The Sequence Listing XML associated with this application is provided in XML format and is hereby incorporated by reference into the specification. The name of the XML file containing the sequence listing is 3915-P1270US_SEQ_final_2022-7-15. The XML file is 135 kb; was created on Jul. 15, 2022; and is being submitted via Patent Center with the filing of the specification.
This invention was made with government support under Grant Nos. R01 CA177272 and U54 CA199090, awarded by the National Institutes of Health. The government has certain rights in the invention.
The present disclosure relates to DNA aptamers that specifically bind the transferrin receptor and can be used, for example, in methods of cell selection or cell depletion.
The manufacturing process of chimeric antigen receptor T cells includes cell selection, activation, gene transduction, and expansion. The T cell selection process from each manufacturing company varies. Some groups collect Ficoll®-enriched peripheral blood mononuclear cells (PBMCs) to obtain increased percentage of T cell content, while other groups use antibodies to isolate helper T cells and cytotoxic T cells separately. However, neither of these methods actively eliminate the cancer cells in the patient's blood collected along with the healthy white blood cells. Alarmingly, it has been found that transduction of these tumor-containing white blood cell specimens could lead to treatment failure. A 2018 case study reported a B-ALL patient who underwent anti-CD19 chimeric antigen receptor (CAR) T cell treatment relapsed due to anti-CD19 CAR expressing leukemic cells. These CAR leukemic cells were generated during the T cell transduction process and were unknowingly grafted back to the patient. As a result, the anti-CD19 CAR in cis masked the CD19 on the same cancer cell, protecting it from being detected by the therapeutic CAR T cells.
Provided herein are aptamers that specifically bind the transferrin receptor on the surface of proliferating or cancerous cells. Such aptamers can be used to deplete such proliferating or cancerous cells from a therapeutic population of cells prior to manipulation and administration (e.g., CAR T cells) to a subject for treatment of a disease. Alternatively, the aptamers can be used herein in methods of enriching transferrin receptor-expressing cells, for example, for the purpose of genomic analysis, diagnosis or prognosis of cancer, or monitoring efficacy of a given treatment of e.g., cancer. Another alternative use relates to the targeted delivery of drugs via binding of aptamer-drug conjugates to the transferrin receptor, which is upregulated in rapidly dividing cells as well as, for example, the brain endothelium.
Accordingly, one aspect provided herein relates to a composition comprising an aptamer that selectively binds to the transferrin receptor (also referred to herein as CD71), wherein the aptamer comprises a sequence having at least 75% sequence identity to a composition comprising an aptamer that selectively binds to CD71, wherein the aptamer comprises a sequence having at least 75% sequence identity to residues 22-54 of SEQ ID NO: 2 (TAAAGGGGGTGTTTGTGCGGTGTGGAGTGCGCG); and wherein the aptamer can further comprise a number (N) of nucleotides at each end wherein each nucleotide is selected independently, and wherein each N comprises from 3 nt to 40 nt, from 3 nt to 30 nt, from 3 nt to 20 nt, or from 3 nt to 10 nt. In another embodiment of this aspect and all other aspects provided herein, the aptamer comprises a sequence having at least 75% identity to SEQ ID NO: 1 (tJBA8.1), SEQ ID NO: 2 (JBA8.1), SEQ ID NO: 26 (tJBA8.26), or SEQ ID NO: 27 (JBA8.26). In another embodiment of this aspect and all other aspects provided herein, the aptamer comprises a central loop region, wherein a stem formed by base pairing of nucleotides from the 5′ and 3′ ends of the aptamer projects from the central loop region, and wherein first and second stem loops project from the central loop region.
In another embodiment of this aspect and all other aspects provided herein, the central loop comprises nucleotides 8-11, 23-26 and 41-44 of SEQ ID NO: 1.
In another embodiment of this aspect and all other aspects provided herein, the central loop comprises nucleotides 9-11, 23-26, and 41-43 of SEQ ID NO: 26.
In another embodiment of this aspect and all other aspects provided herein, nucleotides 8 and 44, 11 and 23, and 26 and 41 or nucleotides 9 and 43, 11 and 23, and 26 and 41 are base paired to each other, respectively.
In one embodiment of this aspect and all other aspects provided herein, the aptamer is attached to a solid support or phase-changing agent.
In another embodiment of this aspect and all other aspects provided herein, the solid support comprises a particle, bead, platform, column, filter or sheet, dish, a microfluidic capture device, capillary tube, electrochemical responsive platform, scaffold, cartridge, resin, fiber, or matrix.
In another embodiment of this aspect and all other aspects provided herein, the aptamer further comprises a detectable moiety, a label, a tag, or a probe.
In another embodiment of this aspect and all other aspects provided herein, the aptamer is formulated for use as a sensor.
Another aspect provided herein relates to an aptamer composition comprising an effective amount of an aptamer as described herein and further comprising a pharmaceutically acceptable carrier. In one embodiment of this aspect, the aptamer is attached to polymeric carrier to facilitate drug, nucleic acid, or nucleoprotein delivery. In another embodiment of this aspect, the aptamer is attached to a lipid formulation to facilitate drug, nucleic acid, and/or nucleoprotien delivery. In another embodiment the aptamer is attached directly to the pharmaceutically active agent, such as an antibody or small molecule drug.
Another aspect provided herein relates to a method for preparing cell a composition depleted of cells expressing CD71, the method comprising: contacting a biological sample comprising a desired cell population with an aptamer of claim 1, under conditions and for a time permitting binding of the aptamer to CD71 expressed on the cells in said sample; and separating cells bound to the aptamer from the cells not bound to the aptamer, thereby providing a preparation of cells depleted in CD71 expressing cells for use in generating a therapeutic cell composition.
Another aspect provided herein relates to a method for preparing T cells to be used in generating a therapeutic cell composition, the method comprising: contacting a biological sample comprising T cells with an aptamer as described herein or a composition thereof under conditions and for a time permitting binding of the aptamer to CD71 expressed on cells in said sample; and separating cells bound to the aptamer from cells not bound to the aptamer, thereby providing a preparation of T cells depleted in CD71 expressing cells, for use in generating a therapeutic cell composition.
In one embodiment of this aspect and all other aspects provided herein, the aptamer is bound to a solid support.
In another embodiment of this aspect and all other aspects provided herein, the method further comprises genetically modifying T cells either before or after separation.
In another embodiment of this aspect and all other aspects provided herein, the genetic modification optionally comprises introducing a nucleic acid construct encoding a chimeric antigen receptor.
Also provided herein, in another aspect, is a method for depleting CD71-expressing cells from a biological sample comprising a plurality of cell types, the method comprising: (i) contacting a biological sample comprising a plurality of cell types with an aptamer of claim 1 under conditions and for a time permitting binding of the aptamer to CD71 expressed on cells in said sample; and (ii) separating cells bound to the aptamer from cells not bound to the aptamer, thereby providing a preparation depleted in CD71 expressing cells from the biological sample.
In one embodiment of this aspect and all other aspects provided herein, the active removal of CD71-expressing cells comprises (i) contacting the biological sample with an aptamer composition as described herein under conditions and for a time to permit cells expressing CD71 to bind to the aptamer, and separating cells bound to the aptamer from cells not bound to the aptamer; and (ii) isolating cells expressing CD71 from the cells not bound to the aptamer.
In one embodiment of this aspect of the method the cell preparation depleted of CD71 expression cells comprises NK cells, monocytes, or macrophage.
In another embodiment of this aspect and all other aspects provided herein, the biological sample is isolated from a subject having circulating tumor cells.
Also, provided herein in another aspect is a method for isolating cells expressing CD71, the method comprising: contacting a biological sample comprising CD71-expressing cells with a composition of claim 1 under conditions and for a time permitting binding of the aptamer to CD71 expressed on cells in said sample; and separating cells bound to the aptamer from cells not bound to the aptamer, thereby isolating cells expressing CD71 from the biological sample.
In one embodiment of this aspect of the method, the isolated cells are tumor cells or fetal nucleated red blood cells.
Also provided herein in another aspect is a method of treating a disease, the method comprising: administering the T cells depleted of the cells expressing CD71 by a method set forth above or an engineered or differentiated cell thereof to a subject in need thereof, thereby treating the disease.
Also provided herein in another aspect is a method of delivering a therapeutic agent, the method comprising administering an aptamer described herein, wherein the aptamer is attached to a therapeutic drug, a formulation comprising a therapeutic drug, or a therapeutic cell.
In one embodiment of this aspect and all other aspects provided herein, the therapeutic drug is a nucleic acid or a ribonucleoprotein.
In another embodiment of this aspect and all other aspects provided herein the delivery comprises receptor-mediated transcytosis.
Also provided herein are aptamer dimers comprising one or more of the sequences provided herein. In one embodiment, the dimer is formed by digestion, followed by ligation (see e.g.,
In other embodiments, the aptamer monomers self-assemble into dimers (see e.g.,
This patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
The compositions and methods described herein are related, in part, on the discovery of DNA based aptamers that can bind to the transferrin receptor (also referred to herein as CD71), which is highly expressed on proliferating cells, including cancer cells. As such, the DNA based aptamers described herein can be used to deplete cancerous cells from a biological sample, for use in cell-based therapeutics. Alternatively, the aptamers described herein can be used to isolate proliferating cells, for example, for use in the analysis of such cells or diagnosis of cancer.
As used herein, the term “isolating a cell of interest” refers to the selective separation or enrichment of a target cell, cell type or class of cells from a sample comprising other cells, cell types of cell classes such that the cell population resulting from such separation has a high degree of cell purity as determined by specific cell markers (e.g., CD71 for a CD71 positive T cell or B cell). Such isolated cells can be used in the analysis of proliferating cells in a subject or diagnosis of cancer.
While higher degrees of cell purity are preferred over lower, cell “isolation” as the term is used herein does not require 100% purity of the resulting cell population. Target cells (e.g., transferrin positive cells) or a population thereof will generally be considered “isolated” as the term is used herein if they comprise at least 60% of a target cell population, such as a CD71 positive cell population, resulting from an isolation method as described herein, and preferably at least 70%, at least 80%, at least 90% or more.
A “plurality” contains at least two members. In certain cases, a plurality may have at least 10, at least 100, at least 1000, at least 10,000, at least 100,000, or at least 1,000,000 or more members. As used herein, a “plurality of cell types” refers to a biological sample comprising cells with distinct cell surface markers or compositions thereof and/or physiological functions.
As used herein, surface markers “specific for” a target cell or cell fraction or population of interest are polypeptides or other molecules expressed on the surface of a target cell or cell fraction that are distinct to that target cell or cell fraction and thereby permit the identification and isolation of that cell or cell fraction using a method as described herein. In some embodiments, a single surface marker is sufficient to identify a target cell, e.g., CD8a identifies a CD8+ T cell while CD71/transferrin receptor identifies proliferating cells such as cancer cells. In other embodiments, two or more markers can together identify a target cell or cell population or fraction. In other embodiments, a single marker can identify a class of target cells, e.g., CD71 identifies proliferating cells or cancer cells as a class. In other embodiments the cell surface marker is a membrane lipid, peptide, polypeptide, or protein.
As used herein, the term “contacting a biological sample,” when used in the context of an aptamer or nucleic acid as described herein, refers to addition of an aptamer to a biological sample under conditions that permit specific or selective binding of the aptamer to a target moiety or marker on the cell surface or extracellular matrix of the cell.
As used herein, the term “conditions that permit forming aptamer-bound cells” refers to incubation of cells at 4° C. for 30 minutes in a binding buffer comprising 0.1 mg/mL tRNA, 0.1 g/L CaCl2, 0.2 g/L KCl, 0.2 g/L KH2PO4, 0.1 g/L MgCl2 hexahydrate, 8.0 g/L NaCl, 2.1716 Na2HPO4 septahydrate, supplemented with 25 mM glucose, 5 mM MgCl2, varying amounts of bovine serum albumin (BSA), with a pH of 7.5, or to conditions that provide binding substantially equivalent to binding permitted under these conditions. In this context, substantially equivalent means±10% of the binding permitted under these conditions.
As used herein, the term “nucleic acid” includes one or more types of: polydeoxyribonucleotides (containing 2-deoxy-D-ribose), polyribonucleotides (containing D-ribose), and any other type of polynucleotide that is an N-glycoside of a purine or pyrimidine base, or modified purine or pyrimidine bases (including abasic sites). The term “nucleic acid,” as used herein, also includes polymers of ribonucleosides or deoxyribonucleosides that are covalently bonded, typically by phosphodiester linkages between subunits, but in some cases by phosphorothioates, methylphosphonates, and the like. “Nucleic acids” include single- and double-stranded DNA, as well as single- and double-stranded RNA. Exemplary nucleic acids include, without limitation, gDNA; hnRNA; mRNA; rRNA, tRNA, micro RNA (miRNA), small interfering RNA (siRNA), small nucleolar RNA (snORNA), small nuclear RNA (snRNA), and small temporal RNA (stRNA), and the like, and any combination thereof.
As described herein, a “compensatory change” refers to a change in a nucleoside or nucleobase pair that maintains predicted secondary structure and/or target binding of an aptamer. Non-limiting examples of compensatory changes include changing a G:C base pair to a C:G base pair or changing an A:T base pair to a T:A base pair, e.g., in a stem-loop structure of a nucleic acid. Because of differences in the hydrogen-bonding characteristics between A:T and C:G base pairs, replacing an A:T base pair with a C:G base pair would be expected to alter the stability of the secondary structure, i.e., to increase its stability. Thus, a compensatory change described herein can further include such change. It is contemplated that a change from a C:G or G:C base pair to an A:T or T:A base pair can sometimes be tolerated without significantly affecting the secondary structure or target binding characteristics. However, this type of compensatory change should be considered in the context of the overall stem stability of the aptamer. As described herein, a compensatory change in the nucleotide sequence of an aptamer can involve, where appropriate, a modified nucleoside selected from but not limited to the nucleobases or nucleosides described in Table 1.
As used herein, the term “polypeptide” is intended to encompass a singular “polypeptide” as well as plural “polypeptides,” and includes any chain or chains of two or more amino acids. Thus, as used herein, terms including, but not limited to “peptide,” “dipeptide,” “tripeptide,” “protein,” “enzyme,” “amino acid chain,” and “contiguous amino acid sequence” are all encompassed within the definition of a “polypeptide,” and the term “polypeptide” can be used instead of, or interchangeably with, any of these terms. The term further includes polypeptides that have undergone one or more post-translational modification(s), including for example, but not limited to, glycosylation, acetylation, phosphorylation, amidation, derivatization, proteolytic cleavage, post-translation processing, or modification by inclusion of one or more non-naturally occurring amino acids. Conventional nomenclature exists in the art for polynucleotide and polypeptide structures. For example, one-letter and three-letter abbreviations are widely employed to describe amino acids: Alanine (A; Ala), Arginine (R; Arg), Asparagine (N; Asn), Aspartic Acid (D; Asp), Cysteine (C; Cys), Glutamine (Q; Gln), Glutamic Acid (E; Glu), Glycine (G; Gly), Histidine (H; His), Isoleucine (I; Ile), Leucine (L; Leu), Methionine (M; Met), Phenylalanine (F; Phe), Proline (P; Pro), Serine (S; Ser), Threonine (T; Thr), Tryptophan (W; Trp), Tyrosine (Y; Tyr), Valine (V; Val), and Lysine (K; Lys). Amino acid residues described herein are preferred to be in the “L” isomeric form. However, residues in the “D” isomeric form may be substituted for any L-amino acid residue provided the desired properties of the polypeptide are retained.
As described herein, a “solid support” is a structure upon which one or more aptamers can be displayed for contact with a target cell. A solid support provides a ready means for isolating or removing bound target cells from a mixture or suspension. A solid support can be in the form, for example, of a particle, bead, filter or sheet, resin, scaffold, matrix, or column. Non-limiting classes of materials that the solid support can comprise include polymer, metal, ceramic, gels, paper, or glass. The materials can include, but are not limited to polystyrene, agarose, gelatin, iron oxide, stainless steel, polycarbonate, polydimethylsiloxane, polyethylene, acrylonitrile butadiene styrene, cyclo-olefin polymers and cyclo-olefin copolymers.
As described herein, a “phase changing agent” is an agent that is soluble in aqueous solution under one set of conditions, but can be induced to an insoluble, precipitating form under another set of conditions. The conditions for both soluble and insoluble forms should be compatible with maintaining the viability of target cells. Non-limiting examples of conditions that change phase include temperature, pH and salt or solute concentration. An example of a phase-changing agent includes poly(N-isopropylacrylamide) phase-changing polymers that are soluble at one temperature and then at a different temperature precipitate out from solution.
As used herein, the term “affinity pair” refers to a pair of moieties that specifically bind each other with high affinity, generally in the low micromolar to picomolar range. When one member of an affinity pair is conjugated to a first element and the other member of the pair is conjugated to a second element, the first and second elements will be brought together by the interaction of the members of the affinity pair. Non-limiting examples of affinity pairs that can be conjugated to an aptamer or solid support include ligand-receptor pairs, antibody-antigen pairs, as well as smaller pairs such as biotin-avidin, or biotin-avidin variant, such as biotin streptavidin or biotin-neutravidin, among others. As but one example, the biotin-streptavidin interaction has a Kd of 10−14 to 10−15 molar.
As used herein, the term “conjugated to” encompasses association of an aptamer with a solid support, a phase-changing agent or a member of an affinity pair by covalent bonding, including but not limited to cross-linking via a cross-linking agent, or by a strong non-covalent interaction that is maintained under conditions in which the conjugate is to be used.
As used herein, the term “hybridize” refers to the phenomenon of a single-stranded nucleic acid or region thereof forming hydrogen-bonded base pair interactions with either another single stranded nucleic acid or region thereof (intermolecular hybridization) or with another single-stranded region of the same nucleic acid (intramolecular hybridization). Hybridization between a “reversal agent” or “antidote” and an aptamer permits the disruption of binding of the aptamer to a target by destabilization of the aptamer's secondary structure, allowing for reversible cell selection to occur. Hybridization is governed by the base sequences involved, with complementary nucleobases forming hydrogen bonds, and the stability of any hybrid being determined by the identity of the base pairs (e.g., G:C base pairs being stronger than A:T base pairs) and the number of contiguous base pairs, with longer stretches of complementary bases forming more stable hybrids.
As used herein, a “magnetoresponsive bead” refers to a solid support particle that can be attracted to a magnetic device or magnetic field. A magnetoresponsive bead coated with or otherwise conjugated to an aptamer can be used to separate aptamer-bound cells from a biological sample. While the term “bead” infers a spherical form, this is not a limitation of the shape of magnetoresponsive solid support that can be used to separate the aptamer-bound cells from non-aptamer bound cells. The shape can be irregular, or some variation of spherical, oval, cuboid, and the like. In various embodiments, a magnetoresponsive bead can be conjugated to an aptamer covalently, e.g., via a cross-linking reaction, or can be conjugated non-covalently, e.g., via the interaction of members of an affinity pair.
As used herein, a “cell fraction” refers to a subset of cells in a sample population that shares a given characteristic, e.g., expression of a certain marker or set of markers. A targeted cell fraction can include more than one cell type; as but one example, where T cells are a cell fraction, that fraction can include, for example, CD4+ T cells and CD8+ T cells, among others. In some embodiments, a targeted cell fraction includes a single cell type. T cells, like other cells, can be identified by cluster of differentiation (CD) markers, or chemokine receptors (CCR). Non-limiting examples of T cell markers include CD8, CD19, CD4, CD3, CD28, CD45, CD62, CD31, CD27, or CCR-7.
As used herein the term “selectively binds transferrin receptor 1 (TfR1)/CD71” refers to the capacity of an aptamer as described herein to bind to TfR1/CD71, under conditions that maintain the viability of mammalian cells, with at least 1,000× greater affinity than it binds to CD8 or to CD4, e.g., at least 1,000× greater, at least 5,000× greater, at least 7,500× greater, at least 10,000× greater, at least 15,000× greater, at least 20,000× greater or more. Affinity can be determined, e.g., as described in the Examples herein.
As used herein, the term “specifically binds the transferrin receptor/CD71” refers to the capacity of an aptamer as described herein to bind to the transferrin receptor (also known as CD71) or a given target cell expressing the receptor thereupon, under conditions that maintain the viability of mammalian cells, such that the aptamer binds the transferrin receptor to a significantly greater degree than it binds to other markers or other cells that do not express the transferrin receptor. At a minimum, an aptamer that specifically binds the transferrin receptor binds with a Kd of 1 micromolar or less, e.g., 1 micromolar or less, 900 nanomolar or less, 800 nanomolar or less, 700 nanomolar or less, 600 nanomolar or less, 500 nanomolar or less, 400 nanomolar or less, 300 nanomolar or less, 200 nanomolar or less, 100 nanomolar or less, 90 nanomolar or less, 80 nanomolar or less, 70 nanomolar or less, 60 nanomolar or less, 50 nanomolar or less, 40 nanomolar or less, 30 nanomolar or less, 20 nanomolar or less, or 10 nanomolar or less.
