US20210363204A1 - Protoxin-ii variants and methods of use - Google Patents

Protoxin-ii variants and methods of use Download PDF

Info

Publication number
US20210363204A1
US20210363204A1 US17/306,572 US202117306572A US2021363204A1 US 20210363204 A1 US20210363204 A1 US 20210363204A1 US 202117306572 A US202117306572 A US 202117306572A US 2021363204 A1 US2021363204 A1 US 2021363204A1
Authority
US
United States
Prior art keywords
variant
protoxin
amino acid
cooh
pain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US17/306,572
Inventor
Mack Flinspach
Alan Wickenden
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Janssen Biotech Inc
Original Assignee
Janssen Biotech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Janssen Biotech Inc filed Critical Janssen Biotech Inc
Priority to US17/306,572 priority Critical patent/US20210363204A1/en
Assigned to JANSSEN BIOTECH, INC. reassignment JANSSEN BIOTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FLINSPACH, MACK, WICKENDEN, ALAN
Publication of US20210363204A1 publication Critical patent/US20210363204A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43513Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from arachnidae
    • C07K14/43518Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from arachnidae from spiders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the present invention relates to Protoxin-II variants, synthetic polynucleotides encoding them, and methods of making and using the foregoing.
  • Voltage-gated sodium channels are present in all excitable cells including cardiac and skeletal muscle cells and central and peripheral neurons. In neuronal cells, sodium channels are responsible for amplifying sub-threshold depolarizations and generating the rapid upstroke of the action potential. As such, sodium channels are essential to the initiation and propagation of electrical signals in the nervous system.
  • Sodium channels are typically a complex of various subunits, the principal one being the pore-forming alpha-subunit, which is alone sufficient for function.
  • Nav1.1-Nav1.9 Nine known members of the family of voltage-gated sodium channel alpha subunits exist in humans, Nav1.1-Nav1.9.
  • the Navl.x subfamily can be pharmacologically subdivided into two groups, the tetrodotoxin (TTX)-sensitive and TTX-resistant.
  • Nav1.7 (a.k.a. PN1 or hNE) is encoded by the SCN9A gene, is TTX-sensitive and is primarily expressed in peripheral sympathetic and sensory neurons.
  • Nav1.7 accumulates at nerve fiber endings and amplifies small subthreshold depolarizations and acts as a threshold channel that regulates excitability.
  • Nav1.7 function is implicated in various pain states, including acute, inflammatory and/or neuropathic pain.
  • gain of function mutations of Nav1.7 have been linked to primary erythemalgia (PE), a disease characterized by burning pain and inflammation of the extremities (Yang et al., J Med Genet 41:171-174, 2004), and paroxysmal extreme pain disorder (PEPD)(Fertleman et al., Neuron 52:767-774, 2006).
  • PE primary erythemalgia
  • PPD paroxysmal extreme pain disorder
  • non-selective sodium channel blockers lidocaine, mexiletine and carbamazepine can provide symptomatic relief in these painful disorders (Legroux-Crespel et al., Ann Dermatol Venereol 130:429-433, 2003; Fertleman et al., Neuron 52:767-774, 2006).
  • CIP congenital indifference to pain
  • Single nucleotide polymorphisms in the coding region of SCN9A have been associated with increased nociceptor excitability and pain sensitivity.
  • a polymorphism rs6746030 resulting in R1150W substitution in human Nav1.7 has been associated with osteoarthritis pain, lumbar discectomy pain, phantom pain, and pancreatitis pain (Reimann et al., Proc Natl Acad Sci USA 107:5148-5153, 2010).
  • DRG neurons expressing the R1150W mutant Nav1.7 display increased firing frequency in response to depolarization (Estacion et al., Ann Neurol 66:862-866, 2009).
  • a disabling form of fibromyalgia has been associated with SCN9A sodium channel polymorphism rs6754031, indicating that some patients with severe fibromyalgia may have a dorsal root ganglia sodium channelopathy (Vargas-Alarcon et al., BMC Musculoskelet Disord 13:23, 2012).
  • mice deletion of the SCN9A gene in nociceptive neurons leads to reduction in mechanical and thermal pain thresholds and reduction or abolition of inflammatory pain responses (Nassar et al., Proc Natl Acad Sci USA 101:12706-12711, 2004). Ablating SCN9A in all sensory neurons abolished mechanical pain, inflammatory pain and reflex withdrawal responses to heat. Deleting SCN9A in both sensory and sympathetic neurons abolished mechanical, thermal and neuropathic pain, and recapitulated the pain-free phenotype seen in humans with Nav1.7 loss-of-function mutations (Minett et al., Nat Commun 3:791, 2012). Nav1.7 inhibitors or blockers may therefore be useful in the treatment of a wide range of pain associated with various disorders.
  • Spider venoms are known to contain a large number of sodium channel blocking peptides, including Huwentoxin-IV (HwTx-IV) (Peng et al., J Biol Chem 277:47564-47571, 2002), Protoxin-1, Protoxin-II (Middleton et al., Biochemistry 41:14734-14747, 2002) and Phrixotoxin-III (Bosmans et al., Mol Pharmacol 69:419-429, 2006).
  • HwTx-IV Huwentoxin-IV
  • Protoxin-1 Protoxin-1
  • Protoxin-II Meddleton et al., Biochemistry 41:14734-14747, 2002
  • Phrixotoxin-III Phrixotoxin-III
  • One embodiment of the invention is an isolated Protoxin-II variant, wherein the Protoxin-II variant inhibits human Nav1.7 activity with an IC 50 value of about 1 ⁇ 10 ⁇ 7 M or less, about 1 ⁇ 10 ⁇ 8 M or less, about 1 ⁇ 10 ⁇ 9 M or less, about 1 ⁇ 10 ⁇ 10 M or less, about 1 ⁇ 10 ⁇ 11 M or less, or about 1 ⁇ 10 ⁇ 12 M or less, wherein the IC 50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7, wherein the Protoxin-II variant has a W7Q and/or a W30L substitution.
  • FRET fluorescence resonance energy transfer
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence of SEQ ID NOs: 30, 40, 44, 52, 56, 59, 65, 78, 109, 110, 111, 114, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 162, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 177, 178, 179, 180, 182, 183, 184, 185, 186, 189, 190, 193, 195, 197, 199,
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKWMQTCDSERKCCEGMVCRLWCKKKLL-COOH); wherein the amino acid sequence has Q at position 7 and L at position 30, when residue numbering is according to SEQ ID NO: 1; and the polypeptide inhibits human Nav1.7 activity with an IC 50 value of about 30 ⁇ 10 ⁇ 9 M or less, wherein the IC 50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • FRET fluorescence resonance energy transfer
  • Another embodiment of the invention is an isolated fusion protein comprising the Protoxin-II variant of the invention conjugated to a half-life extending moiety.
  • Another embodiment of the invention is an isolated polynucleotide encoding the Protoxin-II variant of the invention.
  • Another embodiment of the invention is an vector comprising the isolated polynucleotide of the invention.
  • Another embodiment of the invention is a host cell comprising the vector of the invention.
  • Another embodiment of the invention is a method of producing the isolated Protoxin-II variant of the invention, comprising culturing the host cell of the invention and recovering the Protoxin-II variant produced by the host cell.
  • Another embodiment of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the isolated Protoxin-II variant or fusion protein of the invention and a pharmaceutically acceptable excipient.
  • Another embodiment of the invention is a method of treating Nav1.7-mediated pain in a subject, comprising administering to a subject in need thereof an effective amount of the Protoxin-II variant or the fusion protein of the invention to treat the pain.
  • FIG. 1 shows the genus amino acid sequence of Protoxin-II variants that inhibit Nav1.7 with an IC 50 value of 30 nM or less in a FLIPR Tetra assay. Residue numbering is according to wild-type Protoxin-II of SEQ ID NO: 1. Genus SEQ ID NO: 403.
  • FIGS. 2A and 2B show the IC 50 values for Nav1.7 and Nav1.6 inhibition in a QPatch assay, and selectivity of each variant calculated by ratio of IC 50 (Nav1.6)/IC 50 (Nav1.7) obtained in QPatch assay. SE: standard error.
  • FIGS. 3A, 3B and 3C show the sequences and the genus sequence of Protoxin-II variants that inhibit Nav1.7 with an ICs value of 30 nM or less in a FLIPR Tetra assay, and are over 30-fold selective over Nav1.6. Selectivity of each variant was calculated by ratio of IC 50 (Nav1.6)/IC 50 (Nav1.7) obtained in QPatch assay. Residue numbering is according to wild-type Protoxin-II of SEQ ID NO: 1.
  • FIG. 4A shows efficacy of NV1D3034 (NV1D3034-OH) (SEQ ID NO: 78) against CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1).
  • NV1D3034-OH NV1D3034-OH
  • SEQ ID NO: 78 CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1).
  • FIG. 4B shows efficacy of NV1D3034 (NV1D3034-OH) (SEQ ID NO: 78) in CFA-induced thermal hyperalgesia expressed as percent MPE (maximum possible effect) (MPE %) at each dose on day 1 following peptide administration.
  • MPE maximum possible effect
  • FIG. 5A shows efficacy of NV1 D3368 (NV1 D3368-OH) (SEQ ID NO: 198) against CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1). **P ⁇ 0.01 and ****P ⁇ 0.0001 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 5B shows efficacy of NV1 D3368 (NV1 D3368-OH) (SEQ ID NO: 198) in CFA-induced thermal hyperalgesia expressed as percent MPE (MPE %) at each dose on day 1 following peptide administration.
  • MPE percent MPE
  • FIG. 6A shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) against CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1).
  • FIG. 6B shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) in CFA-induced thermal hyperalgesia expressed as percent MPE (MPE %) at each dose on day 1 following peptide administration.
  • MPE percent MPE
  • FIG. 6C shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) against CFA-induced tactile allodynia. Tactile thresholds of hind paw before (pre-CFA) and after CFA (0) and 1-day after peptide administration (1). ****P ⁇ 0.0001 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 6D shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) against CFA-induced tactile allodynia expressed as percent MPE (MPE %) on day 1 following peptide. ***P ⁇ 0.001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 7A shows time course of NV1 D2775-OH mediated reversal of thermal hyperalgesia in the CFA model as assessed by measurement of paw withdrawal latency in the Hargreaves test before and after CFA and at various time points post-peptide administration. **P ⁇ 0.01 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison. Shaded areas indicate compound delivery period (0-24 hr).
  • FIG. 7B shows time course of NV1 D2775-OH mediated reversal of tactile allodynia in the CFA model as assessed by measurement of tactile threshold before and after CFA and at various time points post-peptide administration. **P ⁇ 0.01 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison. Shaded areas indicate compound delivery period (0-24 hr).
  • FIG. 8 shows that NV1 D2775-OH produced significant analgesia in the hotplate test. Thermal withdrawal latency was evaluated at 50 and 55° C. pre- and post-pump implantation. Pump implantation had no impact on the latency in the control PBS group. One day after pump, NV1 D2775-OH treated-mice exhibited prolonged latency compared to the PBS group. *P ⁇ 0.05 and ****P ⁇ 0.0001 vs. PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 9 shows that NV1 D2775-OH pretreatment protected animals from carrageenan-induced thermal hyperalgesia. Paw withdrawal latencies were measured pre- and on day 1 post-pump before intraplantar carrageenan injection. Latencies were measured again at 2, 3 and 4 hr following carrageenan.
  • FIG. 10 shows the surface representation of the NMR structure of the wild type Protoxin-II.
  • a hydrophobic face shown on left includes residues W5, M6, W7, L23 and W24.
  • a selectivity face is shown on the right and includes residues S11, E12, K14, E17, G18, L29 and W30. Residue numbering according to SEQ ID NO: 1.
  • FIG. 11A shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) after a single intrathecal (IT) administration in the tail flick test. Tail withdrawal latency to a thermal stimulus was measured at the indicated time post-peptide administration.
  • FIG. 11B shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) in the tail flick test expressed as percent area under the curve (AUC %) in the first 120 min after a single intrathecal (IT) administration.
  • AUC % percent area under the curve
  • FIG. 11C shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) after a single intrathecal (IT) administration in the hot plate test (52.5° C.). The latency of a nociceptive response on a hot plate was measured at the indicated time post-peptide administration.
  • FIG. 11D shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) in the hot plate test expressed as percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration.
  • FIG. 11E shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) in the formalin test. Injection of formalin into the rat hindpaw induced a bi-phasic flinching behavior. Total number of flinches in Phase I (0-10 min post formalin) and Phase II (11-60 min post formalin) was measured by an automated device. No statistics were performed in E) due to small group size.
  • FIG. 12A shows efficacy of NV1 D2775-OH after a single intrathecal (IT) administration in the tail flick test. Tail withdrawal latency to a thermal stimulus was measured at the indicated time post-peptide administration.
  • FIG. 12B shows efficacy of NV1 D2775-OH in the tail flick test expressed as percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration.
  • AUC % percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration.
  • FIG. 12C shows efficacy of NV1 D2775-OH after a single intrathecal (IT) administration in the hot plate test (52.5° C.). The latency of a nociceptive response on a hot plate was measured at the indicated time post-peptide administration.
  • FIG. 12D shows efficacy of NV1 D2775-OH in the hot plate test expressed as percent area under the curve (AUC %) in the first 120 min after a single intrathecal (IT) administration. **P ⁇ 0.01 and ****P ⁇ 0.0001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 12E shows efficacy of NV1 D2775-OH in the formalin test.
  • Injection of formalin into the rat hindpaw induced a bi-phasic flinching behavior.
  • Total number of flinches in Phase I (0-10 min post formalin) and Phase II (11-60 min post formalin) was measured by an automated device. **P ⁇ 0.01 vs PBS, phase I, *P ⁇ 0.05 vs PBS, phase II, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 13A shows efficacy of NV1D3034-OH after a single intrathecal (IT) administration in the tail flick test. Tail withdrawal latency to a thermal stimulus was measured at the indicated time post-peptide administration.
  • FIG. 13B shows efficacy of NV1D3034-OH in the tail flick test expressed as percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration. ***P ⁇ 0.005 vs PBS, t-test.
  • FIG. 13C shows efficacy of NV1D3034-OH after a single intrathecal (IT) administration in the hot plate test (52.5° C.). The latency of a nociceptive response on a hot plate was measured at the indicated time post-peptide administration.
  • FIG. 13D shows efficacy of NV1D3034-OH in the hot plate test expressed as percent area under the curve (AUC %) in the first 120 min after a single intrathecal (IT) administration. **P ⁇ 0.01 vs PBS, t-test.
  • FIG. 13E shows efficacy of NV1D3034-OH in the formalin test. Injection of formalin into the rat hindpaw induced a bi-phasic flinching behavior. Total number of flinches in Phase I (0-10 min post formalin) and Phase II (11-60 min post formalin) was measured by an automated device. *P ⁇ 0.05 vs PBS, phase I, **P ⁇ 0.01 vs PBS, phase II, t-test.
  • polypeptide means a molecule that comprises at least two amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than 50 amino acids may be referred to as “peptides.” Polypeptides may also be referred as “proteins.”
  • polynucleotide means a molecule comprising a chain of nucleotides covalently linked by a sugar-phosphate backbone or other equivalent covalent chemistry. Double and single-stranded DNAs and RNAs are typical examples of polynucleotides.
  • complementary sequence means a second isolated polynucleotide sequence that is antiparallel to a first isolated polynucleotide sequence and that comprises nucleotides complementary to the nucleotides in the first polynucleotide sequence.
  • vector means a non-natural polynucleotide capable of being duplicated within a biological system or that can be moved between such systems.
  • Vector polynucleotides typically contain a cDNA encoding a protein of interest and additional elements, such as origins of replication, polyadenylation signal or selection markers, that function to facilitate the duplication or maintenance of these polynucleotides in a biological system.
  • additional elements such as origins of replication, polyadenylation signal or selection markers, that function to facilitate the duplication or maintenance of these polynucleotides in a biological system.
  • Examples of such biological systems may include a cell, virus, animal, plant, and reconstituted biological systems utilizing biological components capable of duplicating a vector.
  • the polynucleotide comprising a vector may be DNA or RNA molecules or a hybrid of these.
  • expression vector means a vector that can be utilized in a biological system or a reconstituted biological system to direct the translation of a polypeptide encoded by a polynucleotide sequence present in the expression vector.
  • variant refers to a polypeptide or a polynucleotide that differs from wild type Protoxin-II polypeptide of SEQ ID NO: 1 or the polynucleotide encoding the wild type Protoxin-II having the sequence of SEQ ID NO: 107 by one or more modifications for example, substitutions, insertions or deletions of nucleotides or amino acids.
  • residues that are substituted in the Protoxin-II variants are numbered corresponding to their position in the wild-type Protoxin-II of SEQ ID NO: 1.
  • Y1A in the specification refers to the substitution of tyrosine at residue position that corresponds to the position 1 in the wild type Protoxin-II of SEQ ID NO:1 with alanine.
  • Codons encoding the initiator methionine may or may not be present in cDNA.
