times daily, once every two days, once weekly, twice weekly, three times weekly, once biweekly, once monthly, once bimonthly, twice a year, or once yearly. Treatment may be discrete (e.g., an injection) or continuous (e.g., treatment via an implant or infusion pump). Subjects may be evaluated for treatment efficacy 1 week, 2 weeks, 1 month, 2 months, 3 months, 4 months, 5 months, 6 months or more following administration of a P2RX2 inhibitor depending on the P2RX2 inhibitor and route of administration used for treatment. Depending on the outcome of the evaluation, treatment may be continued or ceased, treatment frequency or dosage may change, or the patient may be treated with a different P2RX2 inhibitor. Subjects may be treated for a discrete period of time (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months) or until the disease or condition is alleviated, or treatment may be chronic depending on the severity and nature of the disease or condition being treated.


Kits


The invention also features a kit including (a) a pharmaceutical composition including a P2RX2 inhibitor described herein, and (b) instructions for administering the pharmaceutical composition to treat cancer.


EXAMPLES

The following examples are provided to further illustrate some embodiments of the present invention, but are not intended to limit the scope of the invention; it will be understood by their exemplary nature that other procedures, methodologies, or techniques known to those skilled in the art may alternatively be used.


Example 1—Identification of P2RX2 as Essential for Pancreatic Cancer Growth and Proliferation

A library of lentiviral-encoded guide RNAs (gRNA) that individually target neuronal genes was synthesized (Cellecta, Inc.) to obtain a library coverage of six unique gRNAs per gene. Pancreatic cancer cell lines, PANC1, MIAPACA2, and BXPC3, were transduced with a lentiviral-encoded Cas9 nuclease at a high multiplicity of infection (MOI), then transduced with the gRNA lentivirus library at a low MOI of 0.5 to ensure that individual cells received approximately one gRNA. The gRNA library vector also encodes puromycin resistance. One day post-transduction, cells were incubated with puromycin for four days to select for successfully-transduced cells.


Following transduction and selection, 10×106 cells were harvested to serve as the “baseline” population from which the growth effects of individual genes would be compared. For the in vitro arm, 10×106 cells were plated in manufacturer's recommended medium and split twice weekly for four weeks, by re-plating 10×106 cells at each split. For the in vivo subcutaneous arm, 5×106 cells were implanted subcutaneously into NOD-SCID mice, with three mice per replicate, and allowed to grow for four weeks. For the in vivo orthotopic arm, 5×105 cells were implanted orthotopically into the pancreas of NOD-SCID mice, with 10 mice per replicate, and allowed to grow for four weeks.


Following the four weeks of cell/tumor growth, DNA from in vitro and in vivo samples was isolated by tissue homogenization and lysis using a DNA extraction kit (Qiagen DNeasy Blood and Tissue Kit), and concentrated by ethanol precipitation. The DNA samples were amplified by two rounds of PCR using manufacturer's recommended primers and analyzed by next-generation sequencing (BGIAmerica). Sequencing results were analyzed to call hits using a Model-based Analysis of Genome-wide CRISPR-Cas9 Knockout (MAGeCK) algorithm as described by Li W et al., Genome Biology 2014 and Li W et al., Genome Biology 2015. In brief, sequencing reads were normalized to their medians, the variance of read counts for individual gRNAs were estimated and normalized, and individual gRNA read count differences were ranked against each other. Target genes were called based on whether multiple gRNAs targeting a single gene ranked near the top of the gRNA ranking list.


Target genes were quantified along three parameters: the Beta score, essentially the magnitude of the effect (log-fold change in gRNA count); the p-value; and the false discovery rate (FDR). Beta scores <0 indicated that the six gRNAs targeting a single gene were absent from the late stage sample compared to the baseline sample, and were a good indication that the gene was “dropping out” in the course of tumor growth. P-value and FDR both reflected the confidence that the result was not artifactual, with a lower value indicating higher confidence. Thresholds for calling hits were P-value <0.1 and FDR <0.5.


P2RX2 showed up as a significant hit in multiple model systems, as shown in Table 13 below. The data indicate that P2RX2 is essential to the growth and proliferation of pancreatic cancer.