The terms “decrease,” “reduce,” “reduction,” or “inhibit” are all used herein to mean a decrease or lessening of a property, level, or other parameter (such as a disease symptom) by a statistically significant amount. In some embodiments, “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease or reduction in the number of target cells expressing the transferrin receptor (e.g., in a depletion method) by at least 10% as compared to number of transferrin receptor-expressing cells in the starting population prior to depletion and can include, for example, a decrease by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or more. In other embodiments, “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease in at least one symptom of a given disease (e.g., cancer) by at least 10% as compared to the symptom prior to onset of treatment and can include, for example, a decrease by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or more. As used herein, “reduction” or “inhibition” does not encompass a complete inhibition or reduction as compared to a reference level. Rather, the term “complete inhibition” is used to refer to a 100% inhibition as compared to an appropriate reference level. A decrease in a symptom of a given disease can be preferably down to a level accepted as within the range of normal for an individual without the disease.
The terms “increased,” “increase” or “enhance” or “activate” are all used herein to generally mean an increase of a property, level, or other parameter (e.g., number of cells bound to aptamer) by a statistically significant amount; for the avoidance of any doubt, the terms “increased,” “increase” or “enhance” or “activate” means an increase in the number of target cells in a given population, for example, following a cell enrichment method by at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, at least about a 20-fold increase, at least about a 50-fold increase, at least about a 100-fold increase, at least about a 1000-fold increase or more as compared to a reference level.
As used herein, the term “small molecule” refers to a chemical agent which can include, but is not limited to, a peptide, a peptidomimetic, an amino acid, an amino acid analog, a polynucleotide, a polynucleotide analog, an aptamer, a nucleotide, a nucleotide analog, or an organic or inorganic compound (e.g., including heterorganic and organometallic compounds) having a molecular weight less than about 10,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 5,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 1,000 grams per mole, organic or inorganic compounds having a molecular weight less than about 500 grams per mole, and salts, esters, and other forms of such compounds that are compatible with cell viability. In one embodiment, the small molecule comprises a reversal agent and is present at a concentration effective to release an aptamer from a cell surface marker on a cell without undue cell toxicity.
As defined herein, “flow cytometry” refers to a technique for counting and examining microscopic particles, such as cells and chromosomes, by suspending them in a stream of fluid and passing them through an electronic detection apparatus. Flow cytometry allows simultaneous multiparametric analysis of the physical and/or chemical parameters of up to thousands of particles per second, such as fluorescent parameters. Modern flow cytometric instruments usually have multiple lasers and fluorescence detectors. Increasing the number of lasers and detectors allows for labeling by multiple antibodies and can more precisely identify a target population by their phenotypic markers. Certain flow cytometric instruments can take digital images of individual cells, allowing for the analysis of fluorescent signal location within or on the surface of cells.
As used herein, the term “immunotherapy” refers to the treatment of disease via the stimulation, induction, subversion, mimicry, enhancement, augmentation or any other modulation of a subject's immune system to elicit or amplify adaptive or innate immunity (actively or passively) against cancerous or otherwise harmful proteins, cells or tissues. Immunotherapies (i.e., immunotherapeutic agents) include cancer vaccines, immunomodulators, “antibody-based immunotherapies” or monoclonal antibodies (e.g., humanized monoclonal antibodies), immunostimulants, cell-based therapies such as adoptive T-cell therapies or dendritic cell immunotherapies or dendritic cell vaccines, and viral therapies, whether designed to treat existing cancers or prevent the development of cancers or for use in the adjuvant setting to reduce likelihood of recurrence of cancer.
As used herein, the term “toehold” describes a 5-15 nucleotide overhang region of an aptamer that is designed to be single-stranded and complementary to a region of a reversal agent or antidote. The complementary nature of the toehold permits the reversal agent/antidote to readily hybridize to the single stranded region and facilitates the remainder of the reversal agent, which is complementary to an adjacent double-stranded region of the aptamer, to initiate strand displacement at the adjacent double-stranded region. Thus, a toehold will generally be designed or chosen to be adjacent to a region of double stranded sequence one wishes to disrupt or displace, which will generally be a stem structure in an aptamer. The toehold provides a kinetic advantage for a reversal agent to initiate hybridization and strand displacement to thereby efficiently disrupt aptamer binding to its cell surface marker target. A toehold is generally, but not necessarily, added to an aptamer once the aptamer is found to bind a desired target. As an alternative, every member of an aptamer library can, for example, have the same sequence at one end or the other that will serve as a toehold. Accordingly, internal regions of the aptamer that are not base paired under structure prediction can act as toehold sequences. A toehold can include, for example, a relatively high GC content to provide an improvement in strand displacement rate constant for hybridization to its complement relative to a sequence with lower GC content. In one embodiment, the toehold comprises a 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 nucleotide overhang.
As used herein, the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease, or disorder, for example, cancer. For example, the term “treating” and “treatment” refers to administering to a subject an effective amount of a composition, e.g., a therapeutic cell composition that is depleted of transferrin receptor-expressing cells by the aptamers described herein so that the subject has a reduction in at least one symptom of the disease (e.g., fatigue, pain, tumor size, tumor growth, reduction in need for chemotherapeutics, improved prognosis, improvement in predicted mortality, and the like) or an improvement in the disease, for example, beneficial or desired clinical results. For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptoms, diminishment of extent of disease, disease stabilization (e.g., not worsening), delay or slowing of disease progression, amelioration or palliation of the disease state, and remission (whether partial or total), whether detectable or undetectable. In some embodiments, treating can refer to prolonging survival as compared to expected survival if not receiving treatment. Thus, one of skill in the art realizes that a treatment can improve the disease condition but may not be a complete cure for the disease. Successful treatment can also be assessed by a reduction in the need for medical interventions, reduction in hospital or emergency room visits, reduction in fatigue, or other markers of an improved quality of life. In some embodiments, treatment can include prophylaxis. However, in alternative embodiments, treatment does not include prophylaxis.
The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As used herein, the terms “pharmaceutically acceptable”, “physiologically tolerable” and grammatical variations thereof, as they refer to compositions, carriers, diluents, and reagents, are used interchangeably and represent that the materials are capable of administration to or upon a mammal without the production of undesirable physiological effects such as nausea, dizziness, gastric upset, and the like. A pharmaceutically acceptable carrier will not promote the raising of an immune response to an agent with which it is admixed, unless so desired. The preparation of a pharmacological composition that contains active ingredients dissolved or dispersed therein is well understood in the art and need not be limited based on formulation. Typically, such compositions are prepared as injectable either as liquid solutions or suspensions, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared. The preparation can also be emulsified or presented as a liposome composition. The active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof. In addition, if desired, the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient. Therapeutic compositions as described herein can include pharmaceutically acceptable salts of the components therein. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine and the like. Physiologically tolerable carriers are well known in the art. Exemplary liquid carriers are sterile aqueous solutions that contain no materials in addition to the active ingredients and water or contain a buffer such as sodium phosphate at physiological pH value, physiological saline or both, such as phosphate-buffered saline. Still further, aqueous carriers can contain more than one buffer salt, as well as salts such as sodium and potassium chlorides, dextrose, polyethylene glycol and other solutes. Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, and water-oil emulsions. The amount of an active agent used with the methods described herein that will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition and can be determined by standard clinical techniques.
As used herein, the term “subject” includes humans and mammals. The term “mammal” is intended to encompass a singular “mammal” and plural “mammals,” and includes, but is not limited to humans; primates such as apes, monkeys, orangutans, and chimpanzees; canids such as dogs and wolves; felids such as cats, lions, and tigers; equids such as horses, donkeys, and zebras; food animals such as cows, pigs, and sheep; ungulates such as deer and giraffes; rodents such as mice, rats, hamsters and guinea pigs; and bears. In some preferred embodiments, a mammal is a human. A subject can be of any age including a neonate, toddler, child, teen, adult, or a geriatric subject.
As used herein the term “comprising” or “comprises” is used in reference to compositions, methods, and respective component(s) thereof, that are essential to the claimed technology, yet open to the inclusion of unspecified elements, whether essential or not.
As used herein the term “consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the invention.
The term “consisting of” refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.
As used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Thus, for example, references to “the method” includes one or more methods, and/or steps of the type described herein and/or which will become apparent to those persons skilled in the art upon reading this disclosure and so forth.
Nucleic Acid Aptamer Compositions
Nucleic acid aptamers, single-strands of nucleic acid oligonucleotides capable of binding target molecules, are an attractive alternative to antibodies for cell selection. Aptamers can possess binding affinities comparable to or even higher than antibodies. Importantly, aptamers are produced synthetically as well-defined, low variability products with long storage stability. Aptamers can be discovered through a library selection method known as systematic evolution of ligands by exponential enrichment (SELEX), and further optimized for chemical stability. With their favorable attributes, the application field for aptamers has escalated in the last quarter century to encompass areas including sensing, purification, diagnostics, drug delivery, for example, transcytosis and aptamer-mediated delivery of, for example, a nucleic acid or a ribonucleoprotein, and the like, and other therapeutically effective agents.
Nucleic acid aptamers include RNA, DNA, and/or synthetic nucleic acid analogs (e.g., PNA) capable of specifically binding target molecules. Aptamers are an attractive alternative to antibodies for cell selection because of their high level of specificity and affinity for cell surface markers. Synthetic aptamers were first developed in the 1990s by the Szostak and Gold groups, and it was discovered that aptamers can possess binding affinities comparable to or even higher than antibodies.
Provided herein are methods and compositions for generating functional nucleic acid aptamers for cell selection. The composition of the nucleic acid aptamer can include but is not limited to the nucleobases described in Table 1 (below) and can comprise one or more combinations of backbone or nucleobase structure characteristic of DNA, RNA, or synthetic nucleic acid analogs such as PNAs or BNAs.
Aptamers generally consist of relatively short oligonucleotides that typically range from 20 to 80 nucleotides in length, for example, at least 20 nucleotides, at least 30 nucleotides, at least 40 nucleotides, at least 50 nucleotides, at least 60 nucleotides, at least 70 nucleotides, at least 80 nucleotides or more. An aptamer can be attached to a longer sequence, e.g., at one end or the other of the aptamer, although appended sequences that affect the secondary structure of the aptamer can affect aptamer function. In certain embodiments, the aptamer, for example, an aptamer that selectively binds to CD71 and comprises a sequence having 75% sequence identity to residues 22-54 of the aptamer JBA8.1 (SEQ ID NO:2) and wherein the aptamer can further comprise a number (N) of nucleotides (nt) at either the 5′ and/or 3′ end, wherein each nucleotide is independently selected from adenine, thymine/uracil, guanine, and/or cytosine. N can be present independently at either end, and can be from 3 to 40 nucleotides, and optionally from 3 to 30 nucleotides, from 3 to 20, and preferably 3 to 10 nucleotides. Preferably, the number (N) nucleotides at each end form a stem structure.
The functional activity of an aptamer, i.e., binding to a given target molecule such as the transferrin receptor, involves interactions between moieties or elements in the aptamer with moieties or elements on the target molecule. The interactions can include, for example, hydrophobic/hydrophilic interactions, charge or electrostatic interactions, hydrogen bonding, and the like, and the specific interactions of a given aptamer with a given target are determined by the sequence of the aptamer and the secondary and tertiary structure assumed by that sequence under binding conditions. Thus, the occurrence of intramolecular base pairing in the aptamer is a primary factor in aptamer structure and therefore aptamer function. Intramolecular base pairing can result, for example, in double stranded stem structures, stem-loop structures, and exposure of various elements of the aptamer that can participate in binding interactions with a target molecule. Where the secondary structure of an aptamer is defined by its sequence, including the presence of intramolecular base pairs between regions of complementary sequence that fold the molecule into a functional shape, it should be understood that changes in aptamer sequence occurring in a stem structure or that introduce new options for intramolecular base pairing can disrupt the conformation of the molecule and thereby its function. That said, when a change of one nucleotide in a base-paired stem structure is accompanied by a compensatory change in the complementary nucleotide that maintains the ability to base pair, the structure, and thereby the function of the aptamer can be maintained. That is, some aptamers can tolerate some degree of sequence change and still retain binding activity. Furthermore, a truncated or partial sequence of an aptamer as described herein can also retain binding activity provided that the truncation does not alter intramolecular base-pairing necessary for the secondary structure of the aptamer. In particular, it is contemplated that removal of some sequence from the 5′ or 3′ end of an aptamer described herein can result in an aptamer molecule that retains binding activity. Indeed, some changes can improve binding activity. This, of course, is one basis for an iterative selection approach used to identify aptamers that bind a given target and do so with high affinity.
As described herein, an aptamer can additionally or alternatively comprise nucleobase (often referred to in the art simply as “base”) modifications or substitutions. Such substitutions can modify stability of the aptamer or reversal agent, e.g., by reducing susceptibility to enzymatic or chemical degradation, or can modify (increase or decrease) intra- or inter-molecular interactions, including but not limited to base-pairing interactions. Aptamer and reversal agent nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U) or modified or related forms thereof. Modified nucleobases include, as non-limiting examples, other synthetic and natural nucleobases, such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine, among others. The base pairing behavior and preferences of these nucleobases are known in the art.
Synthetic oligonucleotides comprising an aptamer can include but are not limited to peptide nucleic acid (PNA), bridged nucleic acid (BNA), morpholinos, locked nucleic acids (LNA), glycol nucleic acids (GNA), threose nucleic acids (TNA), or any other xeno nucleic acid (XNA) described in the art.
One such oligonucleotide, an oligonucleotide mimetic, that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (PNA). In PNA compounds, the sugar-backbone of an oligonucleotide is replaced with an amide-containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to atoms of the amide portion of the backbone.
In some embodiments, a transferrin receptor-binding aptamer as described herein comprises one of the sequences in Table 2. In another embodiment, a transferrin receptor-binding aptamer as described herein comprises at least 75% sequence identity to one of the sequences in Table 2. In other embodiments, the transferrin receptor-binding aptamer comprises at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99% sequence identity to one of the sequences in Table 2. In certain embodiments, the transferrin receptor-binding aptamer consists essentially of one of the sequences in Table 2. In other embodiments, the transferrin receptor-binding aptamer consists of one of the sequences in Table 2.
In some embodiments, the transferrin receptor-binding aptamer comprises the sequence: GCAGCANCNNANAGGNNGTGTNTNNGNNGTNTGNAGTGNNNNNGCNNCTNN (SEQ ID NO: 32), wherein N is A, T, C, or G. In other embodiments, the transferrin receptor-binding aptamer comprises a sequence selected from the group consisting of:
In some embodiments, the transferrin receptor-binding aptamer described herein comprises at least one mutation in the sequence of SEQ ID NO: 1 (JBA8.1) at the following nucleic acids (in 5′ to 3′ order): G at position 7, G at position 9, A at position 12, G at position 16, G at position 17, T at position 22, G at position 24, T at position 25, C at position 27, G at position 28, G at position 31, G at position 34, C at position 39, G at position 40, C at position 41, G at position 42, T at position 43, T at position 46, G at position 47, G at position 50, or C at position 51. In some embodiments, the aptamer comprises at least two, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 nucleic acid mutations selected from the afore-cited nucleic acid positions in SEQ ID NO: 1 and/or SEQ ID NO: 2, provided that the aptamer retains at least 75% sequence identity to residues 10-42 of SEQ ID NO: 1 or 22-55 of SEQ ID NO:2. The nucleic acid at a given position described above can be mutated to any of the other naturally occurring nucleic acids (e.g., a G, can be mutated to a T, C, or A) or a synthetic nucleic acid as desired. If necessary, the aptamer binding characteristics can be determined using the methods outlined in the Example section for binding to transferrin receptor-expressing cells.
In one embodiment, the transferrin receptor-binding aptamer described herein comprises (or consists essentially or consists of) tJBA8.1 (SEQ ID NO: 1). SEQ ID NO: 1 comprises secondary structure including a first, second and third stem region as follows:
SEQ ID NO: 1 also comprises a first, second and third loop region as follows:
Nucleic Acid Aptamer Secondary Structure and Cell Targeting
As noted above, the aptamers described herein have the ability to fold into 2-dimensional (2D) and 3-dimensional (3D) structures that interact with specific targets. Aptamers generally bind to specific targets through non-covalent interactions with a target, e.g., a cell surface marker, via interactions including but not limited to electrostatic interactions, hydrophobic interactions, and/or their complementary shapes.
It will be understood by one of skill in the art that the aptamer 2-D and 3-D structures can be predicted by any of several methods to define properties such as equilibrium probability and stability. The NUPACK™ web application can be used to generate predicted secondary structures of aptamer sequences (see, e.g., Zadeh, J. N., Steenberg, C. D., Bois, J. S., Wolfe, B. R., Pierce, M. B., Khan, A. R., Dirks, R. M. & Pierce, N. A. NUPACK: Analysis and design of nucleic acid systems. J. Comput. Chem. (2011) doi:10.1002/jcc.21596, which is incorporated herein by reference in its entirety). Additional non-limiting examples of aptamer structure prediction methods include in silico models such as UNPACK™, APTANI™, 3D-DART™, ModeRNA™, or Unified Nucleic Acid Folding and hybridization package (UNAFold™), or any other oligonucleotide structure prediction in silico model known in the art.
Different structure prediction models can produce different predicted structures, and even the same model can produce different predicted structures if different baseline parameter are used, e.g., temperature, ionic strength, etc.
It is contemplated that the reverse, complement, reverse complement, or truncated sequences of the aptamers described in the Examples can be used for isolation of cells, as it is contemplated that these sequences would maintain the secondary structure of the aptamer.
For any given secondary structure prediction model, the maintenance or improvement in aptamer binding to its target when a modification, e.g., a compensatory or non-compensatory change is made on the basis of predicted structure should be tested experimentally.
A cell can be designated “positive” or “high,” “dim” or “low,” or “negative” for the transferrin receptor (i.e., CD71), and such designations can be useful for the practice of the assays and cell isolation/depletion methods described herein. A cell is considered “positive” for a cell-surface marker if it expresses the marker on its cell-surface in amounts sufficient to be detected using methods known to those of skill in the art, such as contacting a cell with an antibody or aptamer that binds selectively or specifically to that marker, and subsequently performing flow cytometric analysis of such a contacted cell to determine whether the antibody or aptamer is bound the cell. It is to be understood that while a cell may express messenger RNA for a cell-surface marker, in order to be considered positive for the assays and methods described herein, the cell must express it on its surface. A cell is considered “dim” or “low” for the transferrin receptor if it expresses the receptor on its cell-surface in amounts sufficient to be detected using methods known to those of skill in the art, such as contacting a cell with an antibody or aptamer that binds selectively or specifically to the transferrin receptor, and subsequently performing flow cytometric analysis of such a contacted cell to determine whether the antibody or aptamer is bound to the cell, but there exists another distinct population of cells that expresses CD71 at a higher level, giving rise to at least two populations that are distinguishable when analyzed using, for example, flow cytometry. Similarly, a cell is considered “negative” for the transferrin receptor/CD71 if it does not express the marker on its cell-surface in amounts sufficient to be detected using methods known to those of skill in the art, such as contacting a cell with an antibody or aptamer that binds selectively or specifically to the transferrin receptor and subsequently performing flow cytometric analysis of such a contacted cell to determine whether the antibody or aptamer is bound the cell.
In some embodiments, the aptamers (e.g., aptamer monomers) described herein can self-associate or be induced to self-associate to form higher secondary structures including, but not limited, to dimers, trimers, tetramers, pentamers and the like. In one embodiment, the aptamers described herein are provided in the form of a dimer. It is specifically contemplated herein that such higher order structures can improve affinity or avidity; or lower the KD of a given aptamer as compared to its monomer, for example, as shown in
Nucleic Acid Aptamer Synthesis and Modifications
Aptamers as described herein can be chemically synthesized using, as a non-limiting example, a nucleoside phosphoramidite approach. Furthermore, aptamers can be isolated from a biological sample by DNA or RNA extraction methods. These methods include but are not limited to column purification, ethanol precipitation, phenol-chloroform extraction, or acid guanidinium thiocyanate-phenol chloroform extraction (AGPC).
Following extraction or synthesis, the aptamers described herein can be characterized by liquid chromatography, mass spectrometry, next generation sequencing, polymerase chain reaction (PCR), gel electrophoresis, or any other method of identifying nucleoside sequences, secondary structures, chemical composition, expression, thermodynamics, binding, or function. Aptamers identified by cell-SELEX can further be characterized by aptamer cell binding assays, flow cytometry, or in vivo function as described in the EXAMPLES.
The aptamers described herein can also be modified or conjugated to a solid support or phase-changing agent for cell selection and cell processing. Non-limiting examples of conjugation methods include chemical, thermodynamic, or structural modifications to the aptamer that allows for separation of the aptamer-bound cells from the aptamer un-bound cells or biological sample.
In certain embodiments, the aptamers as described herein can be labeled. Non limiting examples of labels can include, for example, fluorophores, and or members of an affinity pair.
Non-limiting examples of affinity pairs that can be conjugated to the aptamer include, for example, biotin:avidin, biotin:streptavidin. biotin:neutravidin (or other variants of avidin that bind biotin).
Solid Supports
In certain embodiments, aptamers are bound directly or indirectly to a solid support. Aptamer-bound solid supports described herein can exist in the form of a platform, column, filter or sheet, dish, a microfluidic capture device, capillary tube, electrochemical responsive platform, scaffold, cartridge, resin, matrix, bead, or another solid support known in the art.
In some embodiments, the solid support comprises materials that include, but are not limited to, a polymer, metal, ceramic, gels, paper, or glass. The materials of the solid support can further comprise, as non-limiting examples, polystyrene, agarose, gelatin, alginate, iron oxide, stainless steel, gold nanobeads or particles, copper, silver chloride, polycarbonate, polydimethylsiloxane, polyethylene, acrylonitrile butadiene styrene, cyclo-olefin polymers or cyclo-olefin copolymers, or Sepharose™ resin.