  • cDNA may be synthesized for example by reverse transcription or synthetic gene assembly.
  • Synthetic or “non-natural” as used herein refers to a polynucleotide or a polypeptide molecule not present in nature.
  • Nav1.7 (also referred to as hNE or PN1) or “hNav1.7” as used herein refers to the well-known human sodium channel protein type 9 subunit alpha having a sequence shown in GenBank accession number NP 002968.1 and in SEQ ID NO: 79.
  • wild type Protoxin-II or wild type ProTx-II as used herein refers to the tarantula Thrixopelma pruriens (Peruvian green velvet tarantula) toxin peptide having the amino acid sequence YCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH (SEQ ID NO: 1) as described in Middleton et al., Biochemistry 41(50):14734-47, 2002.
  • Recombinant Protoxin-II refers to the recombinant Protoxin-II obtained from expression and subsequent cleavage of a Protoxin-II fusion protein having the sequence of GPYCQKWMWTCDSERKCCEGMVCRLWCKKKLW-OH as shown in SEQ ID NO: 2.
  • Recombinant Protoxin-II incorporates a two amino acid N-terminal extension (residues G and P) when compared to the wild type Protoxin-II.
  • “Blocks human Nav1.7 activity” or “inhibits human Nav1.7 activity” as used herein refers to the ability of the Protoxin-II variant of the invention to reduce membrane depolarization induced by veratridine (3-veratroylveracevine) with an IC 50 value of about 1 ⁇ 10 ⁇ 7 M or less in a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET), where veratridine-induced depolarization is measured as a reduction in FRET signal using DISBAC2(3) ([bis-(1,3-diethylthiobarbituric acid) trimethine oxonol]) as an acceptor and PTS18 (trisodium 8-octadecyloxypyrene-1,3,6-trisulfonate) as a donor by exciting the donor at 390-420 nm and measuring FRET at 515-575 nm in a cell line stably expressing human Nav1.7.
  • FRET flu
  • FLIPR® Tetra membrane depolarization assay as used herein is the assay described in Example 3.
  • substantially identical means that the two Protoxin-II variant amino acid sequences being compared are identical or have “insubstantial differences”. Insubstantial differences are substitutions of 1, 2, 3, 4, 5, 6, or 7 amino acids in the Protoxin-II variant amino acid sequence that do not adversely affect peptide properties.
  • Amino acid sequences substantially identical to the Protoxin-II variants disclosed herein are within the scope of the application. In some embodiments, the sequence identity can be about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher.
  • Percent identity can be determined for example by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9.0.0 (Invitrogen, Carslbad, Calif.).
  • the protein sequences of the present invention may be used as a query sequence to perform a search against public or patent databases, for example, to identify related sequences.
  • Exemplary programs used to perform such searches are the XBLAST or BLASTP programs (http //www ncbi nlm/nih gov), or the GenomeQuestTM (GenomeQuest, Westborough, Mass.) suite using the default settings.
  • the present invention provides isolated Protoxin-II (ProTx-II) variant polypeptides that inhibit human Nav1.7 activity, polynucleotides encoding them, vectors, host cells, and methods of using the polynucleotides and polypeptides of the invention.
  • the polypeptides of the invention inhibit depolarization resulting from Nav1.7 activation, and therefore may be useful in the treatment of various conditions associated with pain and conditions associated with sensory or sympathetic neuron dysfunction.
  • the variants of the invention are potent inhibitors of Nav1.7.
  • the current invention is based, at least in part, on the finding that certain residue substitutions in Protoxin-II enhance selectivity, synthetic yield and/or homogeneity without adversely affecting the potency of the generated Protoxin-II variants, specifically W7 and M19, and additionally residues Y1 and S11, and further additionally residues E12, R22 and (residue numbering according to SEQ ID NO: 1).
  • substitutions at positions W7 and W30 enhance the Protoxin-II variant folding and improve yield.
  • substitutions at positions S11, E12, K14, E17, G18, L29 and W30 improve selectivity of the resulting Protoxin-II variants to Nav1.7.
  • One embodiment of the invention is an isolated Protoxin-II variant, wherein the Protoxin-II variant inhibits human Nav1.7 activity with an IC 50 value of about 1 ⁇ 10 ⁇ 7 M or less, about 1 ⁇ 10 ⁇ 8 M or less, about 1 ⁇ 10 ⁇ 9 M or less, about 1 ⁇ 10 ⁇ 10 M or less, about 1 ⁇ 10 ⁇ 11 M or less, or about 1 ⁇ 10 ⁇ 12 M or less, wherein the IC 50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 9 M 3veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • FRET fluorescence resonance energy transfer
  • One embodiment of the invention is an isolated Protoxin-II variant, wherein the Protoxin-II variant inhibits human Nav1.7 activity with an ICs value of about 1 ⁇ 10 ⁇ 7 M or less, about 1 ⁇ 10 ⁇ 8 M or less, about 1 ⁇ 10 ⁇ 9 M or less, about 1 ⁇ 10 ⁇ 10 M or less, about 1 ⁇ 10 ⁇ 11 M or less, or about 1 ⁇ 10 ⁇ 12 M or less, wherein the ICs value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7, wherein the Protoxin-II variant has a W7Q and a W30L substitution.
  • FRET fluorescence resonance energy transfer
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the sequence X 1 X 2 X 3 CX 4 X 5 WX 6 QX 7 CX 3 X 9 X 10 X 11 X 12 CCX 13 X 14 X 15 X 16 CX 17 LWCX 13 KKLX 19 (SEQ ID NO: 432), wherein
  • X 1 is G, P, A or deleted
  • X 2 is P, A or deleted
  • X 3 is S, Q, A, R or Y;
  • X 4 is Q, R, K, A, S or Y;
  • X 5 is K, S, Q or R;
  • X 6 is M or F
  • X 7 is T, S, R, K or Q
  • X 8 is D, T, or asparagyl-4-aminobutane
  • X 9 is S, A, R, I or V;
  • X 10 is E, R, N, K, T, Q, Y or glutamyl-4-aminobutane
  • X 11 is R or K
  • X 12 is K, Q, S, A or F
  • X 13 is E, Q, D, L, N, or glutamyl-4-aminobutane
  • X 14 is G, Q or P;
  • X 15 is M
  • X 16 is V or S
  • X 17 is R, T or N-omega methyl-L-arginine
  • X 18 is K or R
  • X 19 is W or L
  • polypeptide inhibits human Nav1.7 activity with an IC 50 value of about 1 ⁇ 10 ⁇ 7 M or less, wherein the IC 50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • FRET fluorescence resonance energy transfer
  • the N-terminal extension comprises the amino acid sequences of SEQ ID NOs: 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384 or 385.
  • the C-terminal extension comprises the amino acid sequence of SEQ ID NOs: 374, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396 or 397.
  • the N-terminal and/or the C-terminal extension is conjugated to the Protoxin-II variant via a linker.
  • the linker comprises the amino acid sequence of SEQ ID NOs: 383, 392, 398, 399, 400, 401 or 402.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the sequence X 1 X 2 X 3 CX 4 X 5 WX 6 QX 7 CX 8 X 9 X 10 X 11 X 12 CCX 13 X 14 FX 15 CX 16 LWCX 17 KKLW (SEQ ID: 403), wherein
  • X 1 is G, P, A or deleted
  • X 2 is P, A or deleted
  • X 3 is S, Q, A, R, or Y;
  • X 4 is Q, R, K, A, or S;
  • X 5 is K, S, Q or R;
  • X 6 is M
  • X 7 is T, S, R, K or Q
  • X 8 is D, S or T
  • X 9 is S, A or R;
  • X 10 is E, R, N, K, T or Q;
  • X 11 is R or K
  • X 12 is K, Q, S, R or A
  • X 13 is E, Q, or D
  • X 14 is G or Q
  • X 15 is V or S
  • X 16 is R or T
  • X 17 is K or R
  • the core amino acid sequence inhibits human Nav1.7 activity with an IC 50 value of about 30 ⁇ 10 ⁇ 9 or less, wherein the IC 50 value is measured using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 6 M 3-veratrolyveracevine in HEK293 cells stably expressing human Nav1.7 and using bis-1,3(diethylthiobarbituric acid) trimethine oxonol as an electron acceptor and trisodium 8-octadecylopxypyrene-1,3,6-trisulfonate as a donor by exciting the donor at 390-420 nm and measuring FRET at 515-575 nm, optionally having an N-terminal extension or a C-terminal extension, wherein the polypeptide inhibits human Nav1.7 activity with an ICs value of about 1 ⁇ 10 ⁇ 7 M or less, wherein the ICs value is measured using a FLIPR® Tetra membrane depolarization assay using fluor
  • the Protoxin-II variants of the invention are potent Nav1.7 inhibitors.
  • Recombinant Protoxin-II (SEQ ID NO: 2) has an ICs value of about 4 ⁇ 10 ⁇ 9 M for human Nav1.7 in a veratridine-induced depolarization inhibition assay measuring decline in FRET (fluorescence resonance energy transfer) in cells stably expressing Nav1.7 using FLIPR® Tetra instrument (Molecular Devices) using experimental details described in Example 3.
  • a Protoxin-II variant is a “potent” Nav1.7 inhibitor when the ICs value in the assay described above and in Experiment 3 is about 30 ⁇ 10 ⁇ 9 M or less i.e., within 10-fold of recombinant Protoxin-II.
  • an ICs of 30 ⁇ 10 ⁇ 9 M is identical to ICs of 3.0 ⁇ 10 ⁇ 8 M.
  • the Protoxin-II variant polypeptides of the invention may be produced by chemical synthesis, such as solid phase peptide synthesis, on an automated peptide synthesizer.
  • the polypeptides of the invention may be obtained from polynucleotides encoding the polypeptides by the use of cell-free expression systems such as reticulocyte lysate based expression systems, or by recombinant expression systems. Those skilled in the art will recognize other techniques for obtaining the polypeptides of the invention.
  • the Protoxin-II variants of the invention are generated by expressing them as human serum albumin (HSA) fusion proteins utilizing a glycine-rich linker such as (GGGGS) 4 (SEQ ID NO:80) or (GGGGS) 6 (SEQ ID NO: 81) coupled to a protease cleavable linker such as a recognition sequence for HRV3C protease (Recombinant type 14 3C protease from human rhinovirus) LEVLFQGP (HRV3C linker) (SEQ ID NO: 82), and cleaving the expressed fusion proteins with the HRV3C protease to release the recombinant Protoxin-II variant peptides.
  • HSA human serum albumin
  • a glycine-rich linker such as (GGGGS) 4 (SEQ ID NO:80) or (GGGGS) 6 (SEQ ID NO: 81) coupled to a protease cleavable linker such
  • Protoxin-II variants of the invention may be purified using methods described herein.
  • Protoxin-II variants of the invention expressed as HSA fusion proteins and cleaved with HRV3C protease may be purified using sold phase extraction (SPE) as described herein.
  • SPE sold phase extraction
  • Protoxin-II variants optionally having N-terminal and/or C-terminal extensions, and Protoxin-II variant fusion proteins is typically achieved at the nucleic acid level.
  • the polynucleotides may be synthesized using chemical gene synthesis according to methods described in U.S. Pat. Nos. 6,521,427 and 6,670,127, utilizing degenerate oligonucleotides to generate the desired variants, or by standard PCR cloning and mutagenesis. Libraries of variants may be generated by standard cloning techniques to clone the polynucleotides encoding the Protoxin-II variants into the vector for expression.
  • the Protoxin-II variants may incorporate additional N- and/or C-terminal amino acids when compared to the wild type Protoxin-II of SEQ ID NO: 1, for example resulting from cloning and/or expression schemes.
  • cleavage from HSA after expression of the variant as HSA-linker-HRV3C cleavable peptide-Protoxin-II variant fusion protein may result in the incorporation of additional two residues to the N-terminus of each Protoxin-II variant, such as G and P.
  • the Protoxin-II variants of the invention are tested for their ability to inhibit human Nav1.7 using methods described herein.
  • An exemplary assay is a veratridine-induced depolarization inhibition assay measuring decline in FRET (fluorescence resonance energy transfer) in cells stably expressing Nav1.7.
  • Another exemplary assay employs electrophysiological recordings to measure changes in Nav1.7-mediated currents using well known patch clamp techniques and as described herein.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence of SEQ ID NOs: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 109, 110, 112, 113, 114, 115, 116, 117, 118, 119, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140
  • the Protoxin-II variants of the invention may inhibit human Nav1.7 with an IC 50 value of about 1 ⁇ 10 ⁇ 7 M or less, about 1 ⁇ 10 ⁇ 8 M about 1 ⁇ 10 ⁇ 9 or less, about 1 ⁇ 10 ⁇ 10 M or less, about 1 ⁇ 10 ⁇ 11 M or less, or about 1 ⁇ 10 ⁇ 12 M or less.
  • variants demonstrating the range of IC 50 values are variants having amino acid sequences shown in SEQ ID NOs: 30, 40, 44, 52, 56, 59, 65, 78, 109, 110, 111, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 162, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 177, 178, 179, 180, 182, 183, 184, 185, 186, 189, 190, 193, 195, 179, 199, 206, 207,
  • Table 2 Table 3 and Table 14 show the sequences of select Protoxin-II variants.
  • the isolated Protoxin-II variant inhibits human Nav1.7 activity with an IC 50 value of about 3 ⁇ 10 ⁇ 9 M or less.
  • the isolated Protoxin-II variant inhibits human Nav1.7 activity with an IC 50 value of between about 3 ⁇ 10 ⁇ 9 M to about 1 ⁇ 10 ⁇ 9 M.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence GPQCX 1 X 2 WX 3 QX 4 CX 5 X 6 X 7 X 3 X 9 CCX 10 X 11 FX 12 CX 13 LWCX 14 KKLL (SEQ ID NO: 433), wherein
  • X 1 is Q, R, K, A or S;
  • X 2 is K, S, Q or R;
  • X 3 is M or F
  • X 4 is T, S, R, K or Q
  • X 5 is D or T
  • X 6 is S, A or R;
  • X 7 is E, R, N, K, T or Q;
  • X 8 is R or K
  • X 9 is K, Q, S or A
  • X 10 is E, Q or D
  • X 11 is G or Q
  • X 12 is V or S
  • X 13 is R or T
  • X 14 is K or R.
  • Exemplary Protoxin-II variants that inhibit human Nav1.7 activity with an ICs value of about 30 ⁇ 10 ⁇ 9 M or less are variants comprising the amino acid sequences of SEQ ID NOs: 56, 78, 111, 114, 117, 118, 119, 122, 123, 129, 130, 131, 132, 133, 134, 135, 136, 138, 139, 140, 141, 142, 145, 146, 147, 149, 150, 151, 152, 153, 154, 156, 158, 159, 165, 172, 173, 175, 177, 178, 183, 184, 185, 186, 189, 190, 193, 197, 199, 207, 210, 211, 216, 217, 224, 266, 273, 282, 335, 408, 409, 410, 422, 424, 425, 426, 427, and 428.
  • the isolated Protoxin-II variant selectively inhibits human Nav1.7.
  • the Protoxin-II variants of the invention may be more selective towards Nav1.7 when compared to the recombinant Protoxin-II (SEQ ID NO: 2).
  • recombinant Protoxin-II has an IC 50 of about 2.2 ⁇ 10 ⁇ 9 M for Nav1.7 and an IC 50 of about 62 ⁇ 10 ⁇ 9 M for Nav1.6, and therefore the ratio of IC 50 for Nav1.6 to IC 50 for Nav1.7 about 28 fold.
  • “Selectivity” or “selective” or “more selective” or “selectively blocks” or “selectively inhibits” when used herein refers to a Protoxin-II variant that has a ratio of IC 50 for Nav1.6 to IC 50 for Nav1.7 (IC 50 (Nav1.6)/IC 50 (Nav1.7)) equal or over about 30.
  • IC 50 for Nav1.6 may be assayed in a QPatch electrophysiology assay using cell lines stably expressing Nav1.6 using similar methods to those described for Nav1.7.
  • Residue positions in Protoxin-II that can be mutagenized to improve selectivity include residues 7, 11, 12, 14, 17, 18 and 19, and optionally residues 1, 20, 22 and 26 (residue numbering according to SEQ ID NO: 1).
  • Exemplary substitutions to improve selectivity are Y1Q, W7Q, S11R, S11A, E12T, M19F, V20S, R22T, and K26R.
  • Exemplary Protoxin-II variants with improved selectivity are variants of SEQ ID NOs: 56, 59, 65, 78, 111, 114, 117, 118, 119, 121, 122, 123, 129, 130, 133, 150, 190, 217, 281, 324, 325 or 326.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the sequence GPX 1 CQKWMQX 2 CDX 3 X 4 RKCCX 5 GFX 6 CX 7 LWCX 8 KKLW (SEQ ID NO: 405); wherein
  • X 1 is Y, Q, A, S or R;
  • X 2 is T or S
  • X 3 is S, R or A
  • X 4 is E, T or N;
  • X 5 is E or Q
  • X 6 is V or S
  • X 7 is R or T
  • X 8 is K or R
  • Protoxin-II variant inhibits human Nav1.7 activity with an IC 50 value of about 3 ⁇ 10 ⁇ 8 M or less, and selectively inhibits human Nav1.7.