TABLE 13







P2RX2 CRISPR RESULTS











Gene
B-Score
P-Value
FDR
Condition














P2RX2
−0.62
0.00025
0.0057
in vitro_Miapaca2


P2RX2
−0.46
0.0052
0.22
subQ_Miapaca2


P2RX2
−0.52
0.012
0.34
subQ_Panc1









Example 2—Generation of a P2RX2-Specific Inhibitory Antibody

The protein P2RX2 is recombinantly expressed in a mammalian cell culture system, e.g., HEK or CHO cells. Membrane vesicle preparation of the transgene-expressing cells is performed using hypertonic vesiculation buffer, a technique described in Del Piccolo et al., Analytical Chemistry, 84:8650, 2012. Using routine methods such as phage display, yeast display, or animal immunization, an antibody is raised that is specific to P2RX2 vesicles compared to vesicles prepared from non-transgene-modified cells as measured by ELISA. To confirm that the antibody prevents cation flux, transgene-expressing cells are labeled with a calcium reporter dye, e.g., Fluo-8 dye (Abcam ab112129), which fluoresces upon binding with calcium. The cells are incubated with extracellular ATP to trigger calcium flux through the P2RX2 channel in the presence or absence of the antibody to evaluate inhibitory antibody function.


Example 3—Treatment of a Patient with Cancer with a P2RX2 Inhibitor

According to the methods disclosed herein, a physician of skill in the art can treat a patient, such as a human patient with cancer (e.g., pancreatic cancer), so as to inhibit cancer growth, reduce tumor burden, increase cancer cell death, or slow disease progression. The method of treatment can include diagnosing or identifying a patient as a candidate for treatment with a P2RX2 inhibitor based on P2RX2 expression in a biopsy. For example, a tissue sample can be collected from a patient's cancer and analyzed for RNA expression by qPCR or RNAseq analysis, and the cancer can be found to express high levels of P2RX2. To treat the patient, a physician of skill in the art can administer a P2RX2 inhibitor that decreases P2RX2 expression or function (e.g., an inhibitory RNA directed to P2RX2, a P2RX2 inhibitory antibody, or a P2RX2 small molecule inhibitor, e.g., CHEMBL494161). The P2RX2 inhibitor can be administered locally (e.g., injected into the tumor or tumor microenvironment) to decrease tumor growth or volume. The P2RX2 inhibitor is administered in a therapeutically effective amount, such as from 10 μg/kg to 500 mg/kg (e.g., 10 μg/kg, 100 μg/kg, 500 μg/kg, 1 mg/kg, 10 mg/kg, 50 mg/kg, 100 mg/kg, 250 mg/kg, or 500 mg/kg). In some embodiments, the P2RX2 inhibitor is administered bimonthly, once a month, once every two weeks, or at least once a week or more (e.g., 1, 2, 3, 4, 5, 6, or 7 times a week or more).


The P2RX2 inhibitor is administered to the patient in an amount sufficient to decrease tumor growth decrease tumor burden, increase cancer cell death, or increase progression free survival by 10% or more (e.g., 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or more). Tumor growth and tumor burden can be assessed using standard imaging methods (e.g., digital radiography, positron emission tomography (PET) scan, computed tomography (CT) scan, or magnetic resonance imaging (MRI) scan). Images from before and after administration of the P2RX2 inhibitor can be compared to evaluate the efficacy of the treatment, and the rate of disease progression can be assessed by comparison to the patient's medical history prior to administration of the P2RX2 inhibitor. A finding of a reduction in the total number of tumors, number of primary tumors, volume of tumors, growth of tumors, or rate of disease progression indicates that the P2RX2 inhibitor has successfully treated the cancer.


Other Embodiments

While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the invention that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follows in the scope of the claims. Other embodiments are within the claims.