The aptamer-bound solid support can further comprise a magnetoresponsive element such as a magnetoresponsive bead. In some embodiments, the magnetoresponsive element or bead is in the form of a sphere, cube, rectangle, cylinder, cone, or any other shape described in the art. Aptamer bound to magnetoresponsive beads provides a simple method of separating aptamer-bound cells from non-bound cells by permitting a suspension of the cells to interact with the aptamer-conjugated beads, and then subjecting the sample to a magnetic field. The beads, with aptamer-bound cells, are attracted to the magnetic source, permitting the removal of non-bound cells, e.g., via pipette. Beads with bound cells can be washed and subjected to the magnetic field again to increase the relative purity of the isolated cell fraction.
In some embodiments, the magnetoresponsive element comprises magnetite, iron (III) oxide, samarium-cobalt, terfenol-D, or any other element attracted to a magnet as described in the art.
In some embodiments the solid support is in contact with an extracellular matrix protein or composition. Non-limiting examples include fibronectin, collagen, laminin, poly-L-lysine, Matrigel™ vitronectin, tenascin, fibrillin, brevican, elastin, or other extracellular matrix protein or composition known in the art.
A solid support bound to the aptamer can also contain a label. In some embodiments, the label is conjugated to the aptamer. In some embodiments, the label is a heterologous protein. In some embodiments, the heterologous protein is a tag, such as a fluorescent protein. Such proteins can facilitate tracking and/or visualization of the aptamers. Examples of fluorescent proteins include, but are not limited to, green fluorescent protein (GFP) from the jellyfish Aequorea victoria; mutant versions of GFP that fluoresce different colors (such as BFP, blue fluorescent protein; YFP, yellow fluorescent protein; and CFP, cyan fluorescent protein); dsRed fluorescent protein (dsRed2FP); eqFP611, a red fluorescent protein isolated from Entacmaea quadricolor; AmCyan1, a cyan fluorescent protein isolated from Anemonia majano, and originally named amFP486; Azami Green, a bright fluorescent protein isolated from Galaxeidae; ZSGREEN™, a fluorescent protein isolated from Zoanthus; or any other fluorescent protein or element described in the art.
In some embodiments the aptamers are labeled with a fluorophore. Non-limiting examples of fluorophores include fluorescein, rhodamine, Oregon green, eosin, Texas red, cyanins, e.g., Cy5.5, among others.
In one embodiment, the aptamer can further comprise a “tag,” which refers to a component that provides a means for attaching or immobilizing an aptamer (and any target molecule that is bound to it) to a solid support, such as a bead, e.g., an agarose bead. A “tag” is a set of copies of one type or species of component that is capable of associating with a probe. “Tags” refers to more than one such set of components. The tag can be attached to or included in the aptamer by any method known in the art. Generally, the tag allows the aptamer to associate, either directly or indirectly, with a probe that is attached to the solid support, e.g., a bead. A tag can enable the localization of an aptamer covalent complex to a spatially defined address on a solid support.
Different tags, therefore, can permit the localization of different aptamer covalent complexes to different spatially defined addresses on a solid support. A tag can be a polynucleotide, a polypeptide, a peptide nucleic acid, a locked nucleic acid, an oligosaccharide, a polysaccharide, an antibody, an affybody, an antibody mimic, a cell receptor, a ligand, a lipid, any fragment or derivative of these structures, any combination of the foregoing, or any other structure with which a probe (or linker molecule, as described below) can be designed or configured to bind or otherwise associate with specificity. Generally, a tag is configured such that it does not interact intramolecularly with either itself or the aptamer to which it is attached or of which it is a part. In one embodiment, the tag is included on the 5′-end of the aptamer. In another embodiment, the tag is included on the 3′-end of the aptamer. In one embodiment, the tag is a biotin molecule. In another embodiment, the tag is a streptavidin molecule.
In another embodiment, an aptamer is attached to a solid support through interactions between the tag and a probe on the beads or solid support. A “probe” is a set of copies of one type or species of component that is capable of associating with a tag. “Probes” refers to more than one such set of components. The probe can be attached to or included in the beads or solid support by any method known in the art. Generally, the probe allows the bead or solid support to associate, either directly or indirectly, with a tag that is attached to the aptamer. A probe can be a polynucleotide, a polypeptide, a peptide nucleic acid, a locked nucleic acid, an oligosaccharide, a polysaccharide, an antibody, an affybody, an antibody mimic, a cell receptor, a ligand, a lipid, any fragment or derivative of these structures, any combination of the foregoing, or any other structure with which a probe can be designed or configured to bind or otherwise associate with specificity with a tag. In one embodiment, the probe is a streptavidin molecule, for example, the streptavidin moiety binds to the biotin groups on the aptamer, thereby localizing the aptamers on the solid support to which the streptavidin-coupled beads are bound. In another embodiment, the probe is a biotin molecule.
In one embodiment, the aptamers and solid supports as described herein can be used in a device for enriching a cell population of CD71-expressing cells or alternatively, depleting a cell population of such CD71-expressing cells. The device can optionally comprise a column containing the beads and the filter. In some embodiments, the column is fitted with a syringe. In some embodiments, the column can be sized to fit in a centrifuge tube. In some embodiments the device further comprises a centrifuge tube housing the column.
In some embodiments, the beads with attached aptamer are packed in a column. In other embodiments, the beads with attached aptamer are present in a suspension and collected by centrifugation. The column containing the beads can be of a size and character to allow release of cells without removal of beads. In some embodiments, the column can be of a volume of about 1 mL, 2 mL, 3 mL, 4 mL, 5 mL, 6 mL, 7 mL, 8 mL, 9 mL, 10 mL, 15 mL, 20 mL, 30 mL, 40 mL, 50 mL, 60 mL, 70 mL, 80 mL, 90 mL, 100 mL, 500 mL, 1000 mL, and the like. To facilitate removal of cells from beads, the column can be sized to fit in a centrifuge tube, for example, a small Eppendorf™ tube or a large falcon tube, such that cells can be collected by centrifugation using either a tabletop centrifuge or a large centrifuge. When beads are packed in a column, the column can contain a filter with pores sized to allow cells to pass through while retaining beads in the column. In one embodiment, the filter has a pore size smaller than the diameter of the beads. In another embodiment, the filter has a pore size larger than the diameter of the cells to be enriched. In some embodiments, the filter has a pore size of, for example, about 10-100 μm, e.g., about 10 μm, 20 μm, 30 μm, 40 μm, 50 μm, 60 μm, 70 μm, 80 μm, 90 μm, or 100 μm. In other embodiments, the filter has a pore size of about 10-50 μm, about 10-30 μm, about 10-25 μm, about 10-20 μm, or about 10-15 μm. In some embodiments, the filter has a pore size of less than 10 μm.
The size of beads to which the aptamer is attached can vary. In some embodiments, the beads have a diameter of about 30-200 μm.
In some embodiments, the beads are separated from the captured transferrin receptor-expressing cells by applying mechanical forces, without the addition of any other reagents. As a result, the isolated cell population is free or substantially free of beads, aptamer, antibody, or any other undesired reagents to maintain cell phenotype for additional analysis. In some embodiments, mechanical forces can be applied to the beads without removing them from the column. Release of captured transferrin receptor-expressing cells can be accomplished by, for example, resuspension in a buffer, shaking, pipetting, or vortexing the aptamer-coupled beads. In other embodiments, the column is fitted with a syringe. The syringe can be used to mechanically agitate the beads/cells, thereby disrupting the interaction between beads and cells. The use of mechanical force to separate cells from the aptamer coated beads allows the bound cells to be released without adding any extraneous reagents that could subsequently contaminate the cell population and limit the use of the cell population in clinical applications.
In other embodiments, captured cells can be released from the device using a change in temperature. For example, the beads can be exposed to elevated temperatures for a minimal amount of time sufficient to denature the nucleic acid aptamer and release the cells, without significantly impacting cell viability. In other embodiments, captured cells can be released from the device using a nucleic acid molecule complementary to all or a part of the nucleic acid aptamer (also referred to herein as a “release agent”). Such release agents can compete for binding to the aptamer with cells containing the target, causing release of cells when the aptamer binds to the complementary nucleic acid. Temperature and/or complementary nucleic acid can be used to release cells from the aptamer independently or in conjunction with mechanical disruption, as described herein.
In alternative embodiments, a phase-changing agent can be used in place of a solid support. Phase change agents can change phase or precipitate under a given set of conditions and can thereby facilitate the separation of aptamer-bound from aptamer-unbound cells. In some embodiments, a phase-changing agent can be bound to the aptamer. For example, the phase-changing agent can be soluble in aqueous solution under one set of conditions, but induced to an insoluble, precipitating form under another set of conditions. Further exemplary conditions that can induce a phase change include temperature, pH, salt or solute concentration, light (e.g., ultraviolet or fluorescent), or mechanical forces. For example, poly(N-isopropylacrylamide) is a phase-changing polymer that is soluble, for example, at one temperature and then at a different temperature precipitates out from solution. Upon induction of precipitation, cells bound to the aptamer will be pulled into the precipitated phase. It is also contemplated that a phase changing agent can act in a similar manner to riboflavin that is activated by ultraviolet light.
Reversal Agents for Reversible Cell Selection
Reversal agents or “antidotes” include molecules that disrupt the structure of an aptamer and thereby cause release of cells or molecules bound by the aptamer, or molecules that bind with the aptamer thereby reducing cell-binding affinity. In some embodiments, a reversal agent (also referred to herein as an ‘antidote’) is specific for one or a limited number of aptamers that share a given sequence motif or structure, while in other embodiments, a reversal agent disrupts aptamer structure non-specifically, such that one reversal agent can be used for any of a number of different aptamers. Such non-specific reversal agents include, but are not limited to, for example, polyanions such as dextran sulfate, heparin sulfate, phytic acid, or polyphosphates. In other embodiments, the aptamer of interest can be fused with a small-molecule binding aptamer so that the reversal agent can comprise a small molecule that binds to and affects the folded structure of an aptamer, thereby causing release of bound target molecules or cells. In another example, reversal agents can be chelators that bind essential ions that promote the secondary structure of the aptamer. A small molecule reversal agent can be non-specific or specific, depending upon how it interacts with the aptamer. For example, small molecules that can be used include but are not limited to ATP, ampicillin, tetracycline, dopamine and sulforhodamine B. Further examples of small molecules that bind to aptamers are described in McKeague and DeRosa, J. Nucleic Acids. Vol. 2012, Article ID No. 748913 (2102), which is incorporated herein by reference in its entirety.
In other embodiments, a reversal agent can comprise an oligonucleotide or oligonucleotide mimic (PNA, LNA, BNA, and the like) that includes sequence complementary to a portion of an aptamer that forms a double-stranded stem structure. The hybridization of the oligonucleotide reversal agent to its complement in the aptamer disrupts the structure of the aptamer and causes release of the aptamer's target molecule. An oligonucleotide reversal agent can also include sequence complementary to a non-double stranded region of an aptamer. One such oligonucleotide reversal agent configuration includes a sequence element complementary to a single-stranded portion of a target aptamer and a sequence element complementary to an adjacent double-stranded portion of the target aptamer. In this configuration, the oligonucleotide reversal agent can efficiently initiate hybridization to the single-stranded portion of the aptamer, and then strand-displace the adjacent double-stranded portion of the aptamer to promote release of the aptamer from its target.
Small molecule reversal agents can be selected from, for example, a library of small molecules, which can include, for example, amino acids, oligopeptides, polypeptides, proteins, or fragments of peptides or proteins; nucleic acids (e.g., antisense; DNA; RNA; or peptide nucleic acids, PNA); carbohydrates or polysaccharides, or an organic or inorganic compound (e.g., including heterorganic and organometallic compounds). Each member of the library can be singular or can be a part of a mixture (e.g., a compressed library). The library can contain purified compounds or can be “dirty” (i.e., containing a significant quantity of impurities). Commercially available libraries can be obtained, for example, from Affymetrix, ArQule, Neose Technologies, Sarco, Ciddco, Oxford Asymmetry, Maybridge, Aldrich, Panlabs, Pharmacopoeia, Sigma, or Tripose, among others.
The use of reversal agents permits a reversible, tag-less cell selection. An advantage of the release of aptamer-bound cells in this manner is that the isolated cells will have relatively little aptamer bound to them. This contrasts with the use of antibodies to isolate cells, in which it is difficult to get the antibodies to dissociate from the cells after the cells are isolated. Depending upon how cells are incubated with aptamer, e.g., whether the aptamers are initially bound to a solid support or are first incubated in solution then pulled down via an affinity pair, and how cells are incubated with reversal agent (e.g., reversal agent concentration, timing, and the like) the amounts of aptamer remaining bound to isolated cells after reversal can be, for example, 50% or less, 40% or less, 30% or less, 20% or less, 10% or less, or even 5% or less of the aptamer used for binding.
As described herein, an oligonucleotide reversal agent can additionally or alternatively comprise nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, aptamer and reversal agent nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U). Modified nucleobases include other synthetic and natural nucleobases, such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine.
Synthetic oligonucleotides of a reversal agent can include but are not limited to peptide nucleic acid (PNA), bridged nucleic acids (BNA), morpholinos, locked nucleic acids (LNA), glycol nucleic acids (GNA), threose nucleic acids (TNA), or any other xeno nucleic acid (XNA) described in the art.
Oligonucleotide reversal agents can be synthesized by the same methods used for the synthesis of nucleic acid aptamers as described herein.
Oligonucleotide reversal agents generally consist of short oligonucleotide strands that typically range from 8 to 50 nucleobases in length but can be as long as the target aptamer or longer. In some embodiments, oligonucleotide reversal agents can comprise polynucleotide lengths of 8 or more, 10 or more, 20 or more, 30 or more, 40 or more, 50 or more, 60 or more, 70 or more, 80 or more, 90 or more, 100 or more and beyond.
Reversal agent selection can include designing complementary oligonucleotide sequences to the 3′ end of an aptamer, or to any region of the aptamer known or predicted to participate in intramolecular base pairing.
In some embodiments dissociation of the aptamer from cells can be carried out by using shear force, changing the temperature, changing the buffer composition, ionic strength of the buffer solution, and the like.
In some embodiments, binding of the cells of interest with 5 nM aptamer can be carried out with secondary fluorescent streptavidin labeling. Labeled cells can be incubated with varying fold-excess (over the amount of aptamer used) in, e.g., 200 microliters (μL) of reversal agent in wash buffer with 1% (weight/volume) BSA for different times and temperatures. Cells can be washed twice with wash buffer and 1% (weight/volume) BSA to remove eluted aptamers, followed by fixation, and/or flow cytometry analysis.
In some embodiments, the concentration of reversal agent added to the mixture of cells is 1 nanomolar or more, 10 nanomolar or more, 100 nanomolar or more, 1 micromolar or more, 10 micromolar or more, 100 micromolar or more, 1 millimolar or more and beyond. The reversal agent can be from 0.1-fold to 100-fold excess of the aptamer concentration.
In some embodiments, a toehold region of an aptamer can facilitate reversal agent hybridization that disrupts the secondary structure of the chosen aptamer. The toehold region can be at the end of the aptamer or internally within the aptamer at a single stranded region, depending on the predicted secondary structure.
In some embodiments, reversal of aptamer binding can be accomplished by degrading the aptamer, e.g., by introducing a nuclease to the aptamer-bound cell composition. Nucleases can be non-specific, such as DNAse I, or specific or cleavage at one or more particular sequences. Other approaches for reversal include, for example, contacting with a single strand binding protein (SSBP); binding of such factors to a single-stranded loop of the aptamer can promote unwinding of hybridized stem regions and promote loss of aptamer binding. Further approaches for reversal can include, for example, changing ionic strength, buffer or temperature conditions, changing the pH, or the presence of divalent cations, such that aptamer tertiary or secondary structure is altered to promote loss of aptamer binding.
Characterization of Aptamer Cell Binding
Nucleic acid aptamers identified by the cell-SELEX method described herein, or by another method as known in the art can be characterized by a number of approaches including, but not limited to, aptamer binding assays, next generation sequencing, gene profiling, functional assays such as cytotoxicity assays, cytokine release assays, and in vivo delivery of, for example, native or modified cells isolated through use of the aptamers to an animal or human model. The above methods can serve as quality control of the compositions and method of cell selection described herein.
To characterize aptamer binding to target cells or cells of interest, an in vitro binding assay can be performed, for example, as follows. A population comprising, e.g., 2×105 or more cells of interest (e.g., Jurkat T cells expressing CD71) is incubated with folded FAM-labeled ssDNA pools or FAM/biotin-labeled individual aptamers for 30 minutes at 4° C. in binding buffer as described herein, and at various concentrations of aptamer. In some embodiments, the aptamers are labeled with biotin, avidin, streptavidin, agarose, or neutravidin. In some embodiments the aptamers are labeled with a fluorophore. Non-limiting examples of fluorophores include fluorescein, rhodamine, Oregon green, eosin, Texas red, cyanins, e.g., Cy5.5, among others.
Following incubation, e.g., in a total volume of 100 μL, cells are washed twice in, e.g., 200 μL of wash buffer supplemented with 1% (weight/volume) BSA to remove excess aptamer. If the aptamers used were biotinylated, cells can undergo a second incubation with 100 μL fluorescently-labeled streptavidin or neutravidin secondary label for 20 min at 4° C. in wash buffer with 1% BSA before again washing twice. Stained cells can be fixed, e.g., in 200 μL wash buffer with 1% BSA (weight/volume) and 0.1% (weight/volume) paraformaldehyde (PFA) before analyzing via flow cytometry.
At a minimum, an aptamer that specifically binds a given target cell or a cell expressing the transferrin receptor with at least 100× greater affinity than the binding of the aptamer to a cell that does not express the transferrin receptor, and preferably with at least 200× greater affinity, at least 300× greater affinity, at least 500× greater affinity, at least 600× greater affinity, at least 700× greater affinity, at least 800× greater affinity, at least 900× greater affinity, at least 1,000× greater affinity or more.
Affinity can be expressed in terms of dissociation constant, or KD. An aptamer that selectively binds the transferrin receptor/CD71 will generally bind with a KD below 1 micromolar (1 μM). Aptamers have been described that bind their targets with KDs in the picomolar (μM) range. However, aptamers useful for cell selection can bind in the range of 1 μM to 10 μM, 1 μM to 100 μM, 1 μM to 200 μM, 1 μM to 300 μM, 1 μM to 400 μM, 1 μM to 500 μM, 1 μM to 600 μM, 1 μM to 700 μM, 1 μM to 800 μM, 1 μM to 900 μM, 1 μM to 1 nM, 1 μM to 10 nM, 1 μM to 50 nM, 1 μM to 100 nM, 1 μM to 150 nM1 μM to 200 nM, 1 μM to 250 nM, 1 μM to 300 nM, 1 μM to 350 nM, 1 μM to 400 nM, 1 μM to 450 nM, 1 μM to 500 nM, 1 μM to 550 nM, 1 μM to 600 nM, 1 μM to 650 nM, 1 μM to 700 nM, 1 μM to 750 nM, 1 μM to 800 nM, 1 μM to 850 nM, 1 μM to 900 nM, 1 μM to 950 nM, less than 500 nM to 10 μM, less than 450 nM to 10 μM, less than 400 nM to 10 μM, less than 350 nM to 10 μM, less than 300 nM to 10 μM, less than 250 nM to 10 μM, less than 200 nM to 10 μM, less than 150 nM to 10 μM, less than 100 nM to 10 μM, less than 50 nM to 10 μM, less than 100 nM to 900 μM, less than 100 nM to 800 μM, less than 100 nM to 700 μM, less than 100 nM to 600 μM, less than 100 nM to 500 μM, less than 100 nM to 400 μM, less than 100 nM to 300 μM, less than 100 nM to 200 μM, less than 100 nM to 100 μM, less than 100 nM to 50 μM, or less than 100 nM to 10 μM.
Various methods are known in the art for determining KD for an aptamer's binding to its target (i.e., CD71). Jing and Bowser, Anal. Chim. Acta 686:9-18, which is incorporated herein by reference, reviews various approaches.
Cell Selection Quality Control and Traceless Cell Selection Methods Using Cell-Selected Aptamer Pools
Flow cytometry analysis along with other methods for cellular identification can be used to evaluate aptamer/cell interactions and the ability to select target cells using a given aptamer or combination of aptamers. OneComp eBeads™ (Invitrogen) can be used to prepare single-color controls for compensation, if needed. Stained biological samples can be analyzed, for example, using a MACSQuant™ Analyzer 10 (Miltenyi), Attune NxT (Invitrogen), or BD LSRFortessa™ (BD Biosciences) flow cytometer.
Traceless cell selection methods using cell-SELEX aptamer pools are described herein and demonstrated in the Examples.
Uses for Cells Selected on the Basis of Aptamer Binding
Large scale preparation or manufacturing of specific cell types is becoming increasingly useful for cellular-based therapies. The methods and compositions described herein provide an efficient approach for the isolation of specific target cell types that is readily scaled up for the large-scale isolation of cells for these and other uses.
Where the aptamers described herein can be used to isolate proliferating cells, such as cancer cells or fetal nucleated red blood cells, such isolated cells are specifically contemplated for use in cellular assays for research purposes, genetic analysis, or diagnosis of cancers (e.g., leukemias). Such cells can be manipulated and/or expanded prior to their use in such settings. Manipulations can include, as non-limiting examples, further cell sorting, stimulation with antigen, induction of differentiation, and/or genetic modification.
In some embodiments, the aptamers described herein can be used to “enrich” transferrin receptor-expressing cells in a cell population. By an enriched cell composition of such cells, it is meant that at least about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the cells express the transferrin receptor, or express the transferrin receptor at high levels. In some instances, the enriched composition will be a substantially pure population of cells expressing the transferrin receptor, whereby “substantially pure” it is meant at least 90% or more of the composition will be of the selected phenotype, e.g., 95%, 98%, and up to 100% of the population.
Depletion of Proliferating Cells for CAR T Cell Cancer Therapy
Currently, the generation and administration of autologous CAR T cell cancer therapy involves harvesting and genetically manipulating T cells before reintroducing the engineered cells back to patients. Prior to re-administration of manipulated T cells, it is important to remove substantially all cancerous cells to prevent the engraftment of genetically modified cancer cells and the generation of a new and possibly treatment-resistant cancer. A brief overview of CAR T cell technology is provided herein.
Chimeric Antigen Receptors (CARs) are recombinant proteins that allow T cells modified to express them to recognize a specific protein (antigen) on tumor cells. T cells engineered to express a CAR, referred to as CAR T cells, are expanded in the laboratory and then infused into the patient. After the infusion, the T cells multiply in the patient's body and, with guidance from their engineered receptor, recognize and kill cancer cells that display the antigen on their surfaces.
The first step in the process, cell harvesting, requires high purity isolation of desired cell populations. For example, it is known in the art that CAR T cells with defined 1:1 CD4+ to CD8+ cell populations are more potent than either pure (CD4+ or CD8+ only) and unselected populations in animal models of leukemia and are also very effective in human clinical trials for ALL.
T cells are typically isolated from peripheral blood mononuclear cells (PBMCs) collected by leukapheresis. The main methods for clinical-scale T cell isolation include (i) immunodepletion of undesired cells (e.g., proliferating or cancerous cells) followed by selection of T cell populations using antibody-conjugated magnetic beads (e.g., CliniMACS®) and (ii) “traceless” selection using Streptamer technology, which is based on fragment antigen-binding (Fab) constructs immobilized on magnetic beads. The aptamers described herein can be used to deplete transferrin receptor expressing cells (i.e., proliferating or cancerous cells) from the PBMCs collected by leukapheresis.
Examples of cancer that can be treated using cells prepared using CAR T cell technology include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More particular examples of such cancers include, but are not limited to, basal cell carcinoma, biliary tract cancer; bladder cancer; bone cancer; brain and CNS cancer; breast cancer; cancer of the peritoneum; cervical cancer; cholangiocarcinoma; choriocarcinoma; colon and rectal cancer; connective tissue cancer; cancer of the digestive system; endometrial cancer; esophageal cancer; eye cancer; cancer of the head and neck; gastric cancer (including gastrointestinal cancer); glioblastoma; hepatic carcinoma; hepatoma; intra-epithelial neoplasm; kidney or renal cancer; larynx cancer; leukemia; liver cancer; lung cancer (e.g., small-cell lung cancer, non-small cell lung cancer, adenocarcinoma of the lung, and squamous carcinoma of the lung); lymphoma including Hodgkin's and non-Hodgkin's lymphoma; melanoma; myeloma; neuroblastoma; oral cavity cancer (e.g., lip, tongue, mouth, and pharynx); ovarian cancer; pancreatic cancer; prostate cancer; retinoblastoma; rhabdomyosarcoma; rectal cancer; cancer of the respiratory system; salivary gland carcinoma; sarcoma; skin cancer; squamous cell cancer; stomach cancer; teratocarcinoma; testicular cancer; thyroid cancer; uterine or endometrial cancer; cancer of the urinary system; vulval cancer; as well as other carcinomas and sarcomas; as well as B-cell lymphoma (including low grade/follicular non-Hodgkin's lymphoma (NHL); small lymphocytic (SL) NHL; intermediate grade/follicular NHL; intermediate grade diffuse NHL; high grade immunoblastic NHL; high grade lymphoblastic NHL; high grade small non-cleaved cell NHL; bulky disease NHL; mantle cell lymphoma; AIDS-related lymphoma; and Waldenstrom's Macroglobulinemia); chronic lymphocytic leukemia (CLL); acute lymphoblastic leukemia (ALL); Hairy cell leukemia; chronic myeloblastic leukemia; and post-transplant lymphoproliferative disorder (PTLD), as well as abnormal vascular proliferation associated with phacomatoses, edema (such as that associated with brain tumors), tumors of primitive origins and Meigs' syndrome.
In addition to cancer, CAR T cells have also been generated as potential anti-HIV therapies. Recent studies by Hale et al., 2017 have shown that T cells utilizing CARs based on single-chain variable fragments (scFvs) derived from high-affinity broadly neutralizing antibodies (bNAbs) and containing second-generation co-stimulatory domains, in parallel with genetic protection from HIV can disrupt CCR5, and effectively target HIV-infected cells.
As used herein, the term “therapeutic cell composition” refers to a composition comprising a population of cells that are depleted of transferrin receptor-expressing cells (e.g., CD71 positive cells). Due to the potentially cancerous nature of transferrin receptor-expressing cells, the goal of depletion of transferrin receptor-expressing cells should be to remove all transferrin receptor-expressing cells. Such complete removal may require multiple rounds of depletion using the aptamers described herein or by use of, e.g., an anti-transferrin receptor antibody for a final round of cell depletion. It is also specifically contemplated herein that one or more additional aptamers can be used in combination with the aptamers described herein to deplete cells. For example, an aptamer that depletes monocytes and/or macrophages can be used in combination with the transferrin receptor-binding aptamers described herein. In one embodiment, the monocyte and/or macrophage binding aptamer comprises the sequence of:
In another embodiment, the monocyte and/or macrophage binding aptamer comprises the sequence of:
ATCCAGAGTGACGCAGCAGAAGAGTAGATGAAACGTTTTTTCGCCCGATAAAAG GGACGTGCGTCAGACATGGACACGGTGGCTTAGT (SEQ ID NO: 56). The therapeutic cells can then be expanded, differentiated in vitro, engineered, or formulated as desired before administration as a therapeutic cell composition.
As used herein, the term “depleting transferrin receptor-expressing cells” refers to the selective removal of cells expressing or highly expressing transferrin/CD71 from a sample comprising other cells, cell types or cell classes such that the resulting cell population lacks or has a reduced number of transferrin receptor-expressing cells. Target cells or a population thereof will generally be considered “depleted” as the term is used herein if the population includes less than 40% transferrin receptor-expressing cells (or cells with high expression of the transferrin receptor) resulting from a depletion method as described herein, and preferably includes less than 30%, less than 20%, less than 10%, less than 5%, less than 1% or more. In some embodiments, the therapeutic cell population following a depletion method as described herein will lack detectable numbers of cells expressing detectable levels of the transferrin receptor (i.e., cells that bind the aptamer). A population of cells is also “depleted” for transferrin receptor-expressing cells if the proportion of transferrin receptor-expressing cells is reduced following a depletion procedure by at least 50% relative to the proportion of transferrin receptor-expressing cells in the starting population. For such a depletion procedure, the population is “depleted” if the resulting population has a proportion of transferrin receptor-expressing cells that is reduced by at least 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99% or more relative to the starting population. Cells that are depleted of transferrin receptor-expressing cells are referred to herein as “therapeutic cells” or a “therapeutic cell composition.” Examples of therapeutic cell compositions depleted of TfR-expressing cells can include cell preparations enriched for NK cells, monocytes, macrophage, and the like. Similarly, the term “depletion” or “depleted” can be applied to the depletion of other cell types or cells expressing a given marker, for example, when using an aptamer that depletes monocytes and/or macrophages from the biological cell sample.
When the resulting therapeutic cells are to be genetically modified, for example production of CAR T cells, it is important to remove all transferrin receptor-expressing cells to prevent their genetic modification and potential escape from CAR T therapy. Multiple rounds of depletion and optionally one or more rounds of expansion can be used to ensure complete (i.e., 100%) removal of transferrin receptor-expressing cells. In addition, one or more rounds of depletion with a transferrin receptor antibody (e.g., in combination with aptamer-based depletion as described herein) can help to ensure the complete removal of such cells.
In one embodiment, the therapeutic cells are obtained directly from the subject to whom they are to be administered (i.e., autologous transplantation). In another embodiment, the transplantation can be non-autologous or allogeneic. As used herein, “allogeneic” refers to therapeutic cells (e.g., CAR T cells) obtained from one or more different donors of the same species, where the genes at one or more loci are not identical. For example, a therapeutic cell composition being administered to a subject can be derived from umbilical cord blood obtained from one more unrelated donor subjects, or from one or more non-identical siblings. In some embodiments, syngeneic cell populations can be used, such as those obtained from genetically identical animals, or from identical twins. In other embodiments of this aspect, the therapeutic cells are autologous cells; that is, the cells are obtained or isolated from a subject and administered to the same subject, i.e., the donor and recipient are the same.
For non-autologous transplantation, the recipient is preferably given an immunosuppressive drug to reduce the risk of rejection of the transplanted cell. Methods of administering cells include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, and epidural routes. The cells can be administered by any convenient route and can be administered together with other biologically active agents. The route of administration is preferably intravenous or intradermal. The titer of therapeutic cells to be transplanted or administered and which will be effective in the treatment of a particular disease or condition will depend on the nature of the disorder or condition and can be determined by standard clinical techniques. In addition, in vitro assays can optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each subject's circumstances.
In some respects, the methods provided herein comprise delivering a plurality of therapeutic cells or their progeny or differentiation product cells to a host tissue. In other aspects, the methods provided herein comprise delivering a plurality of cells that have been depleted of aptamer-binding cells (e.g., transferrin receptor-expressing cells). As described herein, therapeutic cell compositions following aptamer-mediated depletion of transferrin receptor-expressing cells can be delivered according to any method known in the art or can be incorporated into pharmaceutical compositions suitable for administration to a subject, e.g., for in vivo delivery to tissues, or organs of the subject.
The dosage ranges for the therapeutic cell composition includes amounts large enough to produce the desired effect, e.g., treatment of a disease or symptom thereof, e.g., cancer. The dosage should not be so large as to cause unacceptable adverse side effects. Generally, the dosage will vary with the particular characteristics of therapeutic cell composition, and with the age, condition, and sex of the patient. The dosage can be determined by one of skill in the art and, unlike traditional cell therapies, can also be adjusted by the individual physician in the event of any complication.
In some embodiments, the therapeutic cell composition is delivered for a repeated or limited amount of time. In some embodiments, the doses are given once a day, or multiple times a day. The duration of treatment depends upon the subject's clinical progress and responsiveness to therapy.
Compositions comprising a therapeutic cell population depleted of transferrin receptor-expressing cells can be delivered to target cells or tissues by surgical implantation, intravenous administration, intra-arterial administration, intraperitoneal administration, limb perfusion, (optionally, isolated limb perfusion of a leg and/or arm; see, e.g., Arruda et al., (2005) Blood 105:3458-3464), and/or direct intramuscular injection. Administration to a muscle (e.g., the diaphragm) can be by any suitable method including intravenous administration, intra-arterial administration, and/or intra-peritoneal administration.
In some embodiments, one or more additional compounds can also be included with the therapeutic cell composition (i.e., a cell population depleted of aptamer-binding cells) to alleviate symptoms of a disease or to otherwise assist or support the function of the administered cells.
In some embodiments, the additional compound can be a therapeutic agent. The therapeutic agent can be selected from any class suitable for the therapeutic objective. In other words, the therapeutic agent can be selected according to the treatment objective and biological action desired. Furthermore, the active ingredients of the therapeutic agent can be mixed with optional pharmaceutical additives such as excipients or carriers which are pharmaceutically acceptable and compatible with the active ingredient.
Therapeutic cells can further comprise a targeting moiety for a tissue of interest. For example, the targeting moiety can comprise a receptor molecule, including receptors that naturally recognize a specific desired molecule of a target cell (e.g., a tumor cell). Such receptor molecules include receptors that have been modified to increase their specificity of interaction with a target molecule, receptors that have been modified to interact with a desired target molecule not naturally recognized by the receptor, and fragments of such receptors (see, e.g., Skerra, 2000, J. Molecular Recognition, 13:167-187). In other embodiments, the targeting moiety can comprise a ligand molecule, including, for example, ligands which naturally recognize a specific desired receptor on a target cell. Such ligand molecules include ligands that have been modified to increase their specificity of interaction with a target receptor, ligands that have been modified to interact with a desired receptor not naturally recognized by the ligand, and fragments of such ligands. In other embodiments, the targeting moiety can comprise an aptamer that has not been used in the initial cell selection as described herein.
For use in the various aspects described herein, an effective amount of therapeutic cells (e.g., low or lacking expression of CD71) comprises at least 102 cells, at least 5×102 cells, at least 103 cells, at least 5×103 cells, at least 104 cells, at least 5×104 cells, at least 105 cells, at least 2×105 cells, at least 3×105 cells, at least 4×105 cells, at least 5×105 cells, at least 6×105 cells, at least 7×105 cells, at least 8×105 cells, at least 9×105 cells, at least 1×106 cells, at least 2×106 cells, at least 3×106 cells, at least 4×106 cells, at least 5×106 cells, at least 6×106 cells, at least 7×106 cells, at least 8×106 cells, at least 9×106 cells, at least 1×107 cells, at least 2×107 cells, at least 3×107 cells, at least 4×107 cells, at least 5×107 cells, at least 6×107 cells, at least 7×107 cells, at least 8×107 cells, at least 9×107 cells, at least 1×108 cells, at least 2×108 cells, at least 3×108 cells, at least 4×108 cells, at least 5×108 cells, at least 6×108 cells, at least 7×108 cells, at least 8×108 cells, at least 9×108 cells, at least 1×109 cells, at least 2×109 cells, at least 5×109 cells, at least 1×1010 cells, at least 2×109 cells, at least 5×109 cells, at least 1×1010 cells, at least 2×1010 cells, at least 5×1010 cells, at least 1×1011 cells, at least 2×1011 cells, at least 5×1011 cells, at least 1×1012 cells, at least 2×1012 cells, at least 5×1012 cells, at least 1×1013 cells, or multiples thereof. In some embodiments of the aspects described herein, the therapeutic cells are expanded in culture prior to administration to a subject in need thereof. In some embodiments, the therapeutic cells are CAR T cells.
In one embodiment, the term “effective amount” as used herein refers to the amount of a population of therapeutic cells (e.g., those cells having low or undetectable levels of the transferrin receptor) needed to alleviate at least one or more symptom of a disease (e.g., cancer), and relates to a sufficient amount of a composition to provide the desired effect, e.g., treat a subject having a disease, such as cancer. The term “therapeutically effective amount” therefore refers to an amount of therapeutic cells or a composition thereof that is sufficient to promote a particular effect when administered to a typical subject, such as one who has or is at risk of cancer. An effective amount as used herein would also include an amount sufficient to prevent or delay the development of a symptom of the disease, alter the course of a symptom disease (for example but not limited to, slow the progression of a symptom of the disease), or reverse a symptom of the disease. It is understood that for any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using routine experimentation.
As used herein, “administered” refers to the delivery of a composition comprising therapeutic cells as described herein into a subject by a method or route which results in at least partial localization of the cell composition at a desired site. A cell composition can be administered by any appropriate route which results in effective treatment in the subject, i.e., administration results in delivery to a desired location in the subject where at least a portion of the composition delivered, i.e., at least 1×104 therapeutic cells are delivered to the desired site for a period of time. Modes of administration include injection, infusion, instillation, or ingestion. “Injection” includes, without limitation, intravenous (iv), intramuscular (im), intra-arterial, intrathecal, intraventricular, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, intracerebro spinal, and intrasternal injection and infusion. For the delivery of therapeutic cells, a composition thereof, or an aptamer-active agent complex, administration by injection or infusion is generally preferred.
In one embodiment, such therapeutic cells or aptamer-active agent complex as described herein are administered systemically. The phrases “systemic administration,” “administered systemically”, “peripheral administration” and “administered peripherally” as used herein refer to the administration of a population of therapeutic cells other than directly into a target site, tissue, or organ, such that it enters, instead, the subject's circulatory system and, thus, is subject to metabolism and other like processes.
The efficacy of a treatment comprising a therapeutic cell composition, or an aptamer-active agent complex as described herein for the treatment of a given disease can be determined by the skilled clinician. However, a treatment is considered “effective treatment,” as the term is used herein, if any one or all the signs or symptoms of the disease are altered in a beneficial manner, other clinically accepted symptoms or markers of disease are improved or ameliorated, e.g., by at least 10% following treatment with the therapeutic cells. Efficacy can also be measured by failure of an individual to worsen as assessed by hospitalization or need for medical interventions (e.g., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein. Treatment includes any treatment of a disease in an individual or an animal (some non-limiting examples include a human, or a mammal) and includes: (1) inhibiting the disease, e.g., arresting, or slowing the progression of a given disease; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of disease.
Following in vitro or ex vivo cell culture, isolation, or differentiation as described herein, isolated or enriched therapeutic cells are prepared for treatment and/or implantation. The cells are suspended in a physiologically compatible carrier, such as cell culture medium (e.g., Eagle's minimal essential media), phosphate buffered saline, or a T cell specific medium. The volume of cell suspension to be implanted will vary depending on the site of implantation, treatment goal, and cell density in the solution.
It will be appreciated by one of skill in the art that a cell composition useful for treating a given disease does not need to be a pure, homogeneous culture of, e.g., therapeutic cells lacking expression of transferrin receptor or having low expression of the transferrin receptor. Accordingly, in one embodiment, the composition administered comprises at least 2% therapeutic cells. In other embodiments, the composition comprises at least 3%, at least 4%, at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 99% or more therapeutic cells as described herein.
The cells can be administered to a subject by any appropriate route that results in delivery of the therapeutic cells to a desired location in the subject where at least a portion of the cells remain viable. It is preferred that at least 5% remain viable. In other embodiments, at least 10%, at least 20%, at least 30%, at least 40%, or at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, at least 98%, at least 99% or more of the therapeutic cells remain viable after administration into a subject. The period of viability of the therapeutic cells after administration to a subject can be as short as a few hours, e.g., twenty-four hours, to a few days, to as long as a few weeks to months.
To accomplish these methods of administration, the therapeutic cell composition(s) can be inserted into a delivery device that facilitates introduction by injection or implantation of the therapeutic cells into the subject. Typically, the therapeutic cells are injected into the target area as a cell suspension. Alternatively, the cells can be embedded in a solid or semisolid support matrix when contained in such a delivery device.
In some embodiments, administration of a composition comprising therapeutic cells is repeated after a given interval of time (e.g., one day, three days, one week, two weeks, three weeks, one month or more. Repeated treatments can be performed, for example, to establish or maintain a threshold level of engraftment necessary to continue effective treatment, as necessary. In some embodiments, the method is repeated twice, three times, four times, five times or more.
Aptamer Therapeutics and Other Uses
Aptamers have been widely used as targeting ligands to mediate delivery of a variety of payloads to cellular or intracellular targets (e.g., HER2, nucleolin). Payloads include therapeutic drugs, imaging compounds, unstable molecules (e.g., nucleic acids). or ribonucleoproteins. These aptamer pay load complexes can be termed “aptamer-active agent complexes.” In cancer therapy, aptamers can be used to facilitate delivery of therapeutics to cancer cells with high targeting efficiency. In many cancers, tumor cells upregulate specific biomarkers; targeted aptamers can guide nanomedicine systems to deliver therapeutic payloads to these cells, reducing off-target toxicity. These nanomedicine platforms include liposomes, polymeric micelles, or iron oxide nanoparticles. Efficacy of these platforms has been demonstrated both in vitro and in vivo. (See e.g., Alshaer, W. et al. Aptamer-guided nanomedicines for anticancer drug delivery. (2018) Adv. Drug Deliv. Rev. 134:122-137; Ray and White; Aptamers for Targeted Drug Delivery. (2010) Pharmaceuticals 3:1761-1778; Meng, H-M. et al. Aptamer-integrated DNA nanostructures for biosensing, bioimaging and cancer therapy. (2016) Chem. Society Rev. 45:2583-2602).
Thus, specifically contemplated herein is the use of the aptamers described herein as a drug delivery device for targeting a given drug to TfR positive cells or other tissues. Also contemplated herein is the use of such aptamers to target an active agent, such as, for example, a therapeutically active agent to TfR positive cells. In certain embodiments the aptamer is attached to the therapeutically active agent.
In another embodiment, the aptamers described herein are contemplated for use as sensors or as a theranostic. Some aptamers can undergo significant conformation change upon binding to their ligand or receptor. These switching aptamers can be labeled with a reporter molecule (e.g., a fluorophore for fluorescence detection or a redox probe for electrochemical detection) to provide a signal change upon ligand binding. In another example, aptamers can also be designed to be in duplex structures until binding to their target; binding releases a fluorescently labeled complementary strand, called a “beacon” (see e.g., Tuleuova and Revzin. Micropatterning of aptamer beacons to create cytokine-sensing surfaces. (2010) Cell Mol. Bioeng. 3:337-344; Iliuk, et al. Aptamer in bioanalytical applications. (2011) Anal. Chem. 83:4440-4452). The aptamers presented in this application can similarly be used to report the presence and relative number of TfR positive cells by engineering the aptamer into an aptamer reporter.
Pharmaceutical Compositions, Administration and Efficacy
Pharmaceutical or therapeutic compositions comprising an aptamer, aptamer-therapeutically active agent complex, or aptamer-depleted cells for the treatment of a given disease or disorder can contain a physiologically tolerable carrier, wherein the therapeutic agent is dissolved or dispersed therein as an active ingredient(s). In a preferred embodiment, the pharmaceutical composition is not immunogenic when administered to a mammal or human patient for therapeutic purposes. As used herein, the terms “pharmaceutically acceptable”, “physiologically tolerable” and grammatical variations thereof, as they refer to compositions, carriers, diluents, and reagents, are used interchangeably and represent that the materials are capable of administration to or upon a mammal without the production of undesirable physiological effects such as nausea, dizziness, gastric upset, and the like. A pharmaceutically acceptable carrier will not promote the raising of an immune response to an agent with which it is admixed, unless so desired. The preparation of a pharmacological or pharmaceutical composition that contains active ingredients dissolved or dispersed therein is well understood in the art and need not be limited based on formulation. Typically, such compositions are prepared as injectable either as liquid solutions or suspensions, however, solid forms suitable for solution, or suspensions, in liquid prior to use can also be prepared. The preparation can also be emulsified or presented as a liposome composition. The active ingredient can be mixed with excipients which are pharmaceutically acceptable and compatible with the active ingredient and in amounts suitable for use in the therapeutic methods described herein. Suitable excipients include, for example, water, saline, dextrose, glycerol, ethanol or the like and combinations thereof. In addition, if desired, the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents and the like which enhance the effectiveness of the active ingredient. The therapeutic composition comprising an aptamer or a population of aptamer-enriched cells for treatment of a disease or disorder can include pharmaceutically acceptable salts of the components therein. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide) that are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, tartaric, mandelic and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
Physiologically tolerable carriers are well known in the art. Exemplary liquid carriers are sterile aqueous solutions that contain no materials in addition to the active ingredients and water or contain a buffer such as sodium phosphate at physiological pH value, physiological saline, or both, such as phosphate-buffered saline. Still further, aqueous carriers can contain more than one buffer salt, as well as salts such as sodium and potassium chlorides, dextrose, polyethylene glycol and other solutes. Liquid compositions can also contain liquid phases in addition to and to the exclusion of water. Exemplary of such additional liquid phases are glycerin, vegetable oils such as cottonseed oil, and water-oil emulsions. The amount of an active agent used in the methods described herein that will be effective in the treatment of disease or a symptom thereof will depend on the nature of the disorder or condition and can be determined by standard clinical techniques.
A pharmaceutical composition as described herein can be formulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multidose containers with, optionally, an added preservative. The compositions can be suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.
Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients can be prepared as appropriate oily or water-based injection suspensions.
Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters such as ethyl oleate, triglycerides, or liposomes. Aqueous injection suspensions can contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran.
Optionally, the suspension can also contain suitable stabilizers or agents that increase the solubility of the active ingredients, to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient can be in powder form for constitution with a suitable vehicle, e.g., a sterile, pyrogen-free, water-based solution, before use.
In some embodiments, a therapeutic aptamer-active agent complex can be delivered in an immediate release form. In other embodiments, the therapeutic aptamer-active agent complex composition can be delivered in a controlled-release system or sustained-release system. Controlled- or sustained-release pharmaceutical compositions can have a common goal of improving drug therapy over the results achieved by their non-controlled or non-sustained-release counterparts. Advantages of controlled- or sustained-release compositions include extended activity of the therapeutic agents, reduced dosage frequency, and increased compliance. In addition, controlled- or sustained-release compositions can favorably affect the time of onset of action or other characteristics, such as blood levels of the therapeutic agent, and can thus reduce the occurrence of adverse side effects. Controlled- or sustained-release of an active ingredient can be stimulated by various conditions, including but not limited to, changes in pH, changes in temperature, concentration or availability of enzymes, concentration or availability of water, or other physiological conditions or compounds.
In one embodiment, a pump can be used (Langer, (1990) Science 249:1527-1533; Sefton, (1987) CRC Crit. Ref Biomed. Eng. 14:201; Buchwald et al. (1980) Surgery 88:507; and Saudek et al. (1989) N. Engl. J. Med. 321:574). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release (Langer and Wise eds., 1974); Controlled Drug Bioavailability, Drug Product Design and Performance (Smolen and Ball eds., 1984); Ranger and Peppas, (1983) J. Macromol. Sci. Rev. Macromol. Chem. 23:61; Levy et al. (1985) Science 228:190; During et al. (1989) Ann. Neurol. 25:351; and Howard et al. (1989) J. Neurosurg. 71:105). In yet another embodiment, a controlled- or sustained-release system can be placed in proximity of a target of infection, thus requiring only a fraction of the systemic dose.
When in tablet or pill form, a pharmaceutical composition as described herein can be coated (e.g., enterically coated) to delay disintegration and absorption in the gastrointestinal tract, thereby providing a sustained action over an extended period of time. Selectively permeable membranes surrounding an osmotically active driving compound are also suitable for orally administered compositions. In these latter platforms, fluid from the environment surrounding the capsule is imbibed by the driving compound, which swells to displace the agent or agent composition through an aperture. These delivery platforms can provide an essentially zero order delivery profile as opposed to the spiked profiles of immediate release formulations. A time-delay material such as glycerol monostearate or glycerol stearate can also be used. Oral compositions can include standard excipients such as mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, and magnesium carbonate. In one embodiment, the excipients are of pharmaceutical grade.
A pharmaceutical composition as described herein can also be formulated in rectal compositions such as suppositories or retention enemas, using, for example, conventional suppository bases such as cocoa butter or other glycerides.
The appropriate dosage range for a given therapeutic agent depends upon the potency and includes amounts large enough to produce the desired effect, e.g., reduction in at least one symptom of a given disease. The dosage of the therapeutic agent should not be so large as to cause unacceptable or life-threatening adverse side effects or should be used under close supervision by a medical professional. Generally, the dosage will vary with the specific aptamer and/or formulation, and with the age, condition, and sex of the patient. The dosage can be determined by one of skill in the art and can also be adjusted by the individual physician in the event of any complication.
Typically, the dosage of a given therapeutic aptamer and/or aptamer-active agent complex, can range from 0.001 mg/kg body weight to 5 g/kg body weight. In some embodiments, the dosage range is from 0.001 mg/kg body weight to 1 g/kg body weight, from 0.001 mg/kg body weight to 0.5 g/kg body weight, from 0.001 mg/kg body weight to 0.1 g/kg body weight, from 0.001 mg/kg body weight to 50 mg/kg body weight, from 0.001 mg/kg body weight to 25 mg/kg body weight, from 0.001 mg/kg body weight to 10 mg/kg body weight, from 0.001 mg/kg body weight to 5 mg/kg body weight, from 0.001 mg/kg body weight to 1 mg/kg body weight, from 0.001 mg/kg body weight to 0.1 mg/kg body weight, from 0.001 mg/kg body weight to 0.005 mg/kg body weight. Alternatively, in some embodiments the dosage range is from 0.1 g/kg body weight to 5 g/kg body weight, from 0.5 g/kg body weight to 5 g/kg body weight, from 1 g/kg body weight to 5 g/kg body weight, from 1.5 g/kg body weight to 5 g/kg body weight, from 2 g/kg body weight to 5 g/kg body weight, from 2.5 g/kg body weight to 5 g/kg body weight, from 3 g/kg body weight to 5 g/kg body weight, from 3.5 g/kg body weight to 5 g/kg body weight, from 4 g/kg body weight to 5 g/kg body weight, from 4.5 g/kg body weight to 5 g/kg body weight, from 4.8 g/kg body weight to 5 g/kg body weight. In one embodiment, the dose range is from 5 μg/kg body weight to 30 μg/kg body weight. Alternatively, the dose range will be titrated to maintain serum levels between 5 μg/mL and 30 μg/mL.
Administration of the doses recited above or as employed by a skilled clinician can be repeated for a limited and defined period of time. In some embodiments, the doses are given once a day, or multiple times a day, for example, but not limited to three times a day. Typically, the dosage regimen is informed by the half-life of the agent as well as the minimum therapeutic concentration of the agent in blood, serum or localized in a given biological tissue. In a preferred embodiment, the doses recited above are administered daily for several weeks or months. The duration of treatment depends upon the subject's clinical progress and continued responsiveness to therapy. Continuous, relatively low maintenance doses are contemplated after an initial higher therapeutic dose.
A therapeutically effective amount is an amount of an agent that is sufficient to produce a statistically significant, measurable change of a given symptom of disease (see “Efficacy Measurement” below). Such effective amounts can be gauged in clinical trials as well as animal studies for a given agent. For example, reduction of a given symptom of disease can be indicative of adequate therapeutic efficacy of an agent(s).
Agents useful in the methods and compositions described herein can be administered topically, intravenously (by bolus or continuous infusion), orally, by inhalation, intraperitoneally, intramuscularly, subcutaneously, intracavity, and can be delivered by peristaltic means, if desired, or by other means known by those skilled in the art. The agent can be administered systemically, if so desired.
Therapeutic compositions containing at least one therapeutic agent can be conventionally administered in a unit dose. The term “unit dose” when used in reference to a therapeutic composition refers to physically discrete units suitable as unitary dosage for the subject, each unit containing a predetermined quantity of a therapeutic agent calculated to produce the desired therapeutic effect in association with the required physiologically acceptable diluent, i.e., carrier, or vehicle.
The compositions are administered in a manner compatible with the dosage formulation, and in a therapeutically effective amount. The quantity to be administered and timing depends on the subject to be treated, capacity of the subject's system to utilize the active ingredient, and degree of therapeutic effect desired. An agent can be targeted by means of a targeting moiety, such as, e.g., an antibody or targeted liposome technology.
Precise amounts of active ingredient required to be administered depend on the judgment of the practitioner and are particular to each individual. However, suitable dosage ranges for systemic application are disclosed herein and depend on the route of administration. Suitable regimes for administration are also variable but are typified by an initial administration followed by repeated doses at one or more intervals by a subsequent injection or other administration. Alternatively, continuous intravenous infusion sufficient to maintain concentrations in the blood in the ranges specified for in vivo therapies are contemplated.
In some embodiments, at least one additional therapeutic agent is used in combination with the aptamers or cells described herein for the treatment of a given disease.
In some embodiments, a therapeutic aptamer composition is administered to a subject concurrently with a combination therapy. As used herein, the term “concurrently” is not limited to the administration of the two or more agents at exactly the same time, but rather, it is meant that they are administered to a subject in a sequence and within a time interval such that they can act together (e.g., synergistically to provide an increased benefit than if they were administered otherwise). For example, the combination of therapeutics can be administered at the same time or sequentially in any order at different points in time; however, if not administered at the same time, they should be administered sufficiently close in time so as to provide the desired therapeutic effect, preferably in a synergistic fashion. The agents can be administered separately, in any appropriate form and by any suitable route. When each of the therapeutic agents in a combination are not administered in the same pharmaceutical composition, it is understood that they can be administered in any order to a subject in need thereof. For example, the first therapeutic agent can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of the second therapeutic agent, to a subject in need thereof (or vice versa). In other embodiments, the delivery of either therapeutic agent ends before the delivery of the other agent/treatment begins. In some embodiments of either case, the treatment is more effective because of combined administration. For example, the therapeutic agents used in combination are more effective than would be seen with either agent alone. In some embodiments, delivery is such that the reduction in a symptom, or other parameter related to the disorder is greater than what would be observed with either therapeutic agent alone. The effect of such a combination can be partially additive, wholly additive, or greater than additive. The agent and/or other therapeutic agents, procedures or modalities can be administered during periods of active disease, or during a period of persistence or less active disease.
When administered in combination, one or more of the therapeutic agents can be administered in an amount or dose that is higher, lower or the same as the amount or dosage of the given agent used individually, e.g., as a monotherapy. In certain embodiments, the administered amount or dosage of a first therapeutic agent when administered in combination with a second therapeutic agent is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50%) than the amount or dosage of the first agent when used individually. In other embodiments, the amount or dosage of a first therapeutic agent, when administered in combination with a second therapeutic agent, results in a desired effect (e.g., improved cognitive functioning) is lower (e.g., at least 20%, at least 30%, at least 40%, or at least 50% lower) than the amount or dosage of the first (or second) agent required to achieve the same therapeutic effect when administered alone.
The efficacy of a treatment for a given disease can be determined by the skilled clinician. However, a treatment is considered “effective treatment,” as the term is used herein, if any one or all the signs or symptoms of the given disease is/are altered in a beneficial manner, or other clinically accepted symptoms or markers of disease are improved, or ameliorated, e.g., by at least 10% following treatment with a therapeutic aptamer or a population of therapeutic cells as described herein. Efficacy can also be measured by failure of an individual to worsen as assessed by stabilization of the disease, or the need for medical interventions (i.e., progression of the disease is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein. Treatment includes any treatment of a disease in an individual or an animal (some non-limiting examples include a human, or a mammal) and includes: (1) inhibiting the disease, e.g., arresting, or slowing progression of the disease; or (2) relieving the disease, e.g., causing regression of symptoms; and (3) preventing or reducing the likelihood of the development of the disease, or preventing secondary diseases/disorders associated with the disease.
An effective amount for the treatment of a disease means that amount which, when administered to a mammal in need thereof, is sufficient to result in effective treatment as that term is defined herein, for that disease. Efficacy of an agent can be determined by assessing physical indicators of the disease, such as, e.g., pain, fatigue, fever, and the like.
Kits
In one aspect, provided herein are kits containing any one or more of the elements disclosed in the above methods and compositions. Provided herein are kits comprising aptamers as described herein, and formulations thereof. In one embodiment, the kit comprises, consists of, or consists essentially of reagents and instructions for selecting or depleting transferrin receptor-expressing cells from a cell population. In other embodiments, the kit comprises, consists of, or consists essentially of reagents and instructions for genomic or phenotypic analysis of aptamer-selected CD71-expressing cells, generating CAR T cells from a population of cells depleted for aptamer-binding cells and administering such CAR T cells.
In certain embodiments, the kits comprise one or more solid supports or an aptamer conjugated to a solid support. Kits can also comprise columns, including but not limited to affinity chromatography columns and flow cytometry columns.
The kit can comprise cell culture or preparation reagents including labeling means, salts, growth media, serum etc.
In some embodiments, a kit comprises one or more reagents for use in a process utilizing one or more of the elements described herein. Reagents can be provided in any suitable container. For example, a kit can provide one or more reaction or storage buffers. A buffer can be any buffer, including but not limited to a sodium carbonate buffer, a sodium bicarbonate buffer, a borate buffer, a Tris buffer, a MOPS buffer, a HEPES buffer, and combinations thereof. In some embodiments, the buffer is alkaline. In some embodiments, the buffer has a pH from about 7 to about 10.
It is understood that the preceding detailed description and the following examples are illustrative only and are not to be taken as limitations upon the scope of the invention. Various changes and modifications to the disclosed embodiments, which will be apparent to those of skill in the art, may be made without departing from the spirit and scope of the present invention. Further, all patents, patent applications, and publications identified are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the present invention. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents are based on the information available to the applicants and do not constitute any admission as to the correctness of the dates or contents of these documents.
The following provides non-limiting Examples demonstrating and supporting the technology as described herein.
Chimeric antigen receptor (CAR) T-cell therapy has gained significant traction in the oncology field, with five FDA-approved therapies to-date: four for treating relapsed or refractory (r/r) CD19+ B-cell malignancies (Novartis's Kymriah®, Gilead-Kite's Yescarta™ and Tecartus™, and Bristol Myers Squibb-Juno Therapeutic's Breyanzi™) and another two for treating r/r BCMA+ multiple myeloma (Bristol Myers Squibb-bluebird bio's Abecma™ and Janssen Pharmaceuticals-Legend Biotech's Carvykti™).1-8 In these treatments, a patient's T cells journey through an elaborate manufacturing process that consists of 1) enrichment from a leukapheresis product, 2) activation ex vivo, 3) lentiviral or retroviral expression of a CAR that directs T-cell function against a tumor-associated antigen, 4) expansion to therapeutically relevant numbers, and 5) re-infusion into the patient's body for cancer elimination.9 Given the intricate nature of this operation, there is a continual need for further innovation at each production step to reduce the costs and increase the efficacy and safety of these adoptive T-cell therapies.10
T-cell enrichment or selection is a pivotal steps in CAR T-cell manufacturing, as the cell composition and purity used in subsequent activation and transduction steps can influence the outcome of the therapy. For Kymriah®, Yescarta™, and Abecma™, T cells are indirectly enriched by collecting peripheral blood mononuclear cells (PBMCs) from leukapheresis product using counterflow centrifugal elutriation, which removes most monocytes, granulocytes, platelets, and residual red blood cells based on differences in cell size and density relative to lymphocytes.11-13 However, this selection approach is unable to discriminate healthy T cells from circulating cancerous lymphocytes, meaning that tumor cells can be present in downstream manufacturing steps and thereby drive uncontrollable activation and exhaustion of the CAR T-cell product.14 Additionally, transduction of a single leukemic B cell with the CAR gene during CAR T-cell manufacturing caused in cis epitope masking that led to a patient's relapse and eventual death.15 These issues highlight the need for complete removal of cancerous lymphocytes during the selection step prior to further CAR T-cell manufacturing.
To address this problem, especially for patients with high circulating blast and leukemia cell counts, Tecartus™, Breyanzi™, and Carvykti™ rely on direct isolation of T cells. Whereas Tecartus™ and Carvykti™ isolates bulk CD3+ T cells,5, 16 Breyanzi™ separately isolates helper CD4+ T cells and cytotoxic CD8+ T cells for CAR T-cell production and later infuses the patient with a defined 1:1 composition of the subsets.6 Breyanzi is associated with lower rates of cytokine release syndrome and neurotoxicity than Kymriah® and Yescarta™ while remaining equally effective.2, 3, 6 However, the direct isolation of T cells is expensive, typically relying on costly antibody- or multimerized Fab-coated magnetic beads that target either the CD3, CD4, and/or CD8 T-cell markers for positive enrichment or unwanted immune cell markers for negative enrichment.17 Furthermore, these approaches do not actively remove the cancer cells from a patient's leukapheresis product. In contrast to B-cell malignancies, malignant T cells can be difficult to separate from healthy T cells used for manufacturing CAR T cell therapies.18, 19 Accordingly, as autologous CAR T-cell therapies are broadened to treat diverse hematological malignancies, an inexpensive and universal method for removing cancerous cells from healthy PBMCs will be imperative for their safe manufacturing.
DNA aptamers, single-stranded oligonucleotides that fold into sequence-specific secondary structures, are molecular recognition agents that can address the current deficiencies of cancer cell removal in adoptive T-cell manufacturing. Aptamers can bind their targets with affinities comparable to antibodies, but unlike antibodies they are synthesized chemically with high reproducibility and relatively low cost.20 Aptamers can also be controllably modified at any position (5′ end, 3′ end, or internally) for facile immobilization onto solid supports (e.g., hydrogels) for affinity-based separations.21 Aptamer binding to their targets can also be reversed by disrupting aptamer structure, for example, by use of a complementary sequence.21, 22 Demonstrating these points, the inventors previously identified CD8-binding DNA aptamers and used them to isolate CD8+ T cells via magnetic-activated cell sorting (MACS) with comparable purity, yield, and downstream CAR functionality as those isolated from commercial antibody-based methods.23
Aptamers can be selected to bind certain cell types without prior knowledge of receptor identity (i.e., receptor-agnostic panning).24-26 The subtractive evolutionary process in which aptamers go through rounds of positive and negative selection against whole cells, termed cell-SELEX (systematic evolution of ligands by exponential enrichment), has become an attractive method for discovering aptamers that can differentiate malignant cells from healthy normal cells.27 Given these benefits, cell-SELEX holds great promise for discovering DNA aptamers that can selectively bind and deplete circulating leukemia and lymphoma cells from PBMCs at low cost prior to CAR T-cell manufacturing.
Here, in a cell-SELEX intended to identify T-cell antigen-binding aptamers, the inventors discovered and truncated an aptamer, named tJBA8.1, that displays high-affinity binding for Jurkat T-leukemia cells. Further assays identified transferrin receptor 1 (TfR1), an iron-uptake receptor not expressed on resting immune cells but upregulated on actively dividing and cancerous cells, as the binding target of tJBA8.1. The tJBA8.1-TfR1 interaction was characterized by flow cytometry, biolayer interferometry, and competition studies with holo-transferrin, and further compare its binding properties to a previously reported TfR1-binding aptamer. The inventors further employed tJBA8.1 in MACS to remove spiked Raji B-lymphoma cells from PBMCs with high yield and minimal impact on the healthy immune cell composition. Lastly, the off-target binding of tJBA8.1 was studied and a point mutation to the aptamer sequence for affinity improvement are described. Given the broad expression of TfR1 on many cancers, including difficult to distinguish T-cell leukemias and lymphomas, it is contemplated that this method can be universally used for the depletion of circulating cancer cells from patient PBMCs prior to downstream CAR T-cell manufacturing, leading to a safer, more potent, and cost-mindful therapy.
Discovery of the Jurkat-Binding Aptamer 8.1 (JBA8.1) by Cell-SELEX and Stem Truncation to tJBA8.1
In an initial effort to identify aptamers that bind the human CD3 and CD28 T-cell receptors, cell-SELEX was performed using CD3+CD28+ Jurkat T-leukemia cells for positive selection and CD3−CD28− J.RT3-T3.5 cells for negative/counter selection (
Prior to cell binding, aptamer sequences were folded by heating in DPBS supplemented with approximately 0.9 mM CA2+ and approximately 5.5 mM Mg2+ ions for 5 min at 95° C. followed by snap cooling on ice (See infra for details). Flow cytometry binding of aptamer pools from the individual SELEX rounds revealed substantial binding to both Jurkat and J.RT3-T3.5 cells starting in round 5 that plateaued by round 8 (
CGAATTGC
TTAATCGA
GTCCATTC
CTGATTCT
TGGCAGTT
CGTTACTT
CATTGAGT
ACCTCTTG
TTAATCGA
FASTAptamer™ toolkit was used to analyze NGS results and calculate fold-enrichment of unique aptamer sequences over rounds of cell-SELEX.29 Unique sequence reads were low for rounds 1-4, reflecting high aptamer pool diversity until rounds 5-8. These results are consistent with the cell-SELEX round binding results in
Nine Jurkat-binding aptamers from round 8 listed in Table 6 and named JBA8.X (where “X” is the aptamer's rank in Table 5) were chosen for cell binding based on their representation, motif, and enrichment across rounds.
None of the selected fluorescein-labeled aptamers displayed specific binding for Jurkat cells over J.RT3-T3.5 cells, indicating that the SELEX process did not enrich CD3- or CD28-binding aptamers (
Truncation and Binding Affinity of the JBA8.1 Aptamer
To reduce aptamer productions costs in downstream assays, the inventors first sought to truncate the JBA8.1 aptamer. Using the NUPACK™ application to predict the minimum free energy (MFE) structure of JBA8.1,33 it was found that the simulated structure of JBA8.1 conforms well to the intended library design, with the majority of the 45-bp Motif 3 forming a multi-hairpin structure that sits on top of a stem comprised of the partially complementary 18-nt flanking primer sequences (
Identification and Validation of Transferrin Receptor 1 (TfR1) as a Target of tJBA8.1
To investigate the cellular compartment and type of molecule targeted by tJBA8.1, Jurkat cells were labeled with tJBA8.1 at 4° C. followed by enzymatic treatment with trypsin, a cell-impermeable serine endopeptidase that cleaves many extracellular proteins. tJBA8.1 binding was almost completely abolished after trypsin treatment (
Having demonstrated that tJBA8.1 binds to an extracellular membrane protein, the inventors next adapted a reported aptamer-based pull-down assay designed for identification of target membrane receptors.34 Briefly, cell membrane proteins extracted and solubilized from Jurkat cells were depleted for non-specific DNA-binding proteins using the RANL aptamer before incubation with biotinylated tJBA8.1. Aptamer-bound membrane proteins were then purified using streptavidin-coated magnetic beads and stratified by molecular weight via SDS-PAGE. Compared to a control sample that was purified with biotin-saturated streptavidin beads without tJBA8.1, two distinct protein bands that were highly enriched by tJBA8.1 were observed at approximately 200 kDa (band a) and 100 kDa (band b) (
To validate that tJBA8.1 binds TfR1, short interfering RNA (siRNA) duplexes (Table 7) were used to knockdown the expression of TfR1 encoded by the TFRC gene in Jurkat cells and evaluated aptamer binding.
Compared to cells that were nucleofected with non-specific (NS) siRNA, cells nucleofected with TFRC siRNA had 51% reduced TfR1 expression as evaluated by anti-CD71 antibody (CD71 Ab) staining, which matched closely with the observed 49% reduction in tJBA8.1 binding (
Characterization of tJBA Binding to TfR1 and Competition with Other Ligands
To better elucidate the interaction between tJBA8.1 and TfR1, Jurkat cells were co-stained with CD71 Ab (clone CY1G4) and tJBA8.1. A striking positive correlation was observed between CD71 Ab and tJBA8.1 staining, demonstrating that tJBA8.1 can bind TfR1 on cells simultaneously with the CD71 Ab (
Whether aptamers JBA8.3, JBA8.4, JBA8.7, JBA8.11, and JBA8.15 bind to TfR1 was also tested by BLI. Despite these aptamers displaying statistically significant binding to Jurkat cells in
The inventors also evaluated binding of His-tagged mouse recombinant TfR1 protein to immobilized tJBA8.1 by BLI, but results were negative despite positive control antibody binding, suggesting that tJBA8.1 does not target mouse TfR1 (
Iron is delivered intracellularly to cells via a transferrin cycle. Specifically, iron-bound transferrin (holo-Tf) binds to TfR1 for uptake, after which iron is released from transferrin under acidic endosomal pH and the resulting iron-free transferrin (apo-Tf) is recycled to the cell surface for dissociation under neutral pH.40 As demonstrated in
Comparison of tJBA8.1 with a Reported TfR1-Binding DNA Aptamer
XQ-2d is a 56-bp truncated aptamer previously discovered by the Tan group that also targets TfR1.42,43 Aligning the variable sequence regions of tJBA8.1 and XQ-2d, the inventors observed sequence similarity between tJBA8.1 and XQ-2d, with 21 nucleotides overlapping in their respective 45-bp and 42-bp random regions (
Given the discrepancies between tJBA8.1 and XQ-2d binding to Jurkat cells, the inventors also evaluated holo-Tf competition with these aptamers on H9 T-lymphoma cells, which have higher TfR1 expression than Jurkat cells and exhibit more equivalent max binding signal between tJBA8.1 and XQ-2d (
Besides holo-Tf, TfR1 has other natural ligands that may share binding epitopes with tJBA8.1. HFE is a major histocompatibility complex (MHC) class I-like membrane protein that is known to compete with holo-Tf at its TfR1-binding site to regulate iron uptake,44-47 and mutation of HFE can cause iron overload that resembles hereditary hemochromatosis via hepatic hepcidin deficiency.48,49 To ascertain whether tJBA8.1 and XQ-2d differentially bind TfR1 with respect to HFE, the same membrane protein pull-down assay was performed as before but also included Western blotting to examine HFE co-precipitation, if any, with aptamer-enriched TfR1. Bands with the characteristic sizes of TfR1 were highly enriched by both tJBA8.1 and XQ-2d (
tJBA8.1-Mediated Depletion of B-Lymphoma Cells from PBMCs
Because TfR1 is overexpressed in many cancer types including leukemias (both lymphocytic and myeloid),50,51 lymphomas,52,53 and myelomas,39 and the level of TfR1 expression marks the proliferative potential of these malignant cells,54 it was recognized that tJBA8.1 could be utilized as a malignant cell depletion agent in CAR T-cell manufacturing. The inventors therefore developed a MACS-based approach combining biotinylated tJBA8.1 and Anti-Biotin Microbeads (Miltenyi Biotec) for selective depletion of cancerous cells from PBMCs (
Elucidation of TfR1-Independent tJBA8.1 Binding and Affinity Optimization
The previous binding and competition results with tJBA8.1 on Jurkat cells support the hypothesis that tJBA8.1 has a TfR1-independent binding component. Given that tJBA8.1 captured some PBMCs in depletion studies, it was speculated that tJBA8.1 can bind a subpopulation of PBMCs via the same TfR1-independent mechanism. To test this and better understand the nucleotides that contribute to this interaction, the individual full-length aptamers identified from cell-SELEX in
To identify the cellular target that tJBA8.1 binds independently from TfR1, the inventors revisited the pull-down results in
Although CNBP lacks a transmembrane domain and its subcellular localization to-date has only been documented in the cytoplasm,57 it was hypothesized that it may be unconventionally expressed at the surface of certain cells, similar to the DNA/RNA-binding protein nucleolin that has been famously targeted by the G-quadruplex containing AS1411 aptamer.62,63 To test this, the inventors conducted extracellular flow cytometry staining of Jurkat cells, H9 cells, and PBMCs with a monoclonal anti-CNBP antibody (clone 38F10). While extracellular expression of CNBP was not detected on Jurkat and H9 cells with this antibody clone, extracellular CNBP expression was observed on a subset of PBMCs (
tJBA8.1 Affinity Optimization
As these efforts to remove TfR1-independent binding were unsuccessful, the inventors revisited the NGS results to identify alternative Motif 3 aptamers that could have improved affinity for TfR1, which would allow one to lower the concentration of aptamer needed for effective cancer cell depletion and thereby minimize TfR1-indpendent binding. Besides JBA8.1, there were three other aptamers belonging to Motif 3 in the top 50 sequences of the round 8 cell-SELEX pool, namely JBA8.10, JBA8.26, and JBA8.45 (Table 5). Notably, all three aptamers were single-point variants of JBA8.1 that displayed approximately 20-fold enrichment in round 8, which was significantly greater than the 6.9-fold round 8 enrichment for JBA8.1. JBA8.26 was selected for further characterization given that it had the highest fold enrichment of the three aptamers. Analyzing the predicted MFE secondary structure of JBA8.26 by NUPACK™, it was noticed that JBA8.26 has an extended stem region compared to JBA8.1, owing to a T→C mutation that induces complementary base pairing (
As expensive, living drugs that often serve as the final barrier between severely ill cancer patients and terminal disease, CAR T cells require stringent manufacturing to maximize their safety and efficacy. In rare cases, with T-cell isolation approaches currently employed in commercial therapies, malignant cells advance through the isolation process and contaminate enriched T cells, which can inadvertently exhaust produced CAR T cells or even create a cancer resistant to therapy.14, 15 Accordingly, an inexpensive approach that actively removes cancerous cells from patient PBMCs while leaving healthy T cells untouched would be of high value to CAR T-cell manufacturing and safety.
In this work and in the studies described herein and the inventors report the discovery of a nanomolar-affinity DNA aptamer, named tJBA8.1, that binds the iron importer TfR1 overexpressed by cancer cells but not expressed by healthy PBMCs. In proof-of-concept MACS-based depletion studies, tJBA8.1 was capable of efficiently removing Raji B-lymphoma cells spiked into PBMCs at various concentrations, yielding uncontaminated, label-free PBMCs. Importantly, the composition of healthy immune cells was unaltered by the depletion steps, showing the precise partitioning afforded by tJBA8.1. This work provides a straightforward, cost-effective approach for selectively removing malignant cells before CAR T-cell production, enabling safer and more reproducible manufacturing of this precious therapy.
Comprehensive validation and characterization of aptamer binding to their cognate receptors can contribute to their widespread recognition and use. For tJBA8.1, the inventors conducted many assays to both validate its binding to TfR1 and further understand its binding site on the receptor, especially within the context of other TfR1 ligands. Notably, tJBA8.1 binding to TfR1+ cells was competed off by both iron-bearing transferrin and another TfR1-binding aptamer described in the literature, XQ-2d,42, 43 indicating that tJBA8.1 shares overlapping binding epitopes on TfR1 with these ligands. Intriguingly, in pull-down assays using cell membrane proteins, tJBA8.1 failed to co-precipitate HFE, another natural TfR1 ligand that is co-expressed on the cell surface, while XQ-2d could. This establishes a key difference in TfR1 binding between tJBA8.1 and XQ-2d, indicating that their binding interfaces are spread over multiple regions on the receptor, some of which overlap between the two aptamers while others are unique. Indeed, molecular dynamics simulations predicted that XQ-2d binds to TfR1 at two regions,31 and only the nucleotides involved in binding to one region appear to overlap strongly with the tJBA8.1 sequence.
The inventors also demonstrated that tJBA8.1 does not bind mouse TfR1 despite the protein sharing ˜75% identity in its extracellular domain with human TfR1. Mouse TfR1 has mutations at multiple residues in a helices 1, 2, and 3 of the human TfR1 helical domain bound by holo-transferrin (
Besides TfR1, it is likely that tJBA8.1 has other target receptors on Jurkat cells and other cell types. Aptamer binding to Jurkat cells was only partially abrogated by large molar excesses of holo-transferrin and XQ-2d, and aptamer binding to TfR1− monocytes and B cells within PBMCs was also observed. Potentially explaining some of this TfR1-independent binding, an interaction between tJBA8.1 and the DNA/RNA-binding protein CNBP was characterized. As CNBP expression has previously been documented only in the cell cytoplasm, it is reported for the first time herein probable extracellular expression of this protein on CD3− PBMCs that colocalized and correlated with TfR1-independent binding of tJBA8.1. While the monoclonal anti-CNBP antibody that was tested failed to detect extracellular CNBP expression on Jurkat cells, enriched CNBP was found in pull-down assays using tJBA8.1 and Jurkat membrane extracts. It is known that the CNBP gene encodes as many as seven alternatively spliced isoforms,57 some of which may be selectively expressed on the cell surface of certain cells and lack epitopes for monoclonal antibody recognition. Alternatively, tJBA8.1 may be internalized by a two-part macropinocytosis mechanism that is CNBP-dependent, which has been similarly described for the nucleolin-binding AS1411 aptamer and would explain why tJBA8.1 does not greatly deplete TfR1− monocytes and B cells from PBMCs via MACS despite binding these subsets.70
The inventors believe that the TfR1-independent binding of tJBA8.1 is mediated by a motif of five consecutive guanines (i.e., G-quintet) which is shared by multiple aptamers identified from the cell-SLEX. This G-rich patterning may promote the formation of intramolecular or intermolecular G-quadruplexes,59 which are know to interact with DNA/RNA-binding proteins like CNBP.71 Initial attempts have been made to decouple this off-target binding of tJBA8.1 from its intended TfR1 binding by mutating the last three guanines in the G-quintet motif, but these changes to the tJBA8.1 sequence removed both TfR1-dependent and independent binding, suggesting that these nucleotides are important for TfR1 binding and/or tJBA8.1 structure. Despite these results, there are further strategies worth exploring, such as mutating the first two guanines of the G-quintet motif or neighboring nucleotides to make the G-quintet motif partially double stranded.
While B-lymphoma cells were used in depletion studies to mimic cancers currently treated by commercial CAR T-cell therapies, it is anticipated that the depletion strategy described herein will have a greater impact on the treatment of T-cell malignancies. Presently, autologous CAR T-cell manufacturing for treating T-cell leukemias and lymphomas is impractical since malignant T cells are often found in the peripheral blood of patients with these diseases and immunoaffinity purification approaches that target common T-cell antigens are unable to distinguish normal T cells from these malignant T cells.18, 19 To circumvent these challenges, CAR NK cells or allogeneic CAR T cells can be used for treatment,72, 73 but the former is difficult to manufacture at a clinical scale whereas the latter requires gene editing to prevent fatal graft-versus-host-disease and likely has limited persistence due to host-versus-graft effects. For these reasons, CAR T-cell manufacturing for T-cell malignancies would serve to benefit the most from the depletion strategy developed here, as it would allow selective harvesting of healthy autologous T cells that are otherwise unattainable. Furthermore, as T-lineage leukemias and lymphomas have been shown to have greater TfR1 expression and positivity than B-lineage counterparts,51, 53 these cancers should be especially amenable to tJBA8.1 recognition and capture.
It is specifically contemplated that the aptamer-based malignant cell depletion strategy described here could be adapted to affinity chromatography approaches, eliminating cell processing steps associated with MACS. Chemical conjugation of aptamers to chromatography solid supports will also realize a fully-synthetic, low-cost cell depletion system, unlike the anti-biotin coupling used here that relies on expensive antibodies. As contaminating myeloid cells also inhibit the production of CAR T cells,74 tJBA8.1 could be applied in combination with the monocyte-binding aptamer described above to elegantly deplete malignant cells and monocytes from patient PBMC concentrates in a single processing step.75 Besides cell isolation, other applications of tJBA8.1 within CAR T-cell manufacturing are possible. The inventors previously used cationic comb polymers and polymer-lytic peptide conjugates (VIPER) for non-viral gene delivery to T cells, and low uptake was one of the limiting barriers to activated T-cell transfection.76,77 Of relevance, the inventors show in this work that activated T cells have upregulated TfR1 expression and high tJBA8.1 binding, and TfR1 is well known to be rapidly internalized upon ligand binding via clathrin-mediated endocytosis.40 Comb polymer and VIPER formulations could thus be decorated with tJBA8.1 to enhance their binding to activated T cells via TfR1, improving their uptake for T-cell transfection. Outside of CAR T-cell therapy, TfR1 targeting with tJBA8.1 could have implications in the detection of circulating tumor cells and the delivery of drugs across the blood-brain barrier,78, 79 paving the way for tJBA8.1-based diagnostics and therapeutics.
Methods and Materials
Cell line and primary cell culture. The Jurkat, J.RT3-T3.5, H9, and Raji cell lines were purchased from ATCC and cultured in RPMI 1640 medium (Corning, Gibco) with 10% FBS (Life Tech). Primary healthy donor PBMCs were isolated from Leukocyte Reduction System (LRS) chambers (Bloodworks Northwest) by Ficoll-Paque™ density gradient centrifugation (GE). Positively enriched primary CD4+ and CD8+ T cells were gifted by Juno Therapeutics. For T-cell activation studies, T cells were thawed and stimulated 1:1 with Dynabeads™ Human T-Activator CD3/CD28 (Gibco) in RPMI 1640 10% FBS supplemented with 20 ng/mL recombinant human IL-2 (Miltenyi) for CD8+ T cells, 5 ng/mL recombinant human IL-7 (Miltenyi) for CD4+ T cells, and 0.5 ng/mL recombinant human IL-15 (Miltenyi) for both CD4+ and CD8+ T cells. Fresh media exchanges were conducted every 2-3 days to replenish cytokines, and T cells were passaged to larger plates or flasks upon reaching high densities (2-3×106 cells/mL) with visible yellow coloration of the culture media.
Oligonucleotides, PCR reagents, and recombinant proteins. Aptamer sequences used in this work are listed in Table 6. All oligonucleotides, including the randomized naïve library, primers, and individual aptamers, were synthesized by Integrated DNA Technologies with either HPLC purification (naïve library and individual aptamers) or standard desalting (primers). The FAM-labeled forward primer and biotin-labeled reverse primer used to amplify aptamer pools between rounds of cell-SELEX are as previously described.23 dNTPs and Phusion™ High-Fidelity Polymerase used in PCR reactions were purchased from QIAGEN and NEB, respectively. Recombinant human holo-Tf used in competition studies was purchased from Sigma-Aldrich. For BLI studies, the following recombinant proteins were used: biotinylated human TfR1 (TfR-H82E5, ACROBiosystems), His-tagged mouse TfR1 (50741-M07H, Sino Biological), and His-tagged human CNBP (NBP2-22742, Novus Biologicals). Recombinant human holo-Tf used in competition studies was purchased from Sigma-Aldrich.
Flow cytometry reagents and staining. For flow cytometry cell staining and competition studies, the following dyes and antibodies were used: ZOMBIE VIOLET™ (1:500, 107 cells/mL, BioLegend), CM-Dil (1:200, 106 cells/mL, Invitrogen), FITC anti-human CD71 (1:100-1:200, BioLegend, CY1G4), FITC anti-human CD14 (1:100, BioLegend, M5E2), FITC anti-human CD3 antibody (1:100, BioLegend, UCHT1), APC-Cy7 anti-human CD14 (1:200, Invitrogen, 61D3), Super Bright 702 anti-human CD19 (1:100, Invitrogen, SJ25C), Super Bright 600 anti-human CD56 (1:100, Invitrogen, TULY56), PE anti-human CD3 (1:100, BioLegend, HIT3a), APC-Cy7 anti-human CD3 (1:200, BD Biosciences, SP34-2), APC anti-human CD19 (1:200, BioLegend, HIB19), and Alexa Fluor™ 647 streptavidin (1:500, BioLegend), PE streptavidin (1:500, BioLegend), mouse anti-human CNBP (1:20, Novus Biologicals, 38F10), and FITC goat anti-mouse IgG (1:100, BioLegend, Poly4053). Antibody dilutions were adjusted when used as isotype/negative controls to match the concentration of experimental antibody used, or when doing serial dilutions for competition studies.
The base wash buffer used for aptamer folding, cell staining, and washing was comprised of DPBS with calcium and magnesium (Corning; ˜0.9 mM CaCl2 and ˜0.5 mM MgCl2) further supplemented with 5 mM MgCl2 (Fisher) and 25 mM D-glucose (Sigma-Aldrich). For antibody staining without aptamers, wash buffer supplemented with 1% BSA (Miltenyi) was used. For aptamer binding/staining with or without antibodies, wash buffer supplemented with 1% BSA and 0.1 mg/mL yeast tRNA (Invitrogen) was used. Wash buffer supplemented with 1% BSA was used for all wash steps. Aptamers were folded at 1 μM in wash buffer by heating the solution at 95° C. for 5 min followed by snap cooling on ice for at least 10 min before binding. For flow cytometry staining, cells were first removed from culture or thawed and washed with DPBS (Gibco) to remove residual media and serum. Cells were then incubated with live/dead ZOMBIE VIOLET™ dye diluted in DPBS for 15 min at room temperature. After live/dead staining, cells were washed with wash buffer 1% BSA to neutralize remaining ZOMBIE VIOLET™ and aliquoted into a polypropylene U-bottom 96-well plate (Corning) at 1-2×105 cells/well for immortalized cell lines and primary T cells and 2-5×105 cells/well for PBMCs. Cells were then blocked with 10 μL FcR Blocking Reagent (Miltenyi) for 5-10 min at 4° C. (only if they were to be stained with antibodies) and subsequently stained with 100 μL antibodies and/or aptamers for 20-30 min at 4° C. in the appropriate binding buffer at the indicated concentrations. For competition assays, unlabeled anti-CD3 antibody (BioLegend, UCHT1), unlabeled anti-CD71 antibody (BioLegend, CY1G4), recombinant human holo-Tf, or FAM-labeled XQ-2d were added to the primary incubation with aptamer at different concentrations. After staining, cells were washed twice with 200 μL wash buffer 1% BSA to remove excess antibodies and/or aptamers. If the aptamers or the antibodies used were biotinylated or were unlabeled, cells underwent a further incubation with 100 μL of fluorescently labeled secondary for 20-30 min at 4° C. in wash buffer 1% BSA and subsequently washed twice as before. Fully stained cells were lightly fixed in 200 μL wash buffer 1% BSA further supplemented with 0.1-0.2% PFA (Alfa Aesar) and immediately analyzed on an Attune™ NxT Cytometer (Invitrogen). If required, unstained and single-stained cell controls were used for compensation.
Cell-SELEX. The experimental design of cell-SELEX used in this study is summarized in
Next generation sequencing (NGS) and sequence analysis. The aptamer pools from each SELEX round were amplified using the barcoded primers listed in Table 4 and sequenced on the MiSeq™ System (Illumina) using a MiSeq™ Reagent kit v2 (300 cycles). FASTAptamer™ v1.0.3 toolkit was used to analyze the FASTA files from sequencing.29 Specifically, FASTAptamer-Count™ was first used to measure the reads for each unique sequence and rank the sequences by abundance (reads per million or RPM), and FASTAptamer-Enrich™ was then used to analyze the fold-enrichment of each unique sequence across adjacent rounds (e.g., RPM om Round Y divided by RPM om Round X; or later rounds over earlier rounds). MEME™ suite v5.3.3 Motif Discovery tool was used to find recurring motifs within the top 50 aptamers sequences in rounds 5-8 with the following settings: zero or one site per sequence for motif distribution, 0-order model of sequences for the background model, minimum motif width of 6 bases, maximum motif width of 45 bases, minimum sites per motif of 2, maximum sites per motif of 50, and search given strand only.31 Phylogenetic trees of the top 50 aptamer sequences in rounds 5-8 were generated using the FigTree™ v1.4.3 graphical viewer to map sequence similarities and differences.30 The NUPACK™ web application was used to predict the minimum free energy structure of aptamer sequences at 4° C. with 137 mM Na+ and 5.5 mM Mg++ concentrations.33
Subcellular localization of aptamer binding by trypsin flow and confocal imaging. The localization of tJBA8.1 binding on Jurkat cells at 4° C. was evaluated by both flow cytometry and confocal imaging. In the flow cytometry assay, Jurkat cells were first stained with Cy5-labeled tJBA8.1 as described above. After washing away unbound aptamer 50 μL of 0.25% trypsin or wash buffer with 1% BSA was then added to the cells for a 5-min incubation at 37° C. Immediately after incubation, 200 μL RPMI 1640 10% FBS was added to the cells (a 4-time trypsin volume) was added to the cells to neutralize trypsin enzymatic activity, and the cells were washed with 200 μL of wash buffer with 1% BSA before fixation in wash buffer 1% BSA further supplemented with 0.2% PFA. The change in Cy5-labeled tJBA8.1 binding on the Jurkat cell surface due to trypsin treatment was evaluated by flow cytometry.
For confocal imaging of tJBA8.1 subcellular compartment binding at 4° C., 15-mm round glass cover slips (Bellco Glass) were first sanitized in 95% ethanol for 5 min and subsequently dried over a 24-well plate for 15 min. Cover slips were next coated with 150 μL of 10 μg/mL poly-D-lysine (Sigma) for 2 hr at 37° C. in a 24-well plate. Meanwhile, 5×106 Jurkat cells were washed twice with wash buffer 1% BSA and stained with 300 μL of 200 nM Cy5-labeled RANL or tJBA8.1 aptamer in binding buffer for 30 min at 4° C. After aptamer binding, excess aptamer was removed by washing the cells twice with 200 μL of wash buffer 1% BSA, and the cells were resuspended in 200 μL wash buffer 1% BSA fir cover slip plating. For this, poly-D-lysine-coated-cover slips were washed once with 1 mL water in a 24-well plate to remove excess poly-D-lysine before adding cells. Jurkat cells were adhered to the coated cover slips by centrifugation at 650×g for 10 min at 4° C. After cell attachment, the excess buffer was aspirated, and adhered cells were fixed with 200 μL of 0.4% PFA for 10 min at room temperature. Fixative was washed off the cell-coated cover slips three times with 1 ml DPBS for 1 min each, and fixed cells were then permeabilized with 200 μL of 0.1% Triton X-100 (Sigma) for 10 min at room temperature. After washing as before FITC Phalloidin (1:100, Invitrogen) and DAPI (300 nM, Thermo fisher) in 200 μL DPBS were applied onto the fixed and permeabilized cells for 30 min at room temperature. Stained cells were then washed as before except in longer 5-min increments, and the stained cell-coated glass cover slips were mounted onto larger microscope slides (Fisher) with 10 μL polyvinyl acetate. Fluorescent images were taken at 60× magnification on a Leica SP8× confocal microscope and imaging processing, and overlaying was performed using Fiji/ImageJ.82
Target receptor pull-down and identification. About 85 million Jurkat cells (aptamer target cells) were used for each group. The membrane protein extraction and target purification process were modified from a previously published method.34 Jurkat cell were washed three times in DPBS to remove media-derived proteins and then lysed foe 30 min at 4° C. with end-over-end mixing in ˜3.3 mL hypotonic buffer comprised of 10 mM Tris-HCl (pH 7.5) supplemented EDTA-free cOmplete™ Protease Inhibitor Cocktail (Roche) and 1 mM phenylmethylsulfonyl fluoride. The resulting cell membrane debris were pelleted at 16,000×g for 15 min at 4° C. and washed 3 times with 3.3 ml of the same hypotonic buffer to remove released intracellular proteins. The membrane pellets were then each resuspended in 1 mL of wash buffer containing 1% Triton X-100, and same concentration of protease inhibitors and 1 mM PMSF to extract and solubilize membrane proteins. The extraction took place at 4° C. with end-for-end mixing for 30 minutes followed by a brief 5 min sonication in an ice water bath. Afterward, the samples were centrifuged as before and the supernatant containing the extracted proteins was collected and stored at −80° C. until next step or used immediately.
Once processed, the extract was first “pre-cleared.” The extracts (1 mL) were thawed and spiled with 100 nM biotin RANL and 0.1 mg/mL yeast tRNA as anionic blocker. After incubating for 30 min at 4° C. with end-to-end mixing, 200 μL (2 mg) of MyOne™ Streptavidin C1 Dynabeads™ (Thermo Fisher) were added to the extracts and incubated for another 15 min at 4° C. to magnetically remove RANL-bound proteins. For the tJBA8.1 group, the pre-cleared extract was spiked with 0.1 mg/mL salmon sperm DNA (Invitrogen) as an additional anionic blocker and 100 nM biotinylated tJBA8.1. After incubating for 30 min at 4° C. with end-over-end mixing, 150 μL (1.5 mg) Streptavidin Dynabeads™ were added to the binding reaction and incubated for another 15 min at 4° C. to capture tJBA8.1-bound proteins. For the control group, 150 μL (1.5 mg) Streptavidin Dynabeads™ were instead saturated with 50 nmol biotin and added to the pre-cleared extract for a 30-min incubation at 4° C. Afterwards, the beads for both groups were washed 5 times for 5 min each with 1 mL cold wash buffer with 0.01% Triton X-100 to remove unbound proteins. To minimize the stripping off streptavidin from the beads, the captured membrane proteins were mildly eluted by heating the beads for 15 min at 47° C. for 15 minutes in loading buffer composed of 1× Laemmli sample buffer (containing SDS) supplemented with 4.6 M urea, 2.5% 2-mercaptoethanol, 10 mM EDTA, and 0.1% Triton X-100. Eluted proteins were stored at −80° C. unless used immediately.
After removing the Streptavidin Dynabeads™ from the supernatant, the eluted proteins (25 μL) were loaded onto a Novex™ Wedge 8% Tris-Glycine gel (Invitrogen) and separated by electrophoresis. The gel was stained with Colloidal Blue Staining Kit (Invitrogen) and imaged on a Gel Doc EZ system. Enriched bands were excised, and the excised band samples were digested, desalted, and subjected to tandem mass spectrometry (Orbitrap Elite). The data was analyzed with Proteome Discoverer™ 2.2 against Uniprot Human database.
siRNA knockdown study. Some 106 Jurkat cells in logarithmic growth phase were nucleofected with a Human T cell Nucleofector™ Kit (Lonza) with program settings X-001 following the manufacturer instruction. 30 pmol of TFRC1 (Table 7) and NS siRNA (non-specific scrambled control) were used. After nucleofection, the cells were incubated for 22 hours and stained with Cy5 tJBA8.1 and FITC antihuman CD71 antibody. The aptamer staining and CD71 expression was evaluated with flow cytometry.
BLI. BLI studies were performed on an Octet RED96™ instrument (Sartorius) using either streptavidin or Ni-NTA biosensors (Sartorius). The sample buffer used for equilibration, loading, rinse, baseline, association, and dissociation steps was comprised of wash buffer supplemented with 1% BSA, 0.1 mg/mL yeast tRNA, 0.1 mg/mL salmon sperm DNA, and 0.01% Tween-20. All steps were performed at 25° C. with sample agitation at 1000 r.p.m. Biosensors were allowed to equilibrate in buffer for at least 10 min before loading. Loading thresholds were optimized for each capture ligand and analyte used. For streptavidin biosensors, 25 nM recombinant biotinylated human TfR1 was loaded until a 3.5 nm threshold and 50 nM biotinylated aptamers were loaded until a 0.5 nm threshold before performing rinse and baseline steps in buffer alone for 100 s each. For Ni-NTA biosensors, 150 nM recombinant His-tagged mouse TfR1 was loaded until a 0.7 nm threshold and 300 nM recombinant His-tagged human CNBP was loaded until a 3 nm threshold before performing cross-linking with 0.1 M EDC+0.025 M NHS in water for 60 s to stabilize the capture protein. The cross-linking reaction was quenched with 1 M ethanolamine pH 8.5 for 60 s before performing rinse and baseline steps as before. Afterwards, loaded sensors were associated with appropriate analytes at concentrations and times indicated in the figures and figure legends. Lastly, sensors were transferred to wells containing buffer alone for dissociation. Data analysis was done on the Octet™ Data Analysis 9.0 software (Sartorius). Association and dissociation curves were normalized to sensors that received capture ligand alone, and kinetic constants were calculated for datasets with several analyte concentrations by conducting a global fit of the association and dissociation curves to a 1:1 or 2:1 heterogeneous ligand binding model. The quality of the fit was evaluated using R2 and χ2 values which are listed in Table 8 for the applicable datasets along with the calculated kinetic constants.
Aptamer, recombinant protein, and antibody competition studies. Jurkat or H9 cells were incubated with Cy5- or FAM-labeled aptamers at 25 nM with or without various fold-increased concentrations of competitor aptamers, FITC anti-human CD71 antibody or recombinant holo-transferrin proteins for 30 minutes at 4° C. Cells were washed 2 times with wash buffer with 1% BSA and fixed in wash buffer with 1% BSA and 0.2% PFA. Changes in aptamer binding affected by the competitors was evaluated on flow cytometer.
Western blotting. Western blotting was used to identify human TfR1 and HFE protein in the aptamer purified protein mixture. Aptamer-bound membrane proteins were first purified using the pull-down process described above with 100 nM biotinylated tJBA8.1 and XQ-2d. Eluted proteins (5 μL) were loaded on Novex™ WedgeWell™ 4-20% Tris-Glycine gel (Invitrogen) and separated by SDS-PAGE. Separated proteins were then blotted on a PVDF membrane (Bio-Rad) under 50 V for 3 hours on ice. Blotted membranes were subsequently blocked in TBST buffer (20 mM Tris, 150 mM NaCl, 0.1% Tween-20 at pH 7.5) supplemented with 3% BSA at room temperature for 1 hour. After blocking, membranes were washed twice with TBST for 5 min each before cutting into separate matched lanes for the different stains. Separated membranes were then stained with rabbit anti-human TfR1 (1:1000, Cell signaling, D7G9X) or rabbit anti-human HFE (1:1000, Abcam, EPR6750) diluted in TBST supplemented with 1% BSA overnight at 4° C. with gentle shaking. After primary incubation, the membranes were washed for 5 minutes 3 times with TBST and stained with secondary antibody, HRP-conjugated anti-rabbit IgG (1:5000, Sigma-Aldrich, A6154) diluted with TBST supplemented with 1% BSA for 1 hr at room temperature with gentle shaking. After secondary incubation, the membranes were then washed 3 times as before with TBST and imaged using SuperSignal™ West Pico PLUS substrate (Thermo Fisher) on a Xenogen™ IVIS (PerkinElmer) imager with small binning, aperture of f/2, and 10-60 s of exposure.
Raji cell depletion from PBMCs. Raji cells (5×106) were first pre-stained with Vybrant™ CM-Dil dye (1:200 or 5 μM, Invitrogen) in serum-free RPMI 1640 medium at 106 cells/mL for 30 min at room temperature. After washing the stained cells twice with wash buffer, approximately 3×104, 3×105, and 3×106 CM-Dil stained Raji cells were spiked into three aliquots of 30×106 freeze-thawed PBMCs to achieve 0.1%, 1%, and 10% spiked cell percentages. The mixed PBMC-Raji cells were then each stained with 500 μL 80 nM biotinylated tJBA8.1 in wash buffer supplemented with 0.5% BSA and 0.1 mg/mL yeast tRNA for 30 min at 4° C. Meanwhile, three aliquots of 150 μL Anti-Biotin MicroBeads (Miltenyi) were blocked in 500 μL of the same buffer for 30 min at 4° C. to prevent non-specific interactions. After staining, each group of aptamer-labeled mixed cells was washed once with wash buffer 0.5% BSA 0.1 mg/mL yeast tRNA and resuspended in 650 μL of the blocked microbead-containing solution for a 15-min incubation at 4° C. After attaching the microbeads to the aptamer-labeled cells, each group of fully labeled mixed cells was washed once as before and resuspended in 1 mL wash buffer 0.5% BSA 0.1 mg/mL yeast tRNA.
Prior to Raji cell depletion, three LS Columns (Miltenyi) were anchored onto a separation magnet (QuadroMACS Separator, Miltenyi) and each was washed once with 3 mL cold wash buffer with 0.5% BSA and 0.1 mg/mL yeast tRNA. Each column was equipped with a 30 μm Pre-Separation Filter (Miltenyi) before applying the full labeled mixed cells. The Raji cell-depleted PBMCs in the flow through were collected, and each filter-equipped column was washed three times with 3 mL cold wash buffer and 0.5% BSA into the same flow through collection tube. The captured Raji cells were collected by removing the columns from the magnet and flushing each with 5 mL autoMACS Rinsing Solution (Miltenyi, 2 mM EDTA) further supplemented with 3 mM EDTA (Invitrogen) and 0.5% BSA using a plunger. The cell composition in each fraction and corresponding depletion efficiencies were determined by antibody staining and flow cytometry.
Protein sequence alignment of TfR1 with mouse ortholog and hepatic homolog. Protein sequence alignment of human TfR1 (UniProt entry P02786; SEQ ID NO: 144) with mouse TfR1 (UniProt entry Q62351; SEQ ID NO: 146) and human TfR2 (UniProt entry Q9UP52; SEQ ID NO: 145) was performed with Clustal Omega v1.2.4.83,84. The percent identity between the extracellular domain of human TfR1 (residues 89-760) with each of the extracellular domains of mouse TfR1 (residues 89-763) and human TfR2 (residues 105-801) was calculated in the same program.
Statistical analysis. Data are expressed as mean±standard deviation (s.d.), unless otherwise stated, and the number of biological and technical replicates is specified in the figure legends. A two-tailed t-test was used for hypothesis testing when comparing only two populations. ANOVA was used for hypothesis testing comparing more than two populations to each other. Paired hypothesis testing was sometimes performed to account for large donor-to-donor, or experimental (e.g., nucleofection) variability. To correct for multiple comparisons, Tukey's test or Dunnett's test was used to adjust the P values when every mean was compared to every other mean or a control mean, respectively, whereas Šídík correction was used to adjust P values when select sets of means were compared, assuming independence. If comparisons could not be assumed independent from each other Bonferroni correction were used instead of Šídík correction to adjust the P values. Tested differences were considered significant if P<0.05. Unless otherwise stated in the methods, graphing and statistical tests were performed using GraphPad™ Prism v6.01 (GraphPad Software Inc.) for Windows
Summary: Cell isolation systems that purify cells without masking the receptors with labeling materials are beneficial for both scientific studies and pharmaceutical applications. While methods that tracelessly isolate one cell type at a time have been reported using engineered antigen binding fragments and aptamers, a multiplexed cell isolation system with removable labeling reagent is still needed. To address this, two aptamer sequences were used along with their designated complementary strands (antidotes) to tracelessly isolate two target cell types from a single specimen. This technology is useful in circumstances where the source of cells is limited for repeated positive selection of multiple cell types, such as CAR T cell manufacturing that requires both CD4+ and CD8+ T cells from patients that have undergone one or more cytotoxic chemotherapies. In this study, the inventors tested the feasibility of multiplexed traceless cell isolation with JBA8.1 and A3t aptamers. JBA8.1 is a previously developed CD71 protein binder that can target activated T cells and some cancer cell lines. A3t is a previously reported CD8 protein binder used for traceless isolation of CD8 T cells from peripheral blood mononuclear cells (PBMCs). The inventors modified JBA8.1 (into JBA1.2) and developed an antidote that is orthogonal to A3t strand displacement. The JBA1.2 and its antidote were first incorporated into a cell isolation system to separate activated T cells based on their CD71 expression levels. Moving forward, JBA1.2 was tested in combination of A3t to isolate activated CD4+ T cells and resting CD8+ T cells. Orthogonal strand displacements were used to elute these cells into separate fractions after one staining procedure.
Positive Cell Isolation Technology with Removable Targeting Ligand
Isolation systems that purify cells without masking their receptors with purification agents are necessary in biomedical fields such as cell signaling pathways research and are desirable for clinical adoptive cell therapies. Reported traceless cell isolation methods using engineered antigen binding fragments or aptamers have been limited to processing a single cell type at a time. There remains an unmet need for cell isolation processes that rapidly sort for multiple target cell types. Here, two aptamers were utilized along with their designated complementary strands (reversal agents) to tracelessly isolate two cell types from a mixed cell population with one aptamer labeling step and two sequential cell elution steps with reversal agents. A CD71-binding aptamer (rvCD71apt) was engineered and reversal agent pair to be used simultaneously with our previously-reported traceless purification approach using CD8 aptamer (rvCD8apt) and its reversal agent. The compatibility of the two aptamer displacement mechanisms was verified by flow cytometry, and the feasibility of incorporating rvCD71apt with a magnetic solid state. rvCD71apt was then combined with rvCD8apt to respectively isolate activated CD4+ T cells and resting CD8+ cells by eluting these target cells into separate fractions with orthogonal strand displacements. This is the first demonstration of isolating different cell types using two aptamers and reversal agents at the same time. Potentially, different or more aptamers can be included into this traceless multiplexed isolation system for diverse applications with shortened operation time and lower production cost.
Current Development of Removable Targeting Ligands
Immunoaffinity-based cell isolation methods are effective ways to purify specific cell types and include fluorescent-activated cell sorting (FACS)1, magnetic cell separation2, and cell affinity chromatography3,4. In contrast to separation methods based on physical differences (e.g., size, density) between cells that often result in lower isolation resolution5,6, immunoaffinity-based methods rely on labeling cell surface proteins with antibodies or antibody fragments to differentiate between cell populations. The labeled cells are then separated from non-labeled cells using solid supports such as magnetic beads or agarose resins. In positive selection strategies, the desired cells are directly labeled for isolation, whereas in negative selection strategies, the undesired cells are labeled for removal from the desired cells. Positive selections usually result in high purity of target cells7,8 but with certain surface proteins still bound by the ligands and solid supports. Negative selections rely on a panel of antibodies and result in residue-free target cell products but often with reduced purity and yield dependent on sample variability9,10. Since the antibodies and solid supports used for isolation can potentially induce unwanted immunogenicity11, block cell signaling pathways12, and impact cell proliferation13, cells isolated for cell therapy applications such as stem cell transplantation14 and adoptive T cell therapy15,16 are ideally free of cell labeling materials. Thus, there has been a focus in the art on developing positive isolation strategies that employ removable selection agents17.
Fab-Streptamer® is a “quasi-traceless” protein-based isolation system using modified Fabs (antigen binding fragments) that have lower binding affinity to targets in monovalent form and higher affinity as multivalent complexes. Fabs fused with Strep-Tag peptides18,19 will form high affinity tetramers in the presence of Strep-Tactin20, a mutated streptavidin, through non-covalent interaction. The interaction between Fab-Strep-Tag® and Strep-Tactin® is reversable upon competition with high affinity D-biotin for Strep-Tactin® binding. Upon release from Strep-Tactin®, the monovalent Fab has reduced affinity for target and will then separate from the target spontaneously. This technology has been used to purify proteins21, as well as to isolate cells in clinical adoptive cell therapy manufacturing4, 22-24. However, D-biotin is a universal target releasing agent in this system. Isolating multiple cell types with Fab-Streptamer® is therefore a time-consuming process that requires repeated staining and dissociating steps for different targets.
Gao and coworkers developed a DNA nanotechnology method for reversible antibody labeling and sorting of targets by functionalizing antibodies with DNA oligonucleotides consisting of a toehold for on-demand displacement and a targeting probe for anchoring onto the solid state25. Multiple targets can be captured using an antibody cocktail with subsets subsequently recovered by sequence-dependent strand displacement initiated at the toehold domain to detach antibodies from solid supports. This elegant approach has been applied to isolate multiple immune cell types from mouse splenocytes with high purity and allows fast sequential isolation of multiple cell types with a single staining step26, but the end product still bound by antibodies. In addition, antibodies are a relatively costly and thermosensitive reagent.
Aptamers are short, folded single-stranded DNA or RNA oligonucleotides that bind to a wide range of targets including cell surface protein receptors. Typically selected from in vitro library screening, aptamers have comparable to antibodies affinity and specificity, but have the benefits of reversible binding, lower production cost, faster and more consistent manufacturing process 27-29. Aptamer binding can be reversed either by universal reagents, such as DNases, ion chelators, and competitors,30-32 or by complementary nucleic acid strands, also called reversal agents. When the reversal agent anneals to portions of the aptamer, part of the folded aptamer may be displaced, losing its structure essential for binding. This unique characteristic of aptamers has been utilized in various applications. For example, the Ch-9.3t RNA aptamer which serves as an anticoagulant by targeting factor IXa, can be neutralized using a chemically modified RNA sequence complementary to its loop structure33. Another example is E07, an EGFR targeting RNA aptamer that was incorporated into FACS and MACS (magnetic-activated cell sorting) systems to isolate EGFR+ epidermoid carcinoma cells. These EGFR+ target cells were incubated with E07 reversal agent for aptamer removal, and were suitable for downstream studies on EGFR-related pathways34. One can also design highly effective reversal agents by implementing an overhang sequence, also referred to as a toehold, on either the 5′- or 3′-end of the aptamer. With the assistance of toehold-initiated annealing, the strand displacement rate constant can be significantly increased35,36. For example, a CD8 receptor-binding aptamer, A3t, has been previously developed with an 8 nucleotides (nt) toehold for isolating CD8+ T cells from PBMCs. With 20 minutes of incubation using reversal agent at room temperature, over 90% of the DNA aptamer is removed from cells. The isolated CD8+ T cells are free of labeling materials and are ideal for manufacturing adoptive cell therapeutic products16.
Presented herein is a demonstration of the isolation of multiple cell types from a single selection process by using multiple aptamers paired with their reversal agents. To achieve this, different DNA toeholds were introduced into two targeting aptamers so that two cell types can be captured together upon aptamers binding and then sequentially eluted with reversal agents. Aptamers tolerate sequence modifications at the regions not essential for receptor binding. First, an existing CD71-binding aptamer, JBA8.137, was modified to carry a suitable toehold and a displaceable stem structure for reversal agent annealing and branch migration. The new variant was named as rvCD71apt (reversible CD71 aptamer). The corresponding reversal agent was optimized to achieve robust strand displacement. When immobilized on magnetic beads, the rvCD71apt/reversal agent pair separate bulk-stimulated PBMCs into different selection fractions based on their CD71 expression level. Next, rvCD71apt was combined with a previously reported CD8 aptamer used for cell isolation, A3t (renamed as rvCD8apt, reversible CD8 aptamer)16, as a traceless multiplex cell sorting system for purifying activated CD4+ T cells and resting CD8+ cells. In the future, this isolation system can be expanded to incorporate different and/or more aptamers and reversal agent pairs and applied to various types of solid supports for diverse applications.
Cell Preparation
Jurkat cell (human T-ALL) culture was maintained in RPMI 1640 media (Corning) supplemented with 10% FBS (Life Tech). Primary human PBMCs were Ficoll™-enriched apheresis product purchased from Bloodworks Northwest. CD4+ T cell and CD8+ T cell subtypes were isolated from PBMCs using negative cell isolation kits (Miltenyi) to obtain cells without antibody and magnetic bead residue. T cells were stimulated with recombinant cytokines (Miltenyi) and CD3/CD28 stimulation Dynabeads™ (Invitrogen) on day 0. CD4+ T cell culture media consisted of RPMI 1640 media with 10% FBS, recombinant human (rh) IL-7 (5 ng/mL) and IL-15 (0.5 ng/mL). CD8+ T cell culture media consisted of RPMI 1640 with 10% FBS, and rhIL-2 (20 ng/mL) and rhIL-15 (0.5 ng/mL). Unless stated otherwise, the stimulation beads were removed by pipetting cells with p1000 tips and separated on a magnetic rack on day 9. The cells were kept in the complete culture media containing cytokines for expansion until day 21. For bulk-stimulated PBMCs, the T cell fraction in PBMCs was used to calculate the amount of stimulation beads and cytokine needed.
Flow Cytometry and Staining Reagents
The following live/dead staining dye and antibodies were used for staining cells: Zombie Violet (1:500 in 100 μL containing 106 cells, BioLegend), FITC-antihuman CD71 (1:100, BioLegend, CY1G4), Super Bright 702-antihuman CD56 (1:100, Invitrogen, TULY56), APC-Cy7-antihuman CD14 (1:200, Molecular probes, 6ID3), Super Bright 600-antihuman CD19 (1:100, Invitrogen, SJ25C1), PE-antihuman CD3 (1:100, BioLegend, HIT3a), APC-antihuman CD8a (1:100, BioLegend, RPA-T8), FITC-antihuman CD8a (1:100, BioLegend, RPA-T8), APC-Cy7-antihuman CD4 (1:100, BioLegend, RPA-TA), FITC-streptavidin (1:500, BioLegend), Alexa Fluor 647-streptavidin (1:500, BioLegend), and PE-streptavidin (1:500, BioLegend).
In the aptamer staining process, wash buffer and binding buffer were used for washing and staining. Wash buffer, which contains 137 mM Na+ and 5.5 mM Mg2+, was made with 500 mL DPBS (with calcium and magnesium, Corning) plus 2.5 mL 1 M MgCl2 (Fisher) and 2.25 g D-glucose (Sigma-Aldrich). Binding buffer was made of wash buffer supplemented with 0.1 mg/mL yeast tRNA (Invitrogen) and 1% BSA (Miltenyi), unless stated otherwise. Aptamers were labeled with either FAM (6-carboxyfluorescein) fluorophore or biotin. To anneal aptamers, the DNA was adjusted to 1 μM in wash buffer and heated at 95° C. for 5 min followed by snap chilling on ice for at least 15 min. The cell staining process was kept at 4° C. to avoid aptamer internalization. RAN aptamer, a sequence that does not bind to any target cell types in this study, was used as control. The cell-associated fluorescence from aptamer binding was analyzed with an Attune NxT cytometer (Invitrogen).
Oligonucleotide Design
Customized oligonucleotides, including aptamers and antidotes were ordered from Integrated DNA Technology (IDT). Modifications of JBA8.1 was done partially with NUPACK™ web application.16 The settings for annealed aptamer structures were 137 mM Na+ and 5.5 mM Mg2+, and temperature at 4° C. The settings for antidote annealing were the same cation concentrations, with temperature at 25° C. The application simulates aptamer and antidote-annealed aptamer structures when sequences folded into their thermally stable structures.
Aptamer Displacement with Reversal Agents
The cells were first stained with targeting aptamers, and then washed with wash buffer to remove unbound atamers. Based on previously optimized aptamer displacement conditions,16 the inventors used 100× concentration of agents (e.g., applying 100 nM reversal agent to cells stained with 1 nM aptamer) for incubations at room temperature for 10-20 min.16 The reversal agents were first heated at 37° C. in water bath for 5 min, and then applied to the aptamer-stained cells. After washing off the displaced DNA strands, the remaining aptamer binding was evaluated by flow cytometry.
rvCD7apt Cell Sorting with MACS
Bulk-stimulated PBMCs were used in the rvCD71apt MACS (Miltenyi) cell sorting studies. Cells were activated using the method described in section above on day 0. The CD3/CD28 stimulation beads were removed on day 3 by a magnetic rack. 50 million stimulated PBMCs were first applied to an LS column (Miltenyi) anchored on a magnetic separator (Miltenyi) to remove cells with residual stimulation beads. The resulting cells were stained with 1 mL 40 nM biotinylated aptamer in wash buffer with 0.5% BSA and 0.1 mg/mL yeast tRNA for 30 minutes at 4° C. After staining, the cells were washed and labeled with 150 μL anti-biotin Microbeads (Miltenyi) in the same buffer with 15 min incubation at 4° C. 30 μL of anti-biotin Microbead stock solution was used for every 10 million total cells. The labeled cells were spun down and resuspended in buffer to remove excess beads. In the meantime, an LS column was rinsed with wash buffer containing 0.5% BSA and 0.1 mg/mL tRNA to reduce non-specific interactions. After the labeled cells were applied to the column for isolation, the column was washed 3 times with wash buffer containing 0.5% BSA. 1 mL of 4 μM pre-warmed reversal agent (100×) in wash buffer with 0.5% BSA was then applied to the column. An initial 600 μL of reversal agent solution was passed through the column and the remaining volume was trapped in the column with a Luer lock plug. The reversal agent solution was incubated in the column for 10 minutes at room temperature, after which the plug was removed, and the column was washed 3 times for cell collection. The remaining cells that were not eluted by reversal agent displacement, were collected by taking the LS column off the magnetic separator and flushed with wash buffer with a plunger. The CD71 expression and cell type of the isolated cells in each fraction were evaluated by antibody staining and flow cytometry.
Removing Un-Eluted Cells with Dextran Sulfate
Activated CD4+ T cells were isolated and cultured using the method described in section 2.1. Resting CD8+ T cells were isolated using the method described section 2.1. Both cell types were first stained with Zombie Violet™ live/dead staining dye, followed by 60 nM biotinylated rvCD71apt or 5 nM biotinylated rvCD8apt labeling (0.2 million cells in 100 μL 5 binding buffer) for 20 min at 4° C. The surface bound biotinylated aptamer was then labeled with secondary staining, streptavidin-AF647, at 4° C. for 15 min. The cells were washed and incubated in dextran sulfate (6.5-10 kDa, Sigma Aldrich) dissolved in wash buffer with 0.5% BSA at 0, 0.16, 0.31, 0.63, 1.25, 2.5, 5, 10, and 12 mM at room temperature for 20 min, with or without 6 μM rvCD71apt reversal agent or 0.5 μM rvCD8apt reversal agent. The cells were washed, fixed with wash buffer with 1% BSA and 0.1% PFA (paraformaldehyde), and then analyzed by flow cytometry. The AF647 fluorescence associated with cells due to aptamer binding was normalized to the control group without dextran sulfate and the reversal agents.
Traceless Multiflexed Cell Isolation with Serial Reversal Agent Elution
About 45-50 million pre-sort cells consisting of activated CD4+ T cells and PBMCs were washed with DPBS and 0.5% BSA and resuspended in 900 μL binding buffer. In this mixed cell population, the CD4+CD71+ cells (activated CD4+ T cells) were rvCD71apt target cells, while the CD8+ cells (mostly resting CD8+ T cells) were rvCD8apt target cells. This mixed population was pre-cleared using 1 mg MyOne™ Streptavidin C1 Dynabeads™ (Invitrogen) to remove non-specific binding cells by incubation at room temperature for 20 min, followed by bead removal using a magnetic rack (MagRack™ 6, GE). The cleared cells were set on ice during beads preparation. To prepare the isolation beads, 0.64 mg Dynabeads™ were washed and resuspended in 640 μL wash buffer with 0.5% BSA and 0.1 mg/mL yeast tRNA. The beads were incubated in the buffer for at least 20 min at 4° C. to reduce non-specific interaction. Dynabeads™ (0.6 mg) were transferred into 500 μL of 60 nM biotinylated rvCD71apt in wash buffer with 0.5% BSA and 0.1 mg/mL yeast tRNA. The remaining 0.04 mg Dynabeads™ were transferred into 500 μL of 5 nM biotinylated rvCD8apt in the same buffer. These streptavidin beads were incubated with the biotinylated aptamers for 15 min at 4° C. for aptamer immobilization to take place. The aptamer-labeled beads were washed and resuspended in 50 μL wash buffer with 0.5% BSA and 0.1 mg/mL yeast tRNA.
To isolate the target cells, the aptamer-beads were incubated with the cleared cells for 30 min at 4° C. and separated on the magnetic rack. The cell-aptamer-bead complex was applied to the magnetic rack to isolate the bead-labeled target cells. The unbound cells were removed by gentle washing. The rvCD71apt reversal agent (1 mL of 6 μM) in wash buffer with 0.5% BSA was pre-heated at 37° C., and then applied to the cell-aptamer-bead complex. The solution was incubated at room temperature for 20 min on rotation to displace rvCD71apt. The beads were then thoroughly washed to collect the activated CD4+ T cells. Next, 10 mM dextran sulfate was applied for 20 min at room temperature to wash off the un-eluted activated CD4+ T cells. After rinsing dextran sulfate off the cells with wash buffer 0.5% BSA, the solution of 0.5 μM rvCD8apt reversal agent in 1 mL wash buffer containing 0.5% BSA was applied for 15 minutes at room temperature and then collected through washing. The remaining cells were collected along with the magnetic beads. All incubations were done in DNA lo-bind tubes (Fisher) on rotation. The washing steps implemented between each incubation procedure were done by using wash buffer and 0.5% BSA. The cell compositions collected in each step were evaluated by flow cytometry after staining with human CD71, CD4, and CD8 antibodies.
The purity of target cells collected in the reversal agent elution steps is the percentage of cells within the population gates. rvCD71apt target cells were gated on CD4+CD71+ cells. rvCD8apt target cells were gated on CD8+ cells. The yield of the target cells is defined as the target cell number counted in the reversal agent elution steps divided by total target cell number counted in the pre-clearing, reversal agent elution, dextran sulfate wash steps, as well as the number of cells that were not captured by the aptamers and were not eluted by the reversal agents.
Statistical Analysis
Data are shown as mean±s.d of 3 biological replicates unless stated otherwise. Two-tailed unpaired t test was used to compare two populations. If there were more than two populations, one-way ANOVA was used to test the hypothesis. Tukey's test was used for multiple comparison in which every mean was compared to every other means. If P<0.05 after adjustments, the difference is considered significant. Graphing and statistical analysis was performed on GraphPad Prism 6 for Windows, version 6.01.
Modification of CD71-Binding Aptamer to Achieve Multiplexed Sorting
In order to isolate two cell populations from a single aptamer-based selection procedure, two aptamers were needed that could specifically disrupted by their reversal agent sequences. Two aptamers were selected: JBA8.1, a CD71 (also called transferrin receptor 1)-binding aptamer37, and A3t, a CD8 receptor-binding aptamer16. To elute target cells from these individual aptamers, complementary strands (reversal agents) were required that specifically reverse the folded structure of corresponding aptamer sequences. JBA8.1, however, shares overlapped sequences in its stem structure with A3t, renamed to rvCD8apt (reversible CD8 aptamer) for this work (
Optimization of Reversal Agent and Toehold Sequence for rvCD71apt
Next, the rvCD71apt system was optimized for reversibility by testing different reversal agents and different G-C content in the aptamer toehold sequences. Reversal agents with various lengths (21, 28, 33, and 51 nt) were designed for branch migration from the toehold placed on the 3′-end to various sites on rvCD71apt (
Next the impact of G-C content in the aptamer toehold sequence on strand displacement efficacy was explored. Toeholds with higher G-C content favor faster annealing rate of complementary strands due to their lower binding free energy35. Three rvCD71apt sequences were designed containing 8 nt toeholds with different G-C content (
Compatibility of rvCD71apt and rvCD8apt Aptamer/Reversal Agent Pairs
The compatibility between rvCD71apt, rvCD8apt, and their corresponding reversal agent sequences is crucial for isolating target cells at high purity with sequential elution. Here, sequence-specific displacement by the two reversal agents using flow cytometry was evaluated. It had been previously demonstrated that rvCD8apt with its reversal agent can be used for traceless isolation of CD8+ T cells16. rvCD71apt targets CD71 receptor, which is expressed by proliferating, activated T cells but not by resting T cells42. rvCD71apt binds with high affinity to both activated CD4+ and CD8+ T cells, with apparent KD around 28.5 nM and 35 nM, respectively (
rvCD71apt and Reversal Agent Separate PBMCs into Cell Populations Base on CD71 Expression Level
Biotinylated rvCD71apt and its reversal agent were incorporated into the MACS® system (Miltenyi) for cell isolation, which utilizes anti-biotin superparamagnetic particles to anchor ligand-labeled cells in a column with application of magnetic field. MACS with aptamer labeling for isolating CD8+ T cells at high purity have been used previously16. Therefore, this system was chosen to test the feasibility of cell isolation with rvCD71apt.
To generate cells for sorting, bulk PBMCs were treated with cytokines and CD3/CD28 stimulation beads for 3 days. On day 3, the cells were first washed to remove stimulation beads named pre-sort fraction). During this stimulation process, the fraction of non-T cell (i.e., NK cells, B cells, and monocytes) populations reduced while the T cells preferentially proliferated, resulting in 83% T cells in the total pre-sort population (
An Aptamer-Based Multiplexed System for Traceless Cell Selection
The amount of cells retained in the MACS column after reversal agent treatment led to consideration of using a different isolation method (
Therefore, multiplexed cell isolation was tested using MyOne™ Streptavidin C1 Dynabeads™ (Invitrogen) labeled with rvCD71apt and rvCD8apt aptamers (
In contrast to the MACS system, most of the target cells were effectively from the aptamer-functionalized Dynabeads™ (not eluted fraction in
The most common strategy to achieve traceless cell isolation is by capturing undesired cells with a panel of staining reagents, which often results in insufficient yield and purity9, 10, 17. There is therefore significant interest in developing removable affinity agents to capture target cells, including recombinant proteins and synthetic oligonucleotides. Cell sorting systems using Fab-Streptamer® complexes can achieve cell purity over 90%22, 23, 45, with yields between 10-60%22 when used to isolate cell types that require multiple marker labeling such as T cell subsets22,45. MACS-based cell isolation method using a single aptamer and reversal agent pair to purify one target cell type has been reported to provide high isolation purity of around 95%16,34. Herein, a new isolation method has been demonstrated using two pairs of aptamers and reversal agents as cell type-releasing DNA gates25,26 to capture and to serially elute activated CD4+ T cells and CD8+ cells with 85-93% purity without repeated staining and sorting steps (
Non-specific cell binding to the solid support is a potential factor in reduced purity. Dynabeads™ are superparamagnetic beads coated with polystyrene which has been reported to non-specifically bind to cells46,47. Another potential factor that might cause reduced purity is unintentional physical removal of captured cells from the beads by pipetting or by charge repulsion due to the application of anionic reagents. This is a concern especially in the CD8+ cell elution step, which occurs after CD71 reversal agent elution, as any un-eluted CD71+ cells would have less aptamer binding and be retained less securely by the beads. Since there was only a small amount of CD71+ (<1%) cells observed in the rvCD8apt reversal agent elution fraction (
In the process of developing the multiplexed cell isolation method, a varying yield of target cells was noticed between different donors (
In sum, the inventors have demonstrated the feasibility of aptamer-based, traceless isolation of two target cell types from a single cell sorting procedure. The inventors first modified a reported CD71-binding aptamer, rvCD71apt, so that it was suitable for cell retention on the solid supports and independent reversal agent elution of the bound targets. They also optimized the reversal agent for this aptamer that could trigger rapid and efficient strand displacement to reverse target binding. It was also demonstrated that rvCD71apt could separate CD71+ cells in PBMCs according to their CD71 expression level. rvCD71apt was then with rvCD8 apt and traceless serial elution of activated CD4+ T cells (rvCD71apt target) and resting CD8+ cells (rvCD8apt target) from mixed cell populations was demonstrated. The two reversal agent elution steps successfully displaced most target cells captured on the Dynabeads™ through strand displacement with 93% and 85% mean target cells purity. Theoretically, this approach can be scaled up for isolation of even more cell populations with the development of additional orthogonal aptamer-reversal agent pairs. This study shows that as long as the aptamers and reversal agents are designed properly, the multiplexed isolation system can be versatile for a wide range of applications and different setups.
The following variant aptamer sequences are specifically contemplated for improvements in affinity and/or specificity for binding to transferrin receptor 1/CD71.
Aptamer tJBA8.26 sequence: GCAGCAGCGTAAAGGGGGTGTTTGTGCGG TGTGGAGTGCGCGCGCTGCTGC (SEQ ID NO: 26) was fused to a drug loading overhang (DLO) (sequence GCAGAAGAAGAAGAAGAAGAAGAAGAAGAACG; SEQ ID NO: 140). The DLO can be used to load drugs such as nucleic acid binders or nucleic acids with complementary sequences to the DLO. As a model drug, complementary sequence to the DLO with a fluorescein label was used in this example. As a control, a mutated tJBA8.26 sequence was used. The specific sequences used are shown below:
Aptamers were folded and annealed with the labeled complementary strand in one step at 10 μM by incubating at 95° C. for 5 min, and snap cooling on ice for 10 min in Wash Buffer (WB). (
This application claims the benefit of U.S. Patent Application No. 63/222,086, filed Jul. 15, 2021, expressly incorporated herein by reference in its entirety.
This invention was made with government support under Grant Nos. R01 CA177272 and U54 CA199090, awarded by the National Institutes of Health. The government has certain rights in the invention.
Number | Date | Country | |
---|---|---|---|
63222086 | Jul 2021 | US |