  • the isolated Protoxin-II variant comprises the sequence GPQCQKWMQX 1 CDX 2 X 3 RKCCX 4 GFX 5 CX 6 LWCX 8 KKLW (SEQ ID NO: 406); wherein
  • X 1 is Tor S
  • X 2 is S, R or A
  • X 3 is E, T or N;
  • X 4 is E or Q
  • X 5 is V or S
  • X 6 is R or T
  • X 7 is K or R.
  • Another embodiment is an isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKWMQTCDSERKCCEGMVCRLWCKKKLL-COOH); wherein
  • the amino acid sequence has Q at position 7 and L at position 30, when residue numbering is according to SEQ ID NO: 1; and the polypeptide inhibits human Nav1.7 activity with an ICs value of about 30 ⁇ 10 ⁇ 9 M or less, wherein the ICs value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • FRET fluorescence resonance energy transfer
  • Protoxin-II variants having substitutions W7Q and W30L have improved folding, yield and selectivity when compared to the wild type Protoxin-II.
  • Another embodiment is an isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 78 (GPQCQKWMQTCDRERKCCEGFVCTLWCRKKLW-COOH); wherein
  • the amino acid sequence has Q at position 1, Q at position 7 and F at position 19, when residue numbering is according to SEQ ID NO: 1; the polypeptide inhibits human Nav1.7 activity with an IC 50 value of about 30 ⁇ 10 ⁇ 9 M or less, wherein the IC 50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25 ⁇ 10 ⁇ 9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and the polypeptide selectively inhibits Nav1.7.
  • FRET fluorescence resonance energy transfer
  • the isolated Protoxin-II variant has a free C-terminal carboxylic acid, amide, methylamide or butylamide group, which are generated via routine synthetic methods.
  • Another embodiment of the invention is an isolated fusion protein comprising the Protoxin-II variant of SEQ ID NOs: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 109, 110, 112, 113, 114, 115, 116, 117, 118, 119, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140
  • Such second polypeptides may be well known leader or secretory signal sequences, or synthetic sequences resulting for example from cloning steps, or tags such as hexahistidine tag (SEQ ID NO: 108). Such second polypeptide may be a half-life extending moiety.
  • the isolated fusion protein comprises the Protoxin-II variant of the invention conjugated to a half-life extending moiety.
  • Exemplary half-life extending moieties that can be used include well known human serum albumin, transthyretin (TTR), a thyroxine-binding globulin (TGB), albumin-binding domains, or an Fc or fragments thereof.
  • TTR transthyretin
  • TGB thyroxine-binding globulin
  • Fc or fragments thereof include well known human serum albumin, transthyretin (TTR), a thyroxine-binding globulin (TGB), albumin-binding domains, or an Fc or fragments thereof.
  • Biologically suitable polymers or copolymers may also be used, for example ethylene glycol or polyethylene glycol (PEG) molecules, such as PEG5000 or PEG20000, dextran, polylysine, fatty acids and fatty acid esters of different chain lengths, for example laurate, myristate, stearate, arachidate, behenate, oleate, arachidonate, octanedioic acid, tetradecanedioic acid, octadecanedioic acid, docosanedioic acid, and the like, octane, or carbohydrates (dextran, cellulose, oligo- or polysaccharides).
  • PEG polyethylene glycol
  • moieties may be direct fusions with the Protoxin-II variant polypeptides and may be generated by standard cloning and expression techniques. Alternatively, well known chemical coupling methods may be used to attach the moieties to recombinantly produced Protoxin-II variants of the invention.
  • the half-life extending moiety of the fusion protein of the invention is human serum albumin, albumin binding domain (ABD), or polyethylene glycol (PEG).
  • the half-life extending moiety of is conjugated to the Protoxin-II variant via a linker.
  • Suitable linkers are well known and include linkers having the sequence shown in SEQ ID NOs: 80 or 81.
  • Exemplary fusion proteins incorporating Protoxin-II variants of the invention are those having the polypeptide sequence of SEQ ID NOs: 83, 85, 87, 89, 91, 93, 95, 97, 99, 101 or 103.
  • Protoxin-II variants of the invention incorporating additional moieties may be compared for functionality by several well-known assays. For example, pharmacokinetic properties of Protoxin-II variants coupled to PEG may be evaluated in well known in vivo models.
  • Protoxin-II variants and Protoxin-II variant fusion proteins are within the scope of the invention. Additional substitutions to the Protoxin-II variants of the invention can be made as long as the resulting variant or the fusion protein retains similar characteristics when compared to the parent peptide. Exemplary modifications are for example conservative substitutions that will result in Protoxin-II variants with similar characteristics to those of the parent molecules. Conservative replacements are those that take place within a family of amino acids that are related in their side chains.
  • Genetically encoded amino acids can be divided into four families: (1) acidic (aspartate, glutamate); (2) basic (lysine, arginine, histidine); (3) nonpolar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); and (4) uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine). Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids.
  • the amino acid repertoire can be grouped as (1) acidic (aspartate, glutamate); (2) basic (lysine, arginine histidine), (3) aliphatic (glycine, alanine, valine, leucine, isoleucine, serine, threonine), with serine and threonine optionally grouped separately as aliphatic-hydroxyl; (4) aromatic (phenylalanine, tyrosine, tryptophan); (5) amide (asparagine, glutamine); and (6) sulfur-containing (cysteine and methionine) (Stryer (ed.), Biochemistry, 2nd ed, WH Freeman and Co., 1981).
  • Non-conservative substitutions can be made to the Protoxin-II variants that involve substitutions of amino acid residues between different classes of amino acids to improve properties of the Protoxin-II variants and Protoxin-II variant fusion proteins.
  • Whether a change in the amino acid sequence of a polypeptide or fragment thereof results in a functional homolog can be readily determined by assessing the ability of the modified polypeptide or fragment to produce a response in a fashion similar to the unmodified polypeptide or fragment using the assays described herein.
  • Peptides, polypeptides or proteins in which more than one replacement takes place can readily be tested in the same manner.
  • Another embodiment of the invention is an isolated synthetic polynucleotide comprising a polynucleotide encoding the Protoxin-II variant of the invention.
  • Certain exemplary synthetic polynucleotides are disclosed herein, however, other synthetic polynucleotides which, given the degeneracy of the genetic code or codon preferences in a given expression system, encode the Protoxin-II variants and Protoxin-II variant fusion proteins of the invention are also within the scope of the invention.
  • Exemplary synthetic polynucleotides are for example polynucleotide sequences shown in SEQ ID NOs: 84, 86, 88, 90, 92, 94, 96, 98, 100, 102 and 104, which encode the Protoxin-II variant fusion proteins of the invention.
  • the synthetic polynucleotide sequences encoding the Protoxin-II variants or fusion proteins of the invention can be operably linked to one or more regulatory elements, such as a promoter and enhancer, that allow expression of the nucleotide sequence in the intended host cell.
  • the synthetic polynucleotide may be a cDNA.
  • the polynucleotides of the invention may be produced by chemical synthesis such as solid phase polynucleotide synthesis on an automated polynucleotide synthesizer.
  • the polynucleotides of the invention may be produced by other techniques such as PCR based duplication, vector based duplication, or restriction enzyme based DNA manipulation techniques. Techniques for producing or obtaining polynucleotides of known sequences are well known.
  • the polynucleotides of the invention may also comprise at least one non-coding sequence, such as transcribed but not translated sequences, termination signals, ribosome binding sites, mRNA stabilizing sequences, introns and polyadenylation signals.
  • the polynucleotide sequences may also comprise additional sequences encoding additional amino acids. These additional polynucleotide sequences may, for example, encode a marker or well-known tag sequences such as a hexa-histidine (SEQ ID NO: 108) or a HA tag which facilitate the purification of fused polypeptides.
  • Another embodiment of the invention is a vector comprising the polynucleotide of the invention.
  • Such vectors may be plasmid vectors, viral vectors, vectors for baculovirus expression, transposon based vectors or any other vector suitable for introduction of the polynucleotide of the invention into a given organism or genetic background by any means.
  • polynucleotides encoding the Protoxin-II variants or the Protoxin-II variant fusion proteins of the invention are inserted into an expression vector and may be operably linked to control sequences in the expression vector to ensure efficient expression, such as signal sequences, promoters (e.g.
  • the vector Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the proteins encoded by the incorporated polynucleotides.
  • Suitable expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors contain selection markers such as ampicillin-resistance, hygromycin-resistance, tetracycline resistance, kanamycin resistance or neomycin resistance to permit detection of those cells transformed with the desired DNA sequences.
  • Suitable promoter and enhancer elements are known in the art.
  • suitable promoters include, but are not limited to, lacI, lacZ, T3, T7, gpt, lambda P and trc.
  • suitable promoters include, but are not limited to, light and/or heavy chain immunoglobulin gene promoter and enhancer elements; cytomegalovirus immediate early promoter; herpes simplex virus thymidine kinase promoter; early and late SV40 promoters; promoter present in long terminal repeats from a retrovirus; mouse metallothionein-I promoter; and various art-known tissue specific promoters.
  • a suitable promoter is a constitutive promoter such as an ADH1 PGK1, ENO or PYK1 promoter and the like, or a regulatable promoter such as a GALL or GAL10 promoter. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • Bacterial pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene, La Jolla, Calif., USA); pTrc99A, pKK223-3, pKK233-3, pDR540, and pRIT5 (Pharmacia, Uppsala, Sweden).
  • Eukaryotic pWLneo, pSV2cat, pOG44, PXR1, pSG (Stratagene) pSVK3, pBPV, pMSG and pSVL (Pharmacia).
  • An exemplary vector for expression of the Protoxin-II variants or Protoxin-II variant fusion proteins is a vector having ampicillin-resistance selection marker, CMV promoter, CMV intron, signal peptide, neomycin resistance, f1 origin of replication, SV40 polyadenylation signal, and cDNA encoding the Protoxin-II variant or the Protoxin-II variant fusion protein of the invention.
  • host cell refers to a cell into which a vector has been introduced. It is understood that the term host cell is intended to refer not only to the particular subject cell but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein.
  • host cells may be eukaryotic cells, prokaryotic cells, plant cells or archaeal cells.
  • Escherichia coli bacilli, such as Bacillus subtilis , and other enterobacteriaceae, such as Salmonella, Serratia , and various Pseudomonas species, are examples of prokaryotic host cells.
  • Other microbes, such as yeast are also useful for expression. Saccharomyces (e.g., S. cerevisiae ) and Pichia are examples of suitable yeast host cells.
  • Exemplary eukaryotic cells may be of mammalian, insect, avian or other animal origins.
  • Mammalian eukaryotic cells include immortalized cell lines such as hybridomas or myeloma cell lines such as SP2/0 (American Type Culture Collection (ATCC), Manassas, Va., CRL-1581), NSO (European Collection of Cell Cultures (ECACC), Salisbury, Wiltshire, UK, ECACC No. 85110503), FO (ATCC CRL-1646) and Ag653 (ATCC CRL-1580) murine cell lines.
  • An exemplary human myeloma cell line is U266 (ATTC CRL-TIB-196).
  • Other useful cell lines include those derived from Chinese Hamster Ovary (CHO) cells such as CHO-K1SV (Lonza Biologics, Walkersville, Md.), CHO-K1 (ATCC CRL-61) or DG44.
  • a polynucleotide such as a vector
  • Introduction of a polynucleotide, such as a vector, into a host cell can be effected by methods well known to those skilled in the art. Exemplary methods are calcium phosphate transfection, DEAE-Dextran mediated transfection, microinjection, cationic lipid-mediated transfection and electroporation.
  • Another embodiment of the invention is a method for producing the Protoxin-II variant of the invention comprising the steps of providing a host cell of the invention; and culturing the host cell under conditions sufficient for the expression of at least one Protoxin-II variant of the invention.
  • Host cells can be cultured under any conditions suitable for maintaining or propagating a given type of host cell and sufficient for expressing a polypeptide.
  • Culture conditions, media, and related methods sufficient for the expression of polypeptides are well known in the art.
  • many mammalian cell types can be aerobically cultured at 37° C. using appropriately buffered DMEM media while bacterial, yeast and other cell types may be cultured at 37° C. under appropriate atmospheric conditions in LB media.
  • the expression of the Protoxin-II variant can be confirmed using a variety of well-known methods.
  • expression of a polypeptide can be confirmed using detection reagents, such as using SDS-PAGE or HPLC.
  • Another aspect of the invention is a method of modulating the activity of Nav1.7 in a biological tissue, the method comprising contacting the biological tissue expressing Nav1.7 with a Nav1.7-modulating amount of the Protoxin-II variant of the invention.
  • Protoxin-II variants of the invention may be utilized in any therapy where it is desired to treat, reduce or alleviate symptoms of pain or other disorders of sensory or sympathetic neuron dysfunction.
  • Pain treated with the Protoxin-II variants of the invention may be any type of pain, such as chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, pain associated with inflammatory conditions, post-operative pain, thermal pain or pain associated with disease and degeneration.
  • Pain treated with the Protoxin-II variants of the invention may be Nav1.7-mediated pain.
  • Nav1.7-mediated pain refers to pain resulting at least partially from increased Nav1.7 channel activity.
  • the methods of the invention may be used to treat an animal patient belonging to any classification.
  • animals include mammals such as humans, rodents, dogs, cats and farm animals.
  • the pain and/or Nav1.7-mediated pain may result from one or more causes, such as peripheral neuropathy, central neuropathy, nerve compression or entrapment syndromes such as carpal tunnel syndrome, tarsus tunnel syndrome, ulnar nerve entrapment, compression radiculopathy, lumbar spinal stenosis, sciatic nerve compression, spinal root compression, intercostal neuralgia, compression radiculopathy and radicular lower back pain, spinal root lesions, neuritis, autoimmune diseases, general inflammation, chronic inflammatory conditions, arthritis, rheumatic diseases, lupus, osteoarthritis, general gastrointestinal disorders, colitis, gastric ulceration, duodenal ulcers, inflammatory bowel disorders, irritable bowel syndrome, pain associated with diarrhea, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, sunburn, carditis, dermatitis, myositis, neuritis, collagen vascular diseases, inflammatory pain and associated hyperalgesia and allodyn
  • Protoxin-II variants of the invention include itch, cough and asthma.
  • mice global deletion of the SCN9A gene leads to complete insensitivity to histamine-induced itch (Gingras et al., American Pain Society Meeting Abstract 2013 and U.S. Pat. Publ. No. 2012/0185956).
  • peptide Nav1.7 blockers may have utility in the treatment of itch, which may arise from various sources, such as dermatological or inflammatory disorders; or inflammatory disorders such as renal or hepatobiliary disorders, immunological disorders, medication reactions and unknown/idiopathic conditions, including dermatitis, psoriasis, eczema, insect sting or bite.
  • Nav1.7 is also expressed in sensory nerves innervating the airways (Muroi et al., J Physiol. 2011 Dec. 1; 589(Pt 23):5663-76; Muroi et al., Am J Physiol Regul Integr Comp Physiol. 2013 Apr.
  • peptide Nav1.7 blockers may be beneficial in the treatment of cough e.g., acute or chronic cough, or cough caused by irritation from gastroesophageal reflux disease, and inflammatory diseases of the airways such as asthma and allergy-related immune responses, bronchospasm, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, and hiccups (hiccoughs, singultus).
  • Silencing Nav1.7 in vivo in nodose ganglia of guinea pigs using shRNA nearly abolished the cough reflex induced by mechanical probing (Muroi et al., Am J Physiol Regul Integr Comp Physiol. 2013 Apr. 10).
  • One aspect of the invention is a method of alleviating or treating itch, cough or asthma in a subject by administering a therapeutically effective amount of the Protoxin-II variant of the invention to a subject in need thereof for a time sufficient to alleviate the itch, cough or asthma.
  • Another aspect of the invention is a method of alleviating or treating Nav1.7-mediated itch, Nav1.7-mediated cough or Nav1.7-mediated asthma in a subject by administering a therapeutically effective amount of the Protoxin-II variant of the invention to a subject in need thereof for a time sufficient to alleviate the itch, cough or asthma.
  • Nav1.7-mediated itch as used herein refers to itch resulting at least partially from increased Nav1.7 channel activity.
  • Nav1.7-mediated cough refers to cough resulting at least partially from increased Nav1.7 channel activity.
  • Nav1.7-mediated asthma refers to asthma resulting at least partially from increased Nav1.7 channel activity.
  • Protoxin-II variants of the invention may be tested for their effect in reducing or alleviating pain and/or Nav1.7-mediated pain using animal models described herein, and models such as the rat spinal nerve ligation (SNL) model of neuropathic pain, carrageenan induced allodynia model, the Freund's complete adjuvant (CFA)-induced allodynia model, the thermal injury model, the formalin model and the Bennett Model, and other models as described in U.S. Pat. Appl. No. 2011/0124711 and U.S. Pat. No. 7,998,980.
  • Carrageenan induced allodynia and CFA-induced allodynia are models of inflammatory pain.
  • the Bennett model provides an animal model for chronic pain including post-operative pain, complex regional pain syndrome, and reflex sympathetic dystrophy.
  • any of the foregoing animal models may be used to evaluate the efficacy of Protoxin-II variants of the invention inhibitor in treating pain and/or Nav1.7-mediated pain.
  • the efficacy of the Protoxin-II variants of the invention may be compared to a no treatment or placebo control. Additionally or alternatively, efficacy may be evaluated in comparison to one or more known pain-relieving medicaments.
  • the present invention provides methods of treating Nav1.7-mediated pain using the Protoxin-II variants of the invention. It has been discovered in the pending application by the inventors (U.S. Patent Application No. 61/781,276) that administration of Nav1.7 blocking peptides are efficacious in treating and/or alleviating pain in various animal models of pain, contrary to what was disclosed and suggested in the literature.
  • peptide inhibitors of Nav1.7 have been shown to be potent and/or selective towards Nav1.7 in in vitro cell culture models using overexpressed Nav1.7 or on isolated neurons in which the blood-nerve barrier is subverted through desheathing or hypertonic saline injection, they have so far proven non-efficacious in in vivo animal models of pain, where the lack of efficacy has been reported to result from the inability of the peptides to pass the blood-nerve barrier.
  • Several publications describe lack of efficacy of Nav1.7 blocking peptides in animal models of pain or in isolated nerves.
  • Nav1.7 is expressed in the peripheral nervous system e.g., in nociceptive dorsal root ganglions (DRG), most notably in nociceptive small-diameter DRG neurons, in particular in peripheral terminals in the skin, with little representation in the brain.
  • DRG nociceptive dorsal root ganglions
  • Nav1.7 distribution e.g. sensory ending
  • physiology predispose it to a major role in transmitting painful stimuli.
  • One embodiment of the invention is a method of treating Nav1.7-mediated pain by administering a therapeutically effective amount of the Protoxin-II variant of the invention to a subject in need thereof for a time sufficient to treat the Nav1.7-mediated pain.
  • Protoxin-II variants of the invention Nav1.7 may be utilized in any therapy where it is desired to treat Nav1.7mediated pain or other disorders of sensory or sympathetic neuron dysfunction. “Treat” or “treatment” of pain is meant to include partially or completely to prevent, stop, inhibit, reduce, or delay the perception of pain.
  • the Nav1.7-mediated pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, post-operative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis, or pain associated with disease and degeneration.
  • PE primary erythemalgia
  • PEPD paroxysmal extreme pain disorder
  • osteoarthritis osteoarthritis
  • rheumatoid arthritis lumbar discectomy
  • pancreatitis fibromyalgia
  • painful diabetic neuropathy (PDN) post-herpetic neuropathy (PHN)
  • TN trige
  • Neuropathic pain includes for example painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN) or trigeminal neuralgia (TN).
  • PDN painful diabetic neuropathy
  • PPN post-herpetic neuropathy
  • TN trigeminal neuralgia
  • Other causes of neuropathic pain include spinal cord injuries, multiple sclerosis, phantom limb pain, post-stroke pain and HIV-associated pain.
  • Conditions such as chronic back pain, osteoarthritis and cancer may also result in the generation of neuropathic-related pain and thus are potentially suitable for treatment with the Protoxin-II variants of the invention.
  • the Nav1.7-mediated pain is associated with primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis or fibromyalgia.
  • PE primary erythemalgia
  • PEPD paroxysmal extreme pain disorder
  • osteoarthritis rheumatoid arthritis
  • lumbar discectomy pancreatitis or fibromyalgia.
  • the Protoxin-II variants of the invention may be conjugated to a second polypeptide to form a fusion protein.
  • fusion proteins are for example the well-known Fc fusions or fusions to human serum albumin to extend half-life of the peptide inhibitors.
  • the conjugation may be a direct conjugation via a linker, such as a glycine-serine rich linker.
  • linkers are well known in the art.
  • the Protoxin-II variants of the invention incorporating additional moieties may be compared for their Nav1.7 blocking ability and efficacy in treatment or reducing pain using well known methods and those described herein.
  • Protoxin-II variants of the invention include asthma, cough, heart-burn, itch, dermatitis, bladder instability, and Reynaud's disease.
  • the Protoxin-II variants of the invention may be formulated in a pharmaceutically acceptable vehicle or carrier.
  • One embodiment of the invention is a pharmaceutical composition comprising the isolated Protoxin-II variant of the invention and a pharmaceutically acceptable excipient.
  • a suitable vehicle or carrier may be water for injection, physiological saline solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration.
  • Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles.
  • These solutions are sterile and generally free of particulate matter, and may be sterilized by conventional, well-known sterilization techniques (e.g., filtration).
  • the compositions may contain pharmaceutically acceptable excipients as required to approximate physiological conditions, such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc.
  • Suitable vehicles and their formulation and packaging are described, for example, in Remington: The Science and Practice of Pharmacy (21st ed., Troy, D. ed., Lippincott Williams & Wilkins, Baltimore, Md. (2005) Chapters 40 and 41).
  • Protoxin-II variants of the invention may be administered by peripheral administration.
  • Peripheral administration or “administered peripherally” means introducing an agent into a subject outside of the central nervous system.
  • Peripheral administration encompasses any route of administration other than direct administration to the spine or brain.
  • Peripheral administration can be local or systemic. Local administration may be used to concentrate the therapeutic to the site of action, such as local administration to joints, spinal cord, surgical wounds, sites of injury/trauma, peripheral nerve fibers, various organs GI, urogenital, etc.) or inflamed tissues.
  • Systemic administration results in delivery of a pharmaceutical composition to essentially the entire peripheral nervous system of the subject and may also result in delivery to the central nervous system depending on the properties of the composition.
  • Routes of peripheral administration encompass, without limitation, topical administration, intravenous or other injection, and implanted mini-pumps or other extended release devices or formulations.
  • compositions of the invention include formulations involving the Protoxin-II variants of the invention in sustained- or controlled-delivery formulations. These formulations may be achieved through use of for example injectable microspheres, bio-erodible particles, microemulsions, nanoparticles, nanocapsules, macroemulsions, polymeric compounds (such as polyesters, polyamino acids, hydrogels, poly(lactic acid), polyglycolic acid or ethylene vinylacetate copolymers), beads or liposomes, hyaluronic acid or implantable drug delivery devices.
  • injectable microspheres such as polyesters, polyamino acids, hydrogels, poly(lactic acid), polyglycolic acid or ethylene vinylacetate copolymers
  • beads or liposomes such as hyaluronic acid or implantable drug delivery devices.
  • the Protoxin-II variants of the invention may be prepared for use for parenteral (subcutaneous, intramuscular or intravenous), intracerebral (intra-parenchymal), intracerebroventricular, intramuscular, intra-ocular, intra-arterial, intraportal, or intralesional routes; by sustained release systems or by implantation devices, or any other administration, particularly in the form of liquid solutions or suspensions; for buccal or sublingual administration such as in the form of tablets or capsules; or intranasally such as in form of powders, nasal drops or aerosols or certain agents; transdermally in a form of a gel, ointment, lotion, cream or dusting powder, suspension or patch delivery system with chemical enhancers to either modify the skin structure or to increase the drug concentration in the transdermal patch, or with agents that enable the application of formulations containing proteins and peptides onto the skin (Int.
  • composition also may be administered locally via implantation of a membrane, sponge or another appropriate material onto which the desired molecule has been absorbed or encapsulated.
  • the device may be implanted into any suitable tissue or organ, and delivery of the desired molecule may be via diffusion, timed-release bolus, or continuous administration.
  • the concentration of the Protoxin-II variants of the invention or other peptide inhibitors of Nav1.7 in such pharmaceutical formulation can vary widely, for example from about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1.0%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, or between 2% to 5%, up to as much as 15%, 20%, 30%, 40%, 50%, 60% or 70% by weight and will be selected primarily based on fluid volumes, viscosities and other factors, according to the particular mode of administration selected.
  • the Protoxin-II variants of the invention can be lyophilized for storage and reconstituted in a suitable vehicle prior to use. This technique has been shown to be effective with conventional protein preparations. Lyophilization and reconstitution techniques are well known in the art.
  • An exemplary pharmaceutical composition of the present invention may comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, and may further include sorbitol, sucrose, Tween-20 and/or a suitable substitute thereof.
  • An effective dose refers to an amount or dosage sufficient to produce a desired result, i.e. to partially or completely prevent, stop, inhibit, reduce, or delay the perception of pain associated with any painful medical condition.
  • the effective amount may vary depending on the specific vehicle and the Protoxin-II variants of the invention selected, and is also dependent on a variety of factors and conditions related to the subject to be treated and the severity of the pain. For example, factors such as age, weight and health of the subject to be administered with the pharmaceutical compositions of the invention as well as dose response curves and toxicity data obtained in preclinical animal work could be among those considered.
  • a determined dose may, if necessary, be repeated at appropriate time intervals selected as appropriate by a physician or other person skilled in the relevant art (e.g. nurse, veterinarian, or veterinary technician) during the treatment period.
  • a physician or other person skilled in the relevant art e.g. nurse, veterinarian, or veterinary technician
  • the determination of an effective amount or a therapeutically effective amount for a given agent is well within the ability of those skilled in the art.
  • a pharmaceutical composition of the invention for intramuscular injection could be prepared to contain 1 mL sterile buffered water, and between about 1 ng to about 100 mg, about 50 ng to about 30 mg or about 5 mg to about 25 mg of a Protoxin-II variant of the invention.
  • a pharmaceutical composition of the invention for intravenous infusion could be made up to contain about 250 ml of sterile Ringer's solution, and about 1 mg to about 30 mg or about 5 mg to about 25 mg of the Protoxin-II variants of the invention.
  • Actual methods for preparing parenterally administrable compositions are well known and are described in more detail in, for example, “Remington's Pharmaceutical Science,” 15th ed., Mack Publishing Company, Easton, Pa.
  • X 1 is G, P, A or deleted
  • X 2 is P, A or deleted
  • X 3 is S, Q, A, R or Y;
  • X 4 is Q, R, K, A or S;
  • X 5 is K, S, Q or R;
  • X 6 is M or F
  • X 7 is T, S, R, K or Q
  • X 8 is D or T
  • X 9 is S, A or R;
  • X 10 is E, R, N, K, T or Q;
  • X 11 is R or K
  • X 12 is K, Q, S or A
  • X 13 is E, Q or D
  • X 14 is G or Q
  • X 15 is V or S
  • X 16 is R or T
  • X 17 is K or R
  • polypeptide inhibits human Nav1.7 activity with an IC 50 value of about 1 ⁇ 10 ⁇ 7 M or less, wherein the IC 50 value is measured using a FLIPR® Tetra membrane depolarization assay in the presence of 25 ⁇ 10 ⁇ 6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • Protoxin-II variant as described above, wherein the N-terminal extension comprises the amino acid sequence of SEQ ID NOs: 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384 or 385.
  • the Protoxin-II variant as described above, wherein the C-terminal extension comprises the amino acid sequence of SEQ ID NOs: 374, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396 or 397.
  • X 1 is Q, R, K, A or S;
  • X 2 is K, S, Q or R;
  • X 3 is M or F
  • X 4 is T, S, R, K or Q
  • X 5 is D or T
  • X 6 is 5, A or R;
  • X 7 is E, R, N, K, T or Q;
  • X 8 is R or K
  • X 9 is K, Q, S or A
  • X 10 is E, Q or D
  • X 11 is G or Q
  • X 12 is V or S
  • X 13 is R or T
  • X 14 is K or R.
  • X 1 is Y, Q, A, S or R;
  • X 2 is T or S
  • X 3 is S, R or A
  • X 4 is E, T or N;
  • X 5 is E or Q
  • X 6 is V or S
  • X 7 is R or T
  • X 8 is K or R.
  • Protoxin-II variant as described above, comprising the amino acid sequence of SEQ ID NOs: 56, 59, 65, 78, 111, 114, 117, 118, 119, 121, 122, 123, 129, 130, 133, 150, 190, 217, 281, 324, 325 or 326.
  • X 1 is Tor S
  • X 2 is S, R or A
  • X 3 is E, T or N;
  • X 4 is E or Q
  • X 5 is V or S
  • X 6 is R or T
  • X 7 is K or R.
  • Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 78 (GPQCQKWMQTCDRERKCCEGFVCTLWCRKKLW-COH), wherein
  • the fusion protein of claim 17 wherein the half-life extending moiety is human serum albumin (HSA), albumin binding domain (ABD), Fc or polyethylene glycol (PEG).
  • HSA human serum albumin
  • ABS albumin binding domain
  • Fc polyethylene glycol
  • a method of producing a isolated Protoxin-II variant comprising culturing the host cell of as described above and recovering the Protoxin-II variant produced by the host cell.
  • a pharmaceutical composition comprising an isolated Protoxin-II variant as described above and a pharmaceutically acceptable excipient.
  • a method of treating Nav1.7-mediated pain in a subject comprising administering to a subject in need thereof an effective amount of the Protoxin-II variant as described above to treat the pain.
  • the pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, postoperative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis.
  • PE primary erythemalgia
  • PEPD paroxysmal extreme pain disorder
  • osteoarthritis osteoarthritis
  • rheumatoid arthritis rheumatoid arthritis
  • lumbar discectomy pancreatitis
  • fibromyalgia painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal
  • Protoxin-II variant for use as described above, wherein pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, postoperative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis.
  • PE primary erythemalgia
  • PEPD paroxysmal extreme pain disorder
  • osteoarthritis osteoarthritis
  • rheumatoid arthritis rheumatoid arthritis
  • lumbar discectomy pancreatitis
  • fibromyalgia painful diabetic neuropathy (PDN), post-herpetic neuropathy (
  • Protoxin-II variant for use as described above, wherein the Protoxin-II variant is administered locally to a joint, spinal cord, surgical wound, sites of injury or trauma, peripheral nerve fibers, urogenital organs, or inflamed tissues.
  • Protoxin-II single position limited amino acid scanning library substitution was designed to assess to what degree selectivity, peptide yield, and homogeneity can be improved.
  • Protoxin-II variants were designed as HRV3C protease cleavable HSA fusion proteins in the following format from N- to C-terminus: 6 ⁇ His-HSA-linker-HRV3C cleavable peptide-Protoxin-II variant (“6 ⁇ His” disclosed as SEQ ID NO: 108); linker being (GGGGSGGGGSGGGGSGGGGS; SEQ ID NO: 80, HSA having the sequence of SEQ ID NO: 106, HRV3C cleavable peptide having the sequence of SEQ ID NO: 82). Each Protoxin-II variant, after cleavage from HSA had a residual N-terminal GP from the cleavage site.
  • the variants were characterized in membrane depolarization assays using FLIPR® Tetra as described in Example 3 FLIPR® Tetra membrane depolarization assay, and in whole cell patch clamp experiments using the QPatch assay as described in Example 3.
  • Combinatorial libraries were designed to test for additive effects of select single position hits in an attempt to generate Nav1.7 antagonists with further improved potency and selectivity profile compared to the native peptide.
  • the designed Protoxin-II variant genes were generated using synthetic gene assembly technology as described in U.S. Pat. No. 6,521,427.
  • the amino acid sequences of the designed peptide variants were back-translated to DNA sequences using human high-frequency codons.
  • the DNA sequence of each variant gene, together with a portion of vector DNA including the DNA cloning sites, was synthesized as multiple oligonucleotides, some of which contained degenerate codons, and assembled into full-length DNA fragments.
  • the assembled DNA fragments were amplified by PCR and PCR products were subsequently cloned as a pool.
  • HEK 293-F cells were maintained in 293 FreestyleTM media (Invitrogen Cat #12338) and split when the cell concentration was between 1.5 and 2.0 ⁇ 10 6 cells per mL. The cells were grown in suspension, shaking at 125 RPM in a humidified incubator set at 37° C. and 8% CO 2 . HEK 293F cells were transiently transfected using a DNA/lipid complex after they were diluted to 1.0 ⁇ 10 6 cells per mL.
  • the supernatant was separated from the cells by centrifugation at 5,000 ⁇ g for 10 minutes and filtered through a 0.2 ⁇ m filter (Corning; Cat #431153), then concentrated 10 and 50 fold using an Amicon Ultra Concentrator 10K (Cat #UFC901096), and centrifuging for approximately 10 minutes at 3,750 ⁇ g.
  • Protoxin-II variants were expressed as HSA fusion proteins as indicated in Example 1 and the Protoxin-II variant peptides were cleaved with HRV3C protease prior to purification. Two methodologies were tested for efficient purification of the Protoxin-II variants.
  • the secreted proteins were purified from the expression supernatants via IMAC using 1 ml HisTrap HP columns (GE Healthcare Cat #17-5247-01).
  • the chromatography method was run using an AKTA Xpress and protein was eluted from the column using a step gradient of imidazole. Peak fractions were pooled and digested overnight with HRV 3C protease (1 ⁇ g protease/150 ⁇ g fusion).
  • Cleaved peptide-fusion pools were further purified using a Dionex HPLC system with a reverse phase Phenomenex Luna 5 ⁇ m C18(2) column (Cat #00B-4252-PO-AX). Samples were eluted from the column with a 0-68% Acetonitrile (0.05% TFA) linear gradient. Elution fractions were pooled, lyophilized overnight and reconstituted in HEPES buffered saline, pH 7.4 (10 mM HEPES, 137 mM NaC1, 5.4 mM KC1, 5 mM glucose, 2 mM CaC1 2 , 1 mM MgC1 2 ).
  • Table 4 shows yields of Protoxin-II variants purified by RP-HPLC. The average mg yield/L was 0.01615.
  • Protoxin-II Variant yield Protoxin-II Variant yield Peptide ID (mg) Peptide ID (mg) NV1D816 0.0008 NV1D2496 0.0006 NV1D2511 0.0009 NV1D2503 0.0030 NV1D2513 0.0034 NV1D766 0.0054 NV1D2504 0.0071 NV1D770 0.0040 NV1D2260 0.0129 NV1D772 0.0015 NV1D2498 0.0079 NV1D792 0.0016 NV1D2499 0.0076 NV1D815 0.0008 IW1D2512 0.0061 NV1D768 0.0060 NV1D2267 0.0095 NV1D2508 0.0017 NV1D2507 0.0000 NV1D2501 0.0008 NV1D2509 0.0000 NV1D2296 0.0018 NV1D2305 0.0001 NV1D2292 0.0059
  • the secreted proteins were purified from the expression supernatants via IMAC using 1 mL HisTrap HP columns (GE Healthcare Cat #17-5247-01).
  • the chromatography method was run using an AKTA Xpress and protein was eluted from the column using a step gradient of imidazole. Peak fractions were pooled and digested overnight with HRV3C protease (1 ⁇ g protease/150 ⁇ g fusion).
  • the cleaved sample was loaded into a 50 kDa molecular weight cut off centrifugal filter unit (Millipore UFC805096) and cleaved peptide collected in the filtrate fraction.
  • Peptide pools were loaded onto a 96-well solid phase extraction block (Agilent Bond Elut Plexa A3969030) for further purification, desalting, and concentration. Blocks were used in conjunction with a vacuum manifold (Whatman). Peptide samples were loaded and washed in 0.05% TFA in water and eluted with a step gradient of acetonitrile with 0.05% TFA in water. Elution fractions were then lyophilized overnight and reconstituted in HEPES buffered saline, pH 7.4 (10 mM HEPES, 137 mM NaC1, 5.4 mM KC1, 5 mM glucose, 2 mM CaC1 2 , 1 mM MgCl 2 ).
  • Peptides were reconstituted in supplemented HEPES buffered saline, pH 7.4 (10 mM HEPES, 137 mM NaC1, 5.4 mM KC1, 5 mM glucose, 2 mM CaC1 2 , 1 mM MgC1 2 ) and absorbance was measured at 280 nm. Concentration values were then calculated using each sample's extinction coefficient. 2 ⁇ g of each peptide were loaded onto an Invitrogen NuPAGE® Novex® Bis-Tris Gel 15 well gel and run in MES buffer non-reduced.
  • Table 5 shows yields (mg) of Protoxin-II variants purified by SPE. The average mg yield/L was 0.05353.
  • the ability of the generated peptides to inhibit membrane depolarization induced by Nav1.7 agonist veratridine (3-veratroylveracevine; Biomol, Catalog #NA125) was measured with a FRET (fluorescence resonance energy transfer) assay on FLIPR® Tetra using DISBAC2(3) (Invitrogen, K1018) as an electron acceptor and PTS18 (Trisodium 8-octadecyloxypyrene-1,3,6-trisulfonate) (Sigma) as a donor by exciting the donor at 390-420 nm and measuring FRET at 515-575 nm.
  • FRET fluorescence resonance energy transfer
  • HEK293 cells stably expressing human Nav1.7 were cultured in DMEM/F-12 media (1:1), supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 400 ⁇ g/mL geneticin and 100 ⁇ M NEAAs (all reagents from Invitrogen). 50 ⁇ L of harvested cells were plated at 25,000 cells/well into poly-lysine coated 384-well black clear bottom plates. The plates were incubated at room temperature (RT) for 15 min followed by an overnight incubation at 37° C. All incubations were done in the dark unless otherwise stated.
  • RT room temperature
  • the wells were washed 4 times with assay buffer (137 mM NaC1, 4 mM KC1, 2 mM MgC1 2 , 2 mM CaC1 2 , 5 mM Glucose, 10 mM HEPES, pH 7.4), and resuspended in 25 ⁇ L of assay buffer.
  • 2 ⁇ stock (6 ⁇ M) of the PTS18 dye was prepared by suspending the dye in 10% Pluronic F127 in DMSO at 1:1 (v/v ratio). 25 ⁇ L of the 2 ⁇ PTS18 stock was added into the wells and the cells were stained for 30 min at RT, after which the dye was washed off with the assay buffer.
  • Peptides were suspended at 3 ⁇ their final concentration in the assay buffer containing 10 ⁇ M DISBAC2(3) and 400 ⁇ M VABSC-1 to suppress background fluorescence (Sigma, cat #201987). 25 ⁇ L/well of the suspended peptides were added into each well, and incubated for 60 minutes at RT. Depolarization was induced by 25 ⁇ M final concentration of veratridine (by adding 25 ⁇ L/well of 75 ⁇ M (3 ⁇ ) stock solution), and the reduction in the mean intensity of FRET dye fluorescence was measured 30-100 seconds after adding the agonist. A 1.3 ⁇ dilution of each measured peptide occurred after adding veratridine by convention, the concentration at the beginning of the FLIPR® Tetra assay is reported.
  • Concentration-response curves of synthetic Protoxin-II were constructed in each experimental series and were used as controls. Fluorescence counts for each well were converted to % response by normalizing the signal to the difference between negative control (response to agonist veratridine alone) and positive control (response to veratridine in the presence of 10 ⁇ M tetracaine) values. For measurements, “spatial uniformity correction” (all fluorescence traces are normalized to the average initial starting intensity) and “subtract bias value” (subtract the initial starting intensity from each trace) were turned on in FLIPR® Tetra. Each data point represented the response in an individual well.
  • HEK293 cells stably expressing human Nav1.5 (SEQ ID NO: 105), Nav1.7 (SEQ ID NO: 79) or Nav1.6 (SEQ ID NO: 407) were cultured in DMEM/F-12 media (1:1), supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 400 ⁇ g/mL Geneticin and 100 ⁇ M NEAAs (all reagents from Invitrogen). Cells were maintained at 37° C. and in 5% CO 2 and assayed upon reaching ⁇ 50-90% confluency.
  • CHO cells stably expressing human Nav1.6 in a tetracycline-inducible manner were cultured in HAMs F12, supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 10 ⁇ g/mL Blasticidin and 400 ⁇ g/mL Zeocin. Cells were maintained at 37° C. and in 5% CO 2 , and assayed upon reaching ⁇ 50-90% confluency. Nav1.6 expression was induced with 1 ⁇ g/ml of tetracycline, 24-48 h prior to an experiment.
  • the voltage protocol used in the assay was as follows.
  • the first liquid application contained only the control buffer (5 ⁇ L).
  • the voltage protocol was executed 10 times (for a total duration of 10 min) five seconds after the application.
  • the next three liquid applications (5 ⁇ L each) contained a test compound (same compound at the same concentration for all three applications) or control buffer (for control cells only). Five seconds after each of these applications, the voltage protocol was again executed 10 times (also once per minute).
  • the last application contained positive (composed of three 10 ⁇ L sub-applications, each separated by 2 seconds), five seconds after which the same voltage protocol was executed twice to obtain the baseline current. Currents were sampled at 25 kHz and filtered at 5 kHz with an 8-pole Bessel filter. The series resistance compensation level was set at 80%.
  • the peak current amplitude at 0 mV for each current trace in the first four liquid applications was first subtracted from that of the last trace in the presence of positive control and then normalized to that of the last trace in the first (control buffer) application as % inhibition.
  • this (% inhibition) value for each cell in the presence of a test compound was further normalized to the average % inhibition value for control (typically 5-6) cells in the same experiment.
  • the mean of the last two such values in the last compound application i.e., the corrected % inhibition value for each concentration of a test compound
  • the % inhibition values for all cells tested at each compound concentration were averaged and used in concentration response calculations.
  • Combinatorial libraries were designed to test for additive effects of select single position hits in an attempt to generate Nav1.7 antagonists with further improved potency and selectivity profile compared to the native peptide using several approaches.
  • Protoxin-II/Huwentoxin-IV single position chimeras were designed bidirectionally. The purpose of this library was to obtain Protoxin-II variants that retained potency and selectivity profile of the wild type Protoxin-II and would achieve beneficial refolding properties associated with Huwentoxin-IV. Substitutions R22T and E12N were identified from this scan.
  • Peptide NV1G1153 was further engineered by diversifying position Y1 by a limited amino acid scan using R, K, T, A, D, E, Q and S, and by charge cluster engineering, where all sets of charged residues in the three-dimensional structure of the peptide (D10/E12, K4/E17, D10/E12/R13) were mutated.
  • N- and C-terminal extensions were introduced to select peptides, including NV1 G1153 with the purpose of improving peptide distribution to the site of action and of improving half-life of the peptides without significantly increasing the molecular weight of the resulting peptide.
  • the N- and C-terminal extensions that were used are shown in Table 8 and 9, respectively, and are described in Oi et al., Neuroscience Letters 434, 266-272, 2008; Whitney et al., Nature Biotechnology 2011 29:4, 352-356; Sockolosky et al., (2012) 109:40, 16095-16100.
  • Cell penetrating peptides HIV Tat and polyarginine were also used.
  • Various linkers were used to couple the Protoxin-II variant to the N- and/or C-terminal extensions. The linkers used are shown in Table 10.
  • Protoxin-II variants from each campaign were tested for their potency and selectivity for Nav1.7 using methods described in Example 3.
  • the amino acid sequences of the variants that inhibited Nav1.7 with an IC 50 value of 200 nM or less are shown in Table 3.
  • Table 11 shows the amino acid substitutions in select variant when compared to the wild type Protoxin-II, and the IC 50 values for Nav1.7 inhibition in the FLIPR Tetra assay.
  • FIG. 1 shows the sequence genus of generated Protoxin-II variants that inhibit Nav1.7 with an IC 50 value of 30 nM or less.
  • the amino acid sequences of the peptides shown in FIG. 2 are shown in FIG. 3 . All these peptides had W7Q and M19F substitutions when compared to the wild type Protoxin-II.
  • the Protoxin-II variants were expressed and purified as described in Example 1, or synthesized by standard solid phase synthesis methods. The yields of the recombinant or synthetic peptides were compared to the yields of the wild-type protoxin. Table 12 shows that the yields of the select Protoxin-II variants were significantly higher than that of Protoxin-II, indicating improved folding properties of the variants. The scale of the solid-phase synthesis was 0.5 mmol.
  • mice Male C57Bl/6 mice (24-26 g), ordered from Charles River and housed individually, were used for this study.
  • Von Frey Test Mechanical (tactile) threshold was assessed by Von Frey Hairs following the Up-Down method (Dixon, 1980, Chaplan et al., 1994). 7 graded stimuli (von Frey filaments: 0.03, 0.07, 0.16, 0.4, 0.6, 1, 2 g; Stoelting, Wood Dale, Ill.) were used. Von Frey hairs were presented perpendicularly against the center plantar area (between toris) on a hindpaw. Sufficient force was applied to bend the filament slightly and held for 3 seconds. Per the Chaplan paper, a positive response can be either 1) a sharp withdrawal or 2) immediate flinching upon removal of the filament. See Chaplan et al. for more details. Mice were acclimated to the wire mesh in the testing chamber for 30-60 minutes prior to testing.
  • Hargreaves Test A modified Hargreaves box was used to measure thermal paw withdrawal latency (PWL) (Hargreaves et al., 1988, Pain, 32:77-88; Dirig et al., 1997, J Neurosci. Methods, 76:183-191).
  • This box consists of a chamber with a raised glass floor maintained at a constant temperature (27° C.).
  • the thermal nociceptive stimulus originates from a projection bulb light beam below the glass surface. The light beam is aimed at the area between toris (center plantar).
  • the “start” button will turn on the light and start the timer. Movements (such as a sudden withdrawal) of the stimulated paw will trigger the switch to turn off the light and stop the timer.
  • the latency in seconds is displayed.
  • the bulb will be turned off after 20 seconds (cutoff) to prevent tissue injury.
  • the animals were allowed to habituate on the glass surface for 30-60 minutes before PWL measurement. Constant amperage was used throughout the study, which resulted in Pre-test paw withdrawal latencies between 8-12 seconds when averaged over 3 to 6 read-outs taken at least 5 minutes apart.
  • T o threshold on day( ) (post-CFA, pre-pump); T o : threshold on day 1 post pump implantation; Tc: cut-off of the test (20 s for the Hargreaves test and 2 g for the Von Frey test).
  • Hotplate Test Animals were placed on a 10′′ ⁇ 10′′ metal plate surrounded by 4 Plexiglas walls (15 inches high). The plate was maintained at a temperature of either 50 or 55° C. The response latency (time when the animal first flinches or licks its hind paw, jumps, or vocalizes) was measured and the animal removed from the plate. Animals showing no response were removed from the plate after 40 s (50° C.) or 20 s (55° C.) to prevent any possible tissue damage. This trial was repeated 2-5 times every 15-60 minutes in a day.
  • CFA Model Animals were anesthetized with isoflurane (4% induction and 2% maintenance) and 20 ⁇ L of 100% Complete Freund's Adjuvant (CFA; Sigma-Aldrich; Saint Louis, Mo.) was injected into the center plantar area on one hind paw using a 27gauge needle attached to a 50- ⁇ L Hamilton syringe.
  • CFA Complete Freund's Adjuvant
  • Carrageenan model Animals were anesthetized with isoflurane (4% induction and 2% maintenance) and 25 ⁇ L of 2% A-carrageenan (Sigma-Aldrich; Saint Louis, Mo.) dissolved in normal saline was injected into the center plantar area on hind paws using an insulin syringe (BD; Franklin Lakes, N.J.).
  • A-carrageenan Sigma-Aldrich; Saint Louis, Mo.
  • Alzet micro-osmotic mini pumps (Durect Corporation Model 1003D and 2001 D) were filled and primed per manufacturer's guide. Mice were anesthetized with isoflurane (5% induction; 2% maintenance). Their backs were shaved, wiped down with isopropyl alcohol and povidone iodine, and a small incision was made between the scapulae. Using a pair of forceps or hemostat, a small pocket was formed by spreading the subcutaneous connective tissues apart. The pump was inserted into the pocket with the flow moderator pointing away from the incision. The skin incision was then closed using 7-mm staples and the animals were allowed to recover in their home cages.
  • NV1D3034-OH NV1D3034-COOH
  • NV1 D3368-OH NV1 D3368-COOH
  • NV1 D2775-OH NV1 D2775-COOH
  • NV1 D3368-OH fully reversed CFA-induced thermal hyperalgesia at 684 and 1824 ⁇ g/day ( FIGS. 5A and 5B ).
  • NV1 D2775-OH demonstrated strong efficacy in the CFA model. Thermal latencies reached values close to the cut-off following NV1 D2775 administration ( FIGS. 6A and 6B , 1824 ⁇ g/day), suggesting a strong analgesia effect on top of the anti-hyperalgesia effect.
  • NV1 D2775-OH reversed CFA-induced tactile allodynia ( FIGS. 6C and 6D , 1824 ⁇ g/day).
  • NV1 D2775-OH The anti-hyperalgesic effect of NV1 D2775-OH was seen as early as 4 hr post-pump implantation ( FIG. 7A ).
  • the effect reached the maximum at 8 hr in both the thermal and tactile tests ( FIGS. 7A and 7B ), which was maintained at 24 hr.
  • Thermal latency and tactile threshold returned the control level by 48 h post pump implantation (approximately 24 h after the pumps were predicted to be empty) ( FIGS. 7A and 7B ).
  • NV1 D2775-OH also exhibited strong, dose-dependent efficacy in the hotplate test ( FIG. 8 ). Latencies at 50 and 55° C. reached values near cut-off following the administration of 1824 ⁇ g/day. At 228 ⁇ g/day, NV1 D2775-OH produced a modest yet significant increase in the thermal latency, compared to the PBS control.
  • NV1 D2775-OH was evaluated in another model of inflammatory pain, the carrageenan model.
  • Animals were implanted with NV1 D2775-OH or PBS pumps. Thermal withdrawal latencies were measured pre- and on day 1 post-pump. A-carrageenan was injected into the hindpaws and thermal latencies were measured again on 2, 3 and 4 hr following carrageenan.
  • NV1D2775-OH at 1824 ⁇ g/day produced significant analgesia ( FIG. 9 ). Injection of A-carrageenan in the hindpaws induced inflammation (not shown) and lowered thermal paw withdrawal latency in the Hargreaves test over the 4 hr test-period ( FIG. 9 , PBS group). Animals pretreated with NV1 D2775-OH at 1824 ⁇ g/day were fully protected from carrageenan-induced hyperalgesia.
  • Mutant peptides were expressed as recombinant fusions to human serum albumin and site-specifically enzymatically cleaved using HRV3C to generate Protoxin-II variants as described in Example 1.
  • Each Protoxin-II variant after cleavage from HSA had a residual N-terminal GP from the cleavage site.
  • IC 50 values against human Nav1.7 were measured using FLIPR Tetra or Qpatch according to the protocols described in Example 3.
  • Variants demonstrating IC 50 ⁇ 100 nM for human Nav1.7 were counter-screened for selectivity against additional hNav channels using Qpatch electrophysiology. Selective hits were identified and used in the design of combinatorial peptide libraries which were produced using both recombinant expression and solid-phase peptide synthesis. Combinatorial variants were screened using the same strategy as detailed above.
  • positions that can be mutated to improve selectivity include Gln3, Ser11, Glu12, Lys14, Glu17, Gly18, Leu29 and Trp30 (residues numbering according to SEQ ID NO: 1).
  • the solution structure of Protoxin-II was determined by NMR and is shown in FIG. 10 as a surface representation.
  • the left hand side of the Figure shows the previously described (Park et al., J. Med. Chem. 2014, 57:6623-6631) ring of Trp residues, W5/W7/W24, surrounding M6.
  • a selectivity face was identified in this study on Protoxin-II consisting of multiple amino acid positions which can be mutated to improve selectivity for hNav1.7 over other sodium channel isoforms.
  • the residues residing on the selectivity face include residues Ser11, Glu12, Lys14, Glu17, Gly18, Leu29 and Trp30 (residue numbering according to SEQ ID NO: 1).
  • the identification of the selectivity face and multiple positions within responsible for selectivity towards Nav1.7 has not been described earlier.
  • a key step in the synthetic production of Protoxin-II variants is the oxidative refolding of the linear peptide, where the disulfide pairings are formed.
  • the RP-HPLC trace for native Protoxin-II purification following refolding revealed multiple peaks at differing retention times that were of correct mass but demonstrated differing levels of activity, indicative of improper folding of the peptide.
  • the relative abundance of the RP-HPLC major peak, and therefore the relative abundance of correctly folded peptide could be improved by making substitutions at various Protoxin-II positions. Mutation of Trp7 or Trp30 improved folding of the resulting Protoxin-II variant. Mutation of both Trp7 and Trp30 in combination further improved folding of the resulting Protoxin-II variant, and could rescue folding of difficult-to-refold Protoxin-II variants.
  • Protoxin-II belongs to a family 3 of inhibitory cysteine knot peptides (Klint et al., Toxicon 60:478-491, 2012).
  • Trp7 is conserved in all family 3 members, and substitutions at this position as well as at Trp5 and Met6 in Jingzhaotoxin-V, another family 3 inhibitory cysteine knot peptide, resulted in loss in potency, indicating that hydrophobic residues at positions 5, 6 and 7 in Jingzhaotoxin-V are essential to Jingzhaotoxin-V Nav1.7 inhibitory potency (Int. Pat. Publ. No. 2014/165277). Trp5/Met6/Trp7 is also conserved in Protoxin-II, and therefore it was unexpected that polar substitutions at Trp7 can be made without loss of Protoxin-II activity with significantly improved refolding properties. Substitutions at Trp30 were shown to simultaneously improve Nav1.7 selectivity and refolding properties of the variant peptide and were unexpected since individual advantageous substitutions typically only improve a single parameter.
  • Table 14 shows the amino acid sequences of the select generated Protoxin-II variants.
  • Protoxin-II variants were expressed and purified as described in Example 1, or synthesized by standard solid phase synthesis methods. The yields of the recombinant or synthetic peptides were compared to the yields of the wild-type protoxin. Table 17 shows that the yields of the select Protoxin-II variants were significantly higher than that of Protoxin-II, indicating improved folding properties of the variants. The scale of the solid-phase synthesis was 0.1 mmol.
  • NV1D2775-OH, NV1D3034 and 63955918 were used in the studies. Animal models that measure acute thermal pain (tail flick and hot plate) and injury-induced pain (formalin flinching) were used.
  • Tail-flick test The animals were placed on a tail-flick device (Ugo Basile). The device has a focal infrared light heating area (diameter-5 mm). The tail (1 ⁇ 3-1 ⁇ 2 way from distal end) of the animal was placed on the focal heating area. The temperature of the heat source was adjusted to elicit a tail-flick within 10 seconds in animals treated with vehicle. A 15 second cut-off time was used to prevent tissue damage, as is standard in the literature. The time elapsed between the start of the heat stimulus and any avoidance response was measured automatically and recorded for the test groups.
  • Hot plate test The animal was placed on a 10′′ ⁇ 10′′ metal plate surrounded by 4 Plexiglas walls (15 inches high) and maintained at a temperature of 48-55° C. If the animal licked its hind paw, jumped, or vocalized, it was removed from the plate and the response latency was documented. If the animal did not show any response within 20-90 seconds (cut-off time), it was removed from the plate to prevent any possible tissue damage.
  • Formalin Flinching Hindpaw injection of formalin-induced pain behavior (i.e. paw flinches) was measured using an automated “flinch response” measuring device UCSD. The device detects any sudden movement of a metal band glued onto one hind paw of the animal using a motion sensor installed underneath the device floor. One-half to one hour prior to formalin injection, a small metal band was attached to the plantar surface of one hind paw using a small drop of cyanoacrylate and the animal was placed in the testing chamber to be acclimatized. The attachment of the metal band did not appear to be irritating to the animal. Formalin (2.5%, 50 ⁇ L) was injected subcutaneously into the dorsum of the paw with the metal band. The animal was placed in the customized cylinder (25 ⁇ 10 ⁇ 20 cm, San Diego Instrument) immediately after intraplantar formalin injection. Paw flinches were recorded automatically.
  • Protoxin-II variant 63955918 produced potent and prolonged analgesia as indicated by the elevated latency in the tail flick test ( FIG. 11A and FIG. 11B ) and hot plate test ( FIG. 11C , FIG. 11D ) after a single intrathecal administration.
  • the significance and duration of the analgesia was dose-dependent.
  • Hindpaw formalin injection is a commonly used model for injury-induced pain. The injection induces a characteristic, bi-phasic flinching behavior, which indicates pain in test animals. As shown in FIG. 11E , animals pretreated with intrathecal injection of Protoxin-II variant 63955918 demonstrated fewer flinches in the formalin test, suggesting an inhibition of injury-induced pain.
  • peptides NV1 D2775-OH and NV1D3034 demonstrated significant efficacy in the tail flick, hot plate and formalin test ( FIG. 12A , FIG. 12B , FIG. 12C , FIG. 12D , FIG. 12E , FIG. 13A , FIG. 13B , FIG. 13C , FIG. 13D , FIG. 13E ) following a single intrathecal administration.

Abstract

The present invention relates to Protoxin-II variants, polynucleotides encoding them, and methods of making and using the foregoing.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional application of co-pending application Ser. No. 15/583,793 filed May 1, 2017, which was a continuation of application Ser. No. 15/060,158 filed Mar. 3, 2016, which claimed the benefit of priority under 35 U.S.C. § 119(e) to U.S. Provisional Application 62/127,339, filed Mar. 3, 2015, the disclosure of each are herein incorporated by reference in their entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to Protoxin-II variants, synthetic polynucleotides encoding them, and methods of making and using the foregoing.
  • BACKGROUND OF THE INVENTION
  • Voltage-gated sodium channels (VGSC) are present in all excitable cells including cardiac and skeletal muscle cells and central and peripheral neurons. In neuronal cells, sodium channels are responsible for amplifying sub-threshold depolarizations and generating the rapid upstroke of the action potential. As such, sodium channels are essential to the initiation and propagation of electrical signals in the nervous system. Aberrant sodium channel function is thought to underlie a variety of medical disorders (Hubner and Jentsch, Hum Mol Genet 11:2435-2445, 2002), including epilepsy (Yogeeswari et al., Curr Drug Targets 5:589-602, 2004), arrhythmia (Tfelt-Hansen et al., J Cardiovasc Electrophysiol 21:107-115, 2010), myotonia (Cannon and Bean, J Clin Invest 120:80-83, 2010), and pain (Cregg et al., J Physiol 588:1897-1904, 2010). Sodium channels are typically a complex of various subunits, the principal one being the pore-forming alpha-subunit, which is alone sufficient for function.
  • Nine known members of the family of voltage-gated sodium channel alpha subunits exist in humans, Nav1.1-Nav1.9. The Navl.x subfamily can be pharmacologically subdivided into two groups, the tetrodotoxin (TTX)-sensitive and TTX-resistant. Nav1.7, (a.k.a. PN1 or hNE) is encoded by the SCN9A gene, is TTX-sensitive and is primarily expressed in peripheral sympathetic and sensory neurons. Nav1.7 accumulates at nerve fiber endings and amplifies small subthreshold depolarizations and acts as a threshold channel that regulates excitability.
  • Nav1.7 function is implicated in various pain states, including acute, inflammatory and/or neuropathic pain. In man, gain of function mutations of Nav1.7 have been linked to primary erythemalgia (PE), a disease characterized by burning pain and inflammation of the extremities (Yang et al., J Med Genet 41:171-174, 2004), and paroxysmal extreme pain disorder (PEPD)(Fertleman et al., Neuron 52:767-774, 2006). Consistent with this observation, non-selective sodium channel blockers lidocaine, mexiletine and carbamazepine can provide symptomatic relief in these painful disorders (Legroux-Crespel et al., Ann Dermatol Venereol 130:429-433, 2003; Fertleman et al., Neuron 52:767-774, 2006).
  • Loss-of-function mutations of Nav1.7 in humans cause congenital indifference to pain (CIP), a rare autosomal recessive disorder characterized by a complete indifference or insensitivity to painful stimuli (Cox et al., Nature 444:894-898, 2006; Goldberg et al, Clin Genet 71:311-319, 2007; Ahmad et al., Hum Mol Genet 16:2114-2121, 2007).
  • Single nucleotide polymorphisms in the coding region of SCN9A have been associated with increased nociceptor excitability and pain sensitivity. For example, a polymorphism rs6746030 resulting in R1150W substitution in human Nav1.7 has been associated with osteoarthritis pain, lumbar discectomy pain, phantom pain, and pancreatitis pain (Reimann et al., Proc Natl Acad Sci USA 107:5148-5153, 2010). DRG neurons expressing the R1150W mutant Nav1.7 display increased firing frequency in response to depolarization (Estacion et al., Ann Neurol 66:862-866, 2009). A disabling form of fibromyalgia has been associated with SCN9A sodium channel polymorphism rs6754031, indicating that some patients with severe fibromyalgia may have a dorsal root ganglia sodium channelopathy (Vargas-Alarcon et al., BMC Musculoskelet Disord 13:23, 2012).
  • In mice, deletion of the SCN9A gene in nociceptive neurons leads to reduction in mechanical and thermal pain thresholds and reduction or abolition of inflammatory pain responses (Nassar et al., Proc Natl Acad Sci USA 101:12706-12711, 2004). Ablating SCN9A in all sensory neurons abolished mechanical pain, inflammatory pain and reflex withdrawal responses to heat. Deleting SCN9A in both sensory and sympathetic neurons abolished mechanical, thermal and neuropathic pain, and recapitulated the pain-free phenotype seen in humans with Nav1.7 loss-of-function mutations (Minett et al., Nat Commun 3:791, 2012). Nav1.7 inhibitors or blockers may therefore be useful in the treatment of a wide range of pain associated with various disorders.
  • Spider venoms are known to contain a large number of sodium channel blocking peptides, including Huwentoxin-IV (HwTx-IV) (Peng et al., J Biol Chem 277:47564-47571, 2002), Protoxin-1, Protoxin-II (Middleton et al., Biochemistry 41:14734-14747, 2002) and Phrixotoxin-III (Bosmans et al., Mol Pharmacol 69:419-429, 2006). There is a need for identification of additional Nav1.7 blockers for treatment of a wide range of pain indications. In particular, there is a need for new Nav1.7 blockers with selectivity for Nav1.7 over other voltage gated sodium channel isoforms.
  • SUMMARY OF THE INVENTION
  • One embodiment of the invention is an isolated Protoxin-II variant, wherein the Protoxin-II variant inhibits human Nav1.7 activity with an IC50 value of about 1×10−7 M or less, about 1×10−8 M or less, about 1×10−9 M or less, about 1×10−10 M or less, about 1×10−11 M or less, or about 1×10−12 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7, wherein the Protoxin-II variant has a W7Q and/or a W30L substitution.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence of SEQ ID NOs: 30, 40, 44, 52, 56, 59, 65, 78, 109, 110, 111, 114, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 162, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 177, 178, 179, 180, 182, 183, 184, 185, 186, 189, 190, 193, 195, 197, 199, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 224, 226, 227, 231, 232, 243, 244, 245, 247, 249, 252, 255, 258, 261, 263, 264, 265, 266, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 332, 334, 335, 336, 337, 339, 340, 341, 342, 346, 351, 358, 359, 364, 366, 367, 368, 369, 370, 371, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, or 431.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKWMQTCDSERKCCEGMVCRLWCKKKLL-COOH); wherein the amino acid sequence has Q at position 7 and L at position 30, when residue numbering is according to SEQ ID NO: 1; and the polypeptide inhibits human Nav1.7 activity with an IC50 value of about 30×10−9 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • Another embodiment of the invention is an isolated fusion protein comprising the Protoxin-II variant of the invention conjugated to a half-life extending moiety.
  • Another embodiment of the invention is an isolated polynucleotide encoding the Protoxin-II variant of the invention.
  • Another embodiment of the invention is an vector comprising the isolated polynucleotide of the invention. Another embodiment of the invention is a host cell comprising the vector of the invention.
  • Another embodiment of the invention is a method of producing the isolated Protoxin-II variant of the invention, comprising culturing the host cell of the invention and recovering the Protoxin-II variant produced by the host cell.
  • Another embodiment of the invention is a pharmaceutical composition comprising the isolated Protoxin-II variant or fusion protein of the invention and a pharmaceutically acceptable excipient.
  • Another embodiment of the invention is a method of treating Nav1.7-mediated pain in a subject, comprising administering to a subject in need thereof an effective amount of the Protoxin-II variant or the fusion protein of the invention to treat the pain.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows the genus amino acid sequence of Protoxin-II variants that inhibit Nav1.7 with an IC50 value of 30 nM or less in a FLIPR Tetra assay. Residue numbering is according to wild-type Protoxin-II of SEQ ID NO: 1. Genus SEQ ID NO: 403.
  • FIGS. 2A and 2B show the IC50 values for Nav1.7 and Nav1.6 inhibition in a QPatch assay, and selectivity of each variant calculated by ratio of IC50(Nav1.6)/IC50(Nav1.7) obtained in QPatch assay. SE: standard error.
  • FIGS. 3A, 3B and 3C show the sequences and the genus sequence of Protoxin-II variants that inhibit Nav1.7 with an ICs value of 30 nM or less in a FLIPR Tetra assay, and are over 30-fold selective over Nav1.6. Selectivity of each variant was calculated by ratio of IC50(Nav1.6)/IC50(Nav1.7) obtained in QPatch assay. Residue numbering is according to wild-type Protoxin-II of SEQ ID NO: 1.
  • FIG. 4A shows efficacy of NV1D3034 (NV1D3034-OH) (SEQ ID NO: 78) against CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1). ***P<0.001 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 4B shows efficacy of NV1D3034 (NV1D3034-OH) (SEQ ID NO: 78) in CFA-induced thermal hyperalgesia expressed as percent MPE (maximum possible effect) (MPE %) at each dose on day 1 following peptide administration. *P<0.05 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 5A shows efficacy of NV1 D3368 (NV1 D3368-OH) (SEQ ID NO: 198) against CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1). **P<0.01 and ****P<0.0001 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 5B shows efficacy of NV1 D3368 (NV1 D3368-OH) (SEQ ID NO: 198) in CFA-induced thermal hyperalgesia expressed as percent MPE (MPE %) at each dose on day 1 following peptide administration. *P<0.05 and **P<0.01 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 6A shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) against CFA-induced thermal hyperalgesia assessed by measurement of paw withdrawal latency in the Hargreaves test before (pre-CFA) and after CFA injection (0) and 1-day after peptide administration (1). ****P<0.0001 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 6B shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) in CFA-induced thermal hyperalgesia expressed as percent MPE (MPE %) at each dose on day 1 following peptide administration. ***P<0.001 and ****P<0.0001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 6C shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) against CFA-induced tactile allodynia. Tactile thresholds of hind paw before (pre-CFA) and after CFA (0) and 1-day after peptide administration (1). ****P<0.0001 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 6D shows efficacy of NV1 D2775-OH (SEQ ID NO: 56) against CFA-induced tactile allodynia expressed as percent MPE (MPE %) on day 1 following peptide. ***P<0.001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 7A shows time course of NV1 D2775-OH mediated reversal of thermal hyperalgesia in the CFA model as assessed by measurement of paw withdrawal latency in the Hargreaves test before and after CFA and at various time points post-peptide administration. **P<0.01 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison. Shaded areas indicate compound delivery period (0-24 hr).
  • FIG. 7B shows time course of NV1 D2775-OH mediated reversal of tactile allodynia in the CFA model as assessed by measurement of tactile threshold before and after CFA and at various time points post-peptide administration. **P<0.01 vs. PBS, two-way ANOVA followed by Bonferroni's multiple comparison. Shaded areas indicate compound delivery period (0-24 hr).
  • FIG. 8 shows that NV1 D2775-OH produced significant analgesia in the hotplate test. Thermal withdrawal latency was evaluated at 50 and 55° C. pre- and post-pump implantation. Pump implantation had no impact on the latency in the control PBS group. One day after pump, NV1 D2775-OH treated-mice exhibited prolonged latency compared to the PBS group. *P<0.05 and ****P<0.0001 vs. PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 9 shows that NV1 D2775-OH pretreatment protected animals from carrageenan-induced thermal hyperalgesia. Paw withdrawal latencies were measured pre- and on day 1 post-pump before intraplantar carrageenan injection. Latencies were measured again at 2, 3 and 4 hr following carrageenan.
  • FIG. 10 shows the surface representation of the NMR structure of the wild type Protoxin-II. A hydrophobic face shown on left includes residues W5, M6, W7, L23 and W24. A selectivity face is shown on the right and includes residues S11, E12, K14, E17, G18, L29 and W30. Residue numbering according to SEQ ID NO: 1.
  • FIG. 11A shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) after a single intrathecal (IT) administration in the tail flick test. Tail withdrawal latency to a thermal stimulus was measured at the indicated time post-peptide administration.
  • FIG. 11B shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) in the tail flick test expressed as percent area under the curve (AUC %) in the first 120 min after a single intrathecal (IT) administration. ***P<0.001 and ****P<0.0001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 11C shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) after a single intrathecal (IT) administration in the hot plate test (52.5° C.). The latency of a nociceptive response on a hot plate was measured at the indicated time post-peptide administration.
  • FIG. 11D shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) in the hot plate test expressed as percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration. ***P<0.001 and ****P<0.0001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 11E shows efficacy of the Protoxin-II variant 63955918 SEQ ID NO: 422) in the formalin test. Injection of formalin into the rat hindpaw induced a bi-phasic flinching behavior. Total number of flinches in Phase I (0-10 min post formalin) and Phase II (11-60 min post formalin) was measured by an automated device. No statistics were performed in E) due to small group size.
  • FIG. 12A shows efficacy of NV1 D2775-OH after a single intrathecal (IT) administration in the tail flick test. Tail withdrawal latency to a thermal stimulus was measured at the indicated time post-peptide administration.
  • FIG. 12B shows efficacy of NV1 D2775-OH in the tail flick test expressed as percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration. *P<0.05 and **P<0.01 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 12C shows efficacy of NV1 D2775-OH after a single intrathecal (IT) administration in the hot plate test (52.5° C.). The latency of a nociceptive response on a hot plate was measured at the indicated time post-peptide administration.
  • FIG. 12D shows efficacy of NV1 D2775-OH in the hot plate test expressed as percent area under the curve (AUC %) in the first 120 min after a single intrathecal (IT) administration. **P<0.01 and ****P<0.0001 vs PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 12E shows efficacy of NV1 D2775-OH in the formalin test. Injection of formalin into the rat hindpaw induced a bi-phasic flinching behavior. Total number of flinches in Phase I (0-10 min post formalin) and Phase II (11-60 min post formalin) was measured by an automated device. **P<0.01 vs PBS, phase I, *P<0.05 vs PBS, phase II, one-way ANOVA followed by Bonferroni's multiple comparison.
  • FIG. 13A shows efficacy of NV1D3034-OH after a single intrathecal (IT) administration in the tail flick test. Tail withdrawal latency to a thermal stimulus was measured at the indicated time post-peptide administration.
  • FIG. 13B shows efficacy of NV1D3034-OH in the tail flick test expressed as percent area under the curve(AUC %) in the first 120 min after a single intrathecal (IT) administration. ***P<0.005 vs PBS, t-test.
  • FIG. 13C shows efficacy of NV1D3034-OH after a single intrathecal (IT) administration in the hot plate test (52.5° C.). The latency of a nociceptive response on a hot plate was measured at the indicated time post-peptide administration.
  • FIG. 13D shows efficacy of NV1D3034-OH in the hot plate test expressed as percent area under the curve (AUC %) in the first 120 min after a single intrathecal (IT) administration. **P<0.01 vs PBS, t-test.
  • FIG. 13E shows efficacy of NV1D3034-OH in the formalin test. Injection of formalin into the rat hindpaw induced a bi-phasic flinching behavior. Total number of flinches in Phase I (0-10 min post formalin) and Phase II (11-60 min post formalin) was measured by an automated device. *P<0.05 vs PBS, phase I, **P<0.01 vs PBS, phase II, t-test.
  • DETAILED DESCRIPTION OF THE INVENTION
  • All publications, including but not limited to patents and patent applications, cited in this specification are herein incorporated by reference as though fully set forth.
  • As used herein and in the claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly dictates otherwise.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which an invention belongs. Although any compositions and methods similar or equivalent to those described herein can be used in the practice or testing of the invention, exemplary compositions and methods are described herein.
  • The term “polypeptide” means a molecule that comprises at least two amino acid residues linked by a peptide bond to form a polypeptide. Small polypeptides of less than 50 amino acids may be referred to as “peptides.” Polypeptides may also be referred as “proteins.”
  • The term “polynucleotide” means a molecule comprising a chain of nucleotides covalently linked by a sugar-phosphate backbone or other equivalent covalent chemistry. Double and single-stranded DNAs and RNAs are typical examples of polynucleotides.
  • The term “complementary sequence” means a second isolated polynucleotide sequence that is antiparallel to a first isolated polynucleotide sequence and that comprises nucleotides complementary to the nucleotides in the first polynucleotide sequence.
  • The term “vector” means a non-natural polynucleotide capable of being duplicated within a biological system or that can be moved between such systems. Vector polynucleotides typically contain a cDNA encoding a protein of interest and additional elements, such as origins of replication, polyadenylation signal or selection markers, that function to facilitate the duplication or maintenance of these polynucleotides in a biological system. Examples of such biological systems may include a cell, virus, animal, plant, and reconstituted biological systems utilizing biological components capable of duplicating a vector. The polynucleotide comprising a vector may be DNA or RNA molecules or a hybrid of these.
  • The term “expression vector” means a vector that can be utilized in a biological system or a reconstituted biological system to direct the translation of a polypeptide encoded by a polynucleotide sequence present in the expression vector.
  • The term “variant” as used herein refers to a polypeptide or a polynucleotide that differs from wild type Protoxin-II polypeptide of SEQ ID NO: 1 or the polynucleotide encoding the wild type Protoxin-II having the sequence of SEQ ID NO: 107 by one or more modifications for example, substitutions, insertions or deletions of nucleotides or amino acids.
  • Throughout the specification, residues that are substituted in the Protoxin-II variants are numbered corresponding to their position in the wild-type Protoxin-II of SEQ ID NO: 1. For example, “Y1A” in the specification refers to the substitution of tyrosine at residue position that corresponds to the position 1 in the wild type Protoxin-II of SEQ ID NO:1 with alanine.
  • “Complementary DNA” or “cDNA” refers to a well-known synthetic polynucleotide that shares the arrangement of sequence elements found in native mature mRNA species with contiguous exons, with the intervening introns present in genomic DNA are removed. The codons encoding the initiator methionine may or may not be present in cDNA. cDNA may be synthesized for example by reverse transcription or synthetic gene assembly.
  • “Synthetic” or “non-natural” as used herein refers to a polynucleotide or a polypeptide molecule not present in nature.
  • “Nav1.7” (also referred to as hNE or PN1) or “hNav1.7” as used herein refers to the well-known human sodium channel protein type 9 subunit alpha having a sequence shown in GenBank accession number NP 002968.1 and in SEQ ID NO: 79.
  • The term “wild type Protoxin-II” or “wild type ProTx-II” as used herein refers to the tarantula Thrixopelma pruriens (Peruvian green velvet tarantula) toxin peptide having the amino acid sequence YCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH (SEQ ID NO: 1) as described in Middleton et al., Biochemistry 41(50):14734-47, 2002.
  • The term “recombinant Protoxin-II” or “recombinant ProTx-II” as used herein refers to the recombinant Protoxin-II obtained from expression and subsequent cleavage of a Protoxin-II fusion protein having the sequence of GPYCQKWMWTCDSERKCCEGMVCRLWCKKKLW-OH as shown in SEQ ID NO: 2. Recombinant Protoxin-II incorporates a two amino acid N-terminal extension (residues G and P) when compared to the wild type Protoxin-II.
  • “Blocks human Nav1.7 activity” or “inhibits human Nav1.7 activity” as used herein refers to the ability of the Protoxin-II variant of the invention to reduce membrane depolarization induced by veratridine (3-veratroylveracevine) with an IC50 value of about 1×10−7 M or less in a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET), where veratridine-induced depolarization is measured as a reduction in FRET signal using DISBAC2(3) ([bis-(1,3-diethylthiobarbituric acid) trimethine oxonol]) as an acceptor and PTS18 (trisodium 8-octadecyloxypyrene-1,3,6-trisulfonate) as a donor by exciting the donor at 390-420 nm and measuring FRET at 515-575 nm in a cell line stably expressing human Nav1.7.
  • “FLIPR® Tetra membrane depolarization assay” as used herein is the assay described in Example 3.
  • The term “substantially identical” as used herein means that the two Protoxin-II variant amino acid sequences being compared are identical or have “insubstantial differences”. Insubstantial differences are substitutions of 1, 2, 3, 4, 5, 6, or 7 amino acids in the Protoxin-II variant amino acid sequence that do not adversely affect peptide properties. Amino acid sequences substantially identical to the Protoxin-II variants disclosed herein are within the scope of the application. In some embodiments, the sequence identity can be about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or higher. Percent identity can be determined for example by pairwise alignment using the default settings of the AlignX module of Vector NTI v.9.0.0 (Invitrogen, Carslbad, Calif.). The protein sequences of the present invention may be used as a query sequence to perform a search against public or patent databases, for example, to identify related sequences. Exemplary programs used to perform such searches are the XBLAST or BLASTP programs (http //www ncbi nlm/nih gov), or the GenomeQuest™ (GenomeQuest, Westborough, Mass.) suite using the default settings.
  • Conventional one and three-letter amino acid codes are used herein as shown in Table 1.
  • TABLE 1
    Amino acid Three letter code One letter code
    Alanine Ala A
    Arginine Arg R
    Asparagine Asn N
    Aspartate Asp D
    Cysteine Cys C
    Glutamate Glu E
    Glutamine Gln Q
    Glycine Gly G
    Histidine His H
    Isoleucine Ile I
    Leucine Leu L
    Lysine Lys K
    Methionine Met M
    Phenylalanine Phe F
    Proline Pro P
    Serine Ser S
    Threonine Thr T
    Tryptophan Trp W
    Tyrosine Tyr Y
    Valine Val V
  • The present invention provides isolated Protoxin-II (ProTx-II) variant polypeptides that inhibit human Nav1.7 activity, polynucleotides encoding them, vectors, host cells, and methods of using the polynucleotides and polypeptides of the invention. The polypeptides of the invention inhibit depolarization resulting from Nav1.7 activation, and therefore may be useful in the treatment of various conditions associated with pain and conditions associated with sensory or sympathetic neuron dysfunction.
  • The variants of the invention are potent inhibitors of Nav1.7. The current invention is based, at least in part, on the finding that certain residue substitutions in Protoxin-II enhance selectivity, synthetic yield and/or homogeneity without adversely affecting the potency of the generated Protoxin-II variants, specifically W7 and M19, and additionally residues Y1 and S11, and further additionally residues E12, R22 and (residue numbering according to SEQ ID NO: 1). For example, substitutions at positions W7 and W30 enhance the Protoxin-II variant folding and improve yield. Substitutions at positions S11, E12, K14, E17, G18, L29 and W30 improve selectivity of the resulting Protoxin-II variants to Nav1.7.
  • One embodiment of the invention is an isolated Protoxin-II variant, wherein the Protoxin-II variant inhibits human Nav1.7 activity with an IC50 value of about 1×10−7 M or less, about 1×10−8 M or less, about 1×10−9 M or less, about 1×10−10 M or less, about 1×10−11 M or less, or about 1×10−12 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−9 M 3veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • One embodiment of the invention is an isolated Protoxin-II variant, wherein the Protoxin-II variant inhibits human Nav1.7 activity with an ICs value of about 1×10−7 M or less, about 1×10−8 M or less, about 1×10−9 M or less, about 1×10−10 M or less, about 1×10−11 M or less, or about 1×10−12 M or less, wherein the ICs value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7, wherein the Protoxin-II variant has a W7Q and a W30L substitution.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the sequence X1X2X3CX4X5WX6QX7CX3X9X10X11X12CCX13X14X15X16CX17LWCX13KKLX19 (SEQ ID NO: 432), wherein
  • X1 is G, P, A or deleted;
  • X2 is P, A or deleted;
  • X3 is S, Q, A, R or Y;
  • X4 is Q, R, K, A, S or Y;
  • X5 is K, S, Q or R;
  • X6 is M or F;
  • X7 is T, S, R, K or Q;
  • X8 is D, T, or asparagyl-4-aminobutane;
  • X9 is S, A, R, I or V;
  • X10 is E, R, N, K, T, Q, Y or glutamyl-4-aminobutane;
  • X11 is R or K;
  • X12 is K, Q, S, A or F;
  • X13 is E, Q, D, L, N, or glutamyl-4-aminobutane;
  • X14 is G, Q or P;
  • X15 is M;
  • X16 is V or S;
  • X17 is R, T or N-omega methyl-L-arginine; and
  • X18 is K or R; and
  • X19 is W or L,
  • optionally having an N-terminal extension or a C-terminal extension, wherein the polypeptide inhibits human Nav1.7 activity with an IC50 value of about 1×10−7 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7. Substitutions at Protoxin-II positions W7Q and W30L improve refolding and yield of the resulting Protoxin-II variant.
  • In some embodiments, the N-terminal extension comprises the amino acid sequences of SEQ ID NOs: 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384 or 385.
  • In some embodiments, the C-terminal extension comprises the amino acid sequence of SEQ ID NOs: 374, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396 or 397.
  • In some embodiments, the N-terminal and/or the C-terminal extension is conjugated to the Protoxin-II variant via a linker.
  • In some embodiments, the linker comprises the amino acid sequence of SEQ ID NOs: 383, 392, 398, 399, 400, 401 or 402.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the sequence X1X2X3CX4X5WX6QX7CX8X9X10X11X12CCX13X14FX15CX16LWCX17KKLW (SEQ ID: 403), wherein
  • X1 is G, P, A or deleted;
  • X2 is P, A or deleted;
  • X3 is S, Q, A, R, or Y;
  • X4 is Q, R, K, A, or S;
  • X5 is K, S, Q or R;
  • X6 is M;
  • X7 is T, S, R, K or Q;
  • X8 is D, S or T;
  • X9 is S, A or R;
  • X10 is E, R, N, K, T or Q;
  • X11 is R or K;
  • X12 is K, Q, S, R or A;
  • X13 is E, Q, or D;
  • X14 is G or Q;
  • X15 is V or S;
  • X16 is R or T; and
  • X17 is K or R;
  • wherein the core amino acid sequence inhibits human Nav1.7 activity with an IC50 value of about 30×10−9 or less, wherein the IC50 value is measured using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratrolyveracevine in HEK293 cells stably expressing human Nav1.7 and using bis-1,3(diethylthiobarbituric acid) trimethine oxonol as an electron acceptor and trisodium 8-octadecylopxypyrene-1,3,6-trisulfonate as a donor by exciting the donor at 390-420 nm and measuring FRET at 515-575 nm,
    optionally having an N-terminal extension or a C-terminal extension,
    wherein the polypeptide inhibits human Nav1.7 activity with an ICs value of about 1×10−7 M or less, wherein the ICs value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • The Protoxin-II variants of the invention are potent Nav1.7 inhibitors. Recombinant Protoxin-II (SEQ ID NO: 2) has an ICs value of about 4×10−9 M for human Nav1.7 in a veratridine-induced depolarization inhibition assay measuring decline in FRET (fluorescence resonance energy transfer) in cells stably expressing Nav1.7 using FLIPR® Tetra instrument (Molecular Devices) using experimental details described in Example 3. A Protoxin-II variant is a “potent” Nav1.7 inhibitor when the ICs value in the assay described above and in Experiment 3 is about 30×10−9 M or less i.e., within 10-fold of recombinant Protoxin-II. For clarity, an ICs of 30×10−9 M is identical to ICs of 3.0×10−8 M.
  • The Protoxin-II variant polypeptides of the invention may be produced by chemical synthesis, such as solid phase peptide synthesis, on an automated peptide synthesizer. Alternatively, the polypeptides of the invention may be obtained from polynucleotides encoding the polypeptides by the use of cell-free expression systems such as reticulocyte lysate based expression systems, or by recombinant expression systems. Those skilled in the art will recognize other techniques for obtaining the polypeptides of the invention. In an exemplary method, the Protoxin-II variants of the invention are generated by expressing them as human serum albumin (HSA) fusion proteins utilizing a glycine-rich linker such as (GGGGS)4 (SEQ ID NO:80) or (GGGGS)6 (SEQ ID NO: 81) coupled to a protease cleavable linker such as a recognition sequence for HRV3C protease (Recombinant type 14 3C protease from human rhinovirus) LEVLFQGP (HRV3C linker) (SEQ ID NO: 82), and cleaving the expressed fusion proteins with the HRV3C protease to release the recombinant Protoxin-II variant peptides. Hexahistidine (SEQ ID NO: 108) or other tags may be used to facilitate purification using well known methods.
  • Protoxin-II variants of the invention may be purified using methods described herein. In an exemplary method, Protoxin-II variants of the invention expressed as HSA fusion proteins and cleaved with HRV3C protease may be purified using sold phase extraction (SPE) as described herein.
  • Generation of the Protoxin-II variants optionally having N-terminal and/or C-terminal extensions, and Protoxin-II variant fusion proteins is typically achieved at the nucleic acid level. The polynucleotides may be synthesized using chemical gene synthesis according to methods described in U.S. Pat. Nos. 6,521,427 and 6,670,127, utilizing degenerate oligonucleotides to generate the desired variants, or by standard PCR cloning and mutagenesis. Libraries of variants may be generated by standard cloning techniques to clone the polynucleotides encoding the Protoxin-II variants into the vector for expression.
  • The Protoxin-II variants may incorporate additional N- and/or C-terminal amino acids when compared to the wild type Protoxin-II of SEQ ID NO: 1, for example resulting from cloning and/or expression schemes. For example, cleavage from HSA after expression of the variant as HSA-linker-HRV3C cleavable peptide-Protoxin-II variant fusion protein may result in the incorporation of additional two residues to the N-terminus of each Protoxin-II variant, such as G and P.
  • The Protoxin-II variants of the invention are tested for their ability to inhibit human Nav1.7 using methods described herein. An exemplary assay is a veratridine-induced depolarization inhibition assay measuring decline in FRET (fluorescence resonance energy transfer) in cells stably expressing Nav1.7. Another exemplary assay employs electrophysiological recordings to measure changes in Nav1.7-mediated currents using well known patch clamp techniques and as described herein.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence of SEQ ID NOs: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 109, 110, 112, 113, 114, 115, 116, 117, 118, 119, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, or 431.
  • The Protoxin-II variants of the invention may inhibit human Nav1.7 with an IC50 value of about 1×10−7 M or less, about 1×10−8 M about 1×10−9 or less, about 1×10−10 M or less, about 1×10−11 M or less, or about 1×10−12 M or less. Exemplary variants demonstrating the range of IC50 values are variants having amino acid sequences shown in SEQ ID NOs: 30, 40, 44, 52, 56, 59, 65, 78, 109, 110, 111, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 162, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 177, 178, 179, 180, 182, 183, 184, 185, 186, 189, 190, 193, 195, 179, 199, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 224, 226, 227, 231, 232, 243, 244, 245, 247, 249, 252, 255, 258, 261, 263, 264, 265, 266, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 332, 334, 335, 336, 337, 339, 340, 341, 342, 346, 351, 358, 359, 364, 366, 367, 368, 408, 409, 410, 411, 412, 413, 414, 315, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, or 431.
  • Table 2, Table 3 and Table 14 show the sequences of select Protoxin-II variants.
  • TABLE 2
    Protoxin-II
    variant SEQ
    Protein peptide ID
    name name NO: Protein amino acid sequence
    Wild type 2 YCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D12 12 GPYCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D748 3 GPACQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D751 4 GPQCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D2292 5 GPRCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D750 6 GPSCQKWMWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D1328 7 GPYCQKWFWTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D774 8 GPYCQKWMQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D786 9 GPYCQKWMWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1D2300 10 GPYCQKWMWTCDRERKCCEGMVCRLWCKKKLW-COOH
    NV1D791 11 GPYCQKWMWTCDSKRKCCEGMVCRLWCKKKLW-COOH
    NV1D1332 12 GPYCQKWMWTCDSNRKCCEGMVCRLWCKKKLW-COOH
    NV1D2512 13 GPYCQKWMWTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1D1336 14 GPYCQKWMWTCDSERKCCEGLVCRLWCKKKLW-COOH
    NV1D1337 15 GPYCQKWMWTCDSERKCCEGMVCTLWCKKKLW-COOH
    NV1D2308 16 GPYCQKWMWTCDSERKCCEGMVCRLWCRKKLW-COOH
    NV1G953 NV1D2670 17 GPACQKWMQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1G951 NV1D2674 18 GPACQKWMWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G909 NV1D2664 19 GPACQKWMWTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1G963 NV1D2671 20 GPQCQKWMQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1G949 NV1D2675 21 GPQCQKWMWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G977 NV1D2665 22 GPQCQKWMWTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1G957 NV1D2668 23 GPRCQKWMQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1G965 NV1D2672 24 GPRCQKWMWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G973 NV1D2662 25 GPRCQKWMWTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1G975 NV1D2669 26 GPSCQKWMQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1G971 NV1D2673 27 GPSCQKWMWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G995 NV1D2663 28 GPSCQKWMWTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1G961 NV1D2676 29 GPYCQKWMQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G911 NV1D2666 30 GPYCQKWMQTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1D2816 31 GPACQKWFQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1G905 NV1D2735 32 GPACQKWMQTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1G919 NV1D2739 33 GPACQKWMWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1G979 NV1D2731 34 GPACQKWMQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1D2810 35 GPQCQKWFQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1G1099 NV1D2732 36 GPQCQKWMQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G1011 NV1D2740 37 GPQCQKWMWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2819 38 GPRCQKWFWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G1105 NV1D2729 39 GPRCQKWMQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G1013 NV1D2733 40 GPRCQKWMQTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1D2814 41 GPSCQKWFQTCDSERKCCEGMVCRLWCKKKLW-COOH
    NV1D2820 42 GPSCQKWFWTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G983 NV1D2730 43 GPSCQKWMQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G1003 NV1D2734 44 GPSCQKWMQTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1G1009 NV1D2738 45 GPSCQKWMWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2851 46 GPYCQKWFKTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1D2850 47 GPYCQKWFQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1G987 NV1D2667 48 GPYCQKWMWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2867 49 GPACQKWFQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1D2881 50 GPACQKWFQTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1D2882 51 GPACQKWFQTCDSERKCCEGLVCRLWCKKKLW-COOH
    NV1G899 NV1D2774 52 GPACQKWMQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1G1077 NV1D2902 53 GPACQKWMQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2861 54 GPQCQKWFQTCDAERKCCEGMVCRLWCKKKLW-COOH
    NV1D2870 55 GPQCQKWFQTCDSERKCCEGLVCRLWCKKKLW-COOH
    NV1G1007 NV1D2775 56 GPQCQKWMQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1G1067 NV1D2893 57 GPQCQKWMQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2887 58 GPRCQKWFWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1G1005 NV1D2772 59 GPRCQKWMQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1G1061 NV1D2896 60 GPRCQKWMQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2877 61 GPSCQKWFQTCDSERKCCEGFVCRLWCKKKLW-COOH
    NV1D2878 62 GPSCQKWFQTCDSERKCCEGLVCRLWCKKKLW-COOH
    NV1D2889 63 GPSCQKWFWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2889 64 GPSCQKWFWTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1G1001 NV1D2773 65 GPSCQKWMQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2890 66 GPSCQKWFWTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1G1109 NV1D2899 67 GPSCQKWMQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2905 68 GPYCQKWFQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2906 69 GPYCQKWFQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2921 70 GPACQKWFQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2922 71 GPACQKWFQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2909 72 GPQCQKWFQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2910 73 GPQCQKWFQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2913 74 GPRCQKWFQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2914 75 GPRCQKWFQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1D2917 76 GPSCQKWFQTCDAERKCCEGFVCRLWCKKKLW-COOH
    NV1D2918 77 GPSCQKWFQTCDAERKCCEGLVCRLWCKKKLW-COOH
    NV1G1153 NV1D3034 78 GPQCQKWMQTCDRERKCCEGFVCTLWCRKKLW-COOH
  • TABLE 3
    Protoxin-II variant SEQ ID
    Protein name peptide name NO: Protein amino acid sequence
    (-GP) NV1G1001 (-GP) NV1D2773 109 SCQKWMQTCDAERKCCEGFVC
    RLWCKKKLW-COOH
    (-GP) (-GP) 110 SCQKWMQTCDAERKCCEGFVC
    NV1G1001-NH- NV1D2773-NH2 RLWCKKKLW-NH2
    NV1G1007-NH2 NV1D2775-NH2 111 GPQCQKWMQTCDAERKCCEG
    FVCRLWCKKKLW-NH2
    NV1G1107-NH2 NV1D2890-NH2 112 GPSCQKWFWTCDAERKCCEGL
    VCRLWCKKKLW-NH2
    NV1G1137 NV1D2974 113 GPQCQKWMQTCDAERKCCEG
    FSCTLWCKKKLW-COOH
    (-GP) N-Ac- (-GP) N-Ac- 114 Ac-QCQKWMQTCDAERKCCEG
    NV1G1137-NH2 NV1D2974-NH2 FSCTLWCKKKLW-NH2
    (-GP) N-Ac- (-GP) N-Ac- 115 Ac-QCQKWMQTCDAERKCCEG
    NV1G1137- NV1D2974 FSCTLWCKKKLW-COOH
    NV1G1153 NV1D3034 116 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1153-NH2 NV1D3034-NH2 117 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-NH2
    NV1G1153-NH- NV1D3034-NH- 118 GPQCQKWMQTCDRERKCCE
    butyl butyl GFVCTLWCRKKLW-NH-butyl
    NV1G1153-NH- NV1D3034-NH- 119 GPQCQKWMQTCDRERKCCE
    methyl methyl GFVCTLWCRKKLW-NH-methyl
    (-GP) N-Ac- (-GP) N-Ac- 120 Ac-QCQKWMQTCDRERKCCE
    NV1G1153 NV1D3034 GFVCTLWCRKKLW-COOH
    (-GP) N-Ac- (-GP) N-Ac- 121 Ac-QCQKWMQTCDRERKCCE
    NV1G1153-NH2 NV1D3034-NH2 GFVCTLWCRKKLW-NH2
    NV1G1818 NV1D3368 122 GPQCQKWMQTCDRTRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1818-NH2 NV1D3368-NH2 123 GPQCQKWMQTCDRTRKCCE
    GFVCTLWCRKKLW-NH2
    NV1G1147 NV1D2969 124 GPSCQKWMQTCDAERKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1145 NV1D2970 125 GPSCQKWMQTCDAERKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1143 NV1D2971 126 GPSCQKWMQTCDAERKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1141 NV1D2972 127 GPQCQKWMQTCDAERKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1139 NV1D2973 128 GPQCQKWMQTCDAERKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1137 NV1D2974 129 GPQCQKWMQTCDAERKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1137-NH2 NV1D2974-NH2 130 GPQCQKWMQTCDAERKCCE
    GFSCTLWCKKKLW-NH2
    NV1G1517 NV1D3004 131 GPQCQKWMQTCDRERKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1515 NV1D3005 132 GPQCQKWMQTCDANRKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1519 NV1D3006 133 GPQCQKWMQTCDARRKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1513 NV1D3007 134 GPQCQKWMQTCDAERKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1523 NV1D3012 135 GPQCQKWMQTCDRNRKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1525 NV1D3013 136 GPQCQKWMQTCDRRRKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1255 NV1D3014 137 GPQCQKWMQTCDRERKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1187 NV1D3015 138 GPQCQKWMQTCDRERKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1257 NV1D3016 139 GPQCQKWMQTCDANRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1221 NV1D3017 140 GPQCQKWMQTCDARRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1521 NV1D3018 141 GPQCQKWMQTCDANRKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1531 NV1D3019 142 GPQCQKWMQTCDARRKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1239 NV1D3020 143 GPQCQKWMQTCDAERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1583 NV1D3030 144 GPQCQKWMQTCDRNRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1527 NV1D3031 145 GPQCQKWMQTCDRRRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1511 NV1D3032 146 GPQCQKWMQTCDRNRKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1509 NV1D3033 147 GPQCQKWMQTCDRRRKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1231 NV1D3035 148 GPQCQKWMQTCDANRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1211 NV1D3036 149 GPQCQKWMQTCDARRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1267 NV1D3044 150 GPQCQKWMQTCDRNRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1269 NV1D3045 151 GPQCQKWMQTCDRRRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1215 NV1D3048 152 GPQCQKWMQTCDAKRKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1593 NV1D3050 153 GPQCQKWMQTCDRKRKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1263 NV1D3051 154 GPQCQKWMQTCDAKRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1585 NV1D3052 155 GPQCQKWMQTCDAKRKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1623 NV1D3056 156 GPQCQKWMQTCDRKRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1613 NV1D3057 157 GPQCQKWMQTCDRKRKCCE
    GFVCRLWCRKKLW-COOH
    NV1G1259 NV1D3058 158 GPQCQKWMQTCDAKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1265 NV1D3062 159 GPQCQKWMQTCDRKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1273 NV1D3109 160 GPQCQKWMWTCDARRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1225 NV1D3121 161 GPQCQKWMWTCDRKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1886 NV1D3249 162 GPAAAAAQCQKWMQTCDAER
    KCCEGFVCRLWCKKKLW-
    COOH
    NV1G1633 NV1D3251 163 GPAPAPAQCQKWMQTCDAER
    KCCEGFVCRLWCKKKLW-
    COOH
    NV1G1631 NV1D3252 164 GPQCQKWMQTCDAERKCCE
    GFVCRLWCKKKLWAPAPA-
    COOH
    NV1G1885 NV1D3254 165 GPQCQKWMQTCDAERKCCE
    GFVCRLWCKKKLWGGGGG-
    COOH
    NV1G1884 NV1D3256 166 GPCCNCSSKWCRDHSRCCG
    RGSAPAPAPAPAPGSQCQKW
    MQTCDAERKCCEGFVCRLWC
    KKKLW-COOH
    NV1G1881 NV1D3257 167 GPQCQKWMQTCDAERKCCE
    GFVCRLWCKKKLWGSAPAPA
    PAPAPGSCCNCSSKWCRDHS
    RCC-COOH
    NV1G1879 NV1D3259 168 GPQCQKWMQTCDAERKCCE
    GFVCRLWCKKKLWGSAPAPA
    PAPAPAPAPAPAPAPGSCCNC
    SSKWCRDHSRCCGR-COOH
    NV1G1883 NV1D3260 169 GPCCNCSSKWCRDHSRCCG
    RGSAPAPAPAPAPAPAPAPAP
    APGSQCQKWMQTCDAERKC
    CEGFVCRLWCKKKLW-COOH
    NV1G1880 NV1D3261 170 GPQCQKWMQTCDAERKCCE
    GFVCRLWCKKKLWGSAPAPA
    PAPAPAPAPAPAPAPGSCCNC
    SSKWCRDHSRCC-COOH
    NV1G1882 NV1D3262 171 GPCCNCSSKWCRDHSRCCG
    SAPAPAPAPAPAPAPAPAPAP
    GSQCQKWMQTCDAERKCCE
    GFVCRLWCKKKLW-COOH
    NV1G1776 NV1D3339 172 GPQCRKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1775 NV1D3340 173 GPQCKKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1768 NV1D3341 174 GPQCTKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1777 NV1D3342 175 GPQCAKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1770 NV1D3344 176 GPQCEKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1767 NV1D3345 177 GPQCSKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1769 NV1D3346 178 GPQCQRWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1774 NV1D3347 179 GPQCQTWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1771 NV1D3348 180 GPQCQAWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1778 NV1D3349 181 GPQCQDWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1773 NV1D3350 182 GPQCQEWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1779 NV1D3351 183 GPQCQQWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1772 NV1D3352 184 GPQCQSWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1868 NV1D3353 185 GPQCQKWMQRCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1824 NV1D3354 186 GPQCQKWMQKCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1863 NV1D3356 187 GPQCQKWMQDCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1826 NV1D3357 188 GPQCQKWMQECDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1810 NV1D3358 189 GPQCQKWMQQCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1836 NV1D3359 190 GPQCQKWMQSCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1834 NV1D3360 191 GPQCQKWMQTCRRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1829 NV1D3361 192 GPQCQKWMQTCKRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1820 NV1D3362 193 GPQCQKWMQTCTRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1828 NV1