Claims
  • 1. A method of treating a human subject identified as having pancreatic cancer, the method comprising administering to the subject an amount of a small molecule P2RX2 antagonist effective to reduce growth of the pancreatic cancer, thereby treating the subject, wherein the small molecule P2RX2 antagonist is selected from the group consisting of CHEMBL494161, CHEMBL119416, CHEMBL604158, CHEMBL1672098, CHEMBL495204, CHEMBL 499580, CHEMBL598857, CHEMBL1671997, CHEMBL523173, CHEMBL1672107, CHEMBL597820, CHEMBL1671996, CHEMBL492300, CHEMBL523043, CHEMBL597591, CHEMBL1671993, CHEMBL494159, CHEMBL521983, CHEMBL597203, CHEMBL1671992, CHEMBL494353, CHEMBL500550, CHEMBL596982, CHEMBL134193, CHEMBL494160, CHEMBL492299, CHEMBL524284, CHEMBL133576, CHEMBL494158, CHEMBL504607, CHEMBL524064, CHEMBL131271, CHEMBL526307, CHEMBL494176, CHEMBL522725, CHEMBL118007, CHEMBL492934, CHEMBL493547, CHEMBL522053, CHEMBL116926, CHEMBL492933, CHEMBL493546, CHEMBL521709, CHEMBL492729, CHEMBL494582, CHEMBL446310, CHEMBL499428, CHEMBL521820, CHEMBL492907, CHEMBL69727, CHEMBL498038, CHEMBL494940, CHEMBL492703, CHEMBL331358, CHEMBL496229, CHEMBL492789, CHEMBL1672104, CHEMBL494833, CHEMBL496022, CHEMBL69234, CHEMBL495203, CHEMBL509572, CHEMBL495834, CHEMBL401735, CHEMBL1672105, CHEMBL496030, CHEMBL495796, CHEMBL494834, CHEMBL448525, CHEMBL1671995, CHEMBL450832, CHEMBL494832, CHEMBL271672, CHEMBL523000, CHEMBL404659, CHEMBL494772, CHEMBL496401, CHEMBL492968, CHEMBL404450, CHEMBL494181, CHEMBL413145, CHEMBL271688, CHEMBL403051, CHEMBL257495, CHEMBL119180, CHEMBL494581, CHEMBL402239, CHEMBL117766, CHEMBL502618, CHEMBL45413, CHEMBL256864, CHEMBL495195, CHEMBL444469, CHEMBL331250, CHEMBL256688, CHEMBL493740, CHEMBL1672106, CHEMBL492967, CHEMBL256057, CHEMBL492562, CHEMBL493741, CHEMBL492744, CHEMBL1672103, CHEMBL477339, CHEMBL443930, CHEMBL606414, CHEMBL1672102, CHEMBL492935, CHEMBL604300, CHEMBL1672099, CHEMBL522184, and CHEMBL492745.
Foreign Referenced Citations (2)
Number Date Country
WO-2018022668 Feb 2018 WO
WO-2018022668 Feb 2018 WO
Non-Patent Literature Citations (8)
Entry
Burnstock G and Di Virgilio F “Purinergic signalling and cancer” Purinergic Signaling 9:491-540. (Year: 2013).
Cui et al. “Targeting calcium signaling in cancer therapy” Acta Pharmaceutica Sinica B 7:3-17. (Year: 2017).
Di Virgilio F and Adinolfi E “Extracellular purines, purinergic receptors and tumor growth” Oncogene 36:293-303. (Year: 2016).
Di Virgilio et al. “Extracellular ATP and P2 purinergic signalling in the tumour microenvironment” Nature Reviews Cancer 18:601-618. (Year: 2018).
Anonymous “Expression of P2RX2 in cancer—Summary—The Human Protein Atlas” https://www.proteinatlas.org/NESG00000187848-P2RX2/pathology#gene_information (Year: 2018).
Kendrick N “A gene's mRNA level does not usually predict its protein level” Kendrick Labs, Inc. (Year: 2014).
Bhargava et al. “Suramin Inhibits Not Only Tumor Growth and Metastasis but Also Angiogenesis in Experimental Pancreatic Cancer” J. Gastrointest. Surg. 11:171-178. (Year: 2007).
Trujillo et al. “Inhibition Mechanism of the Recombinant Rat P2X2 Receptor in Glial Cells by Suramin and TNP-ATP” Biochemistry 45:224-233. (Year: 2006).
Provisional Applications (1)
Number Date Country
62623236 Jan 2018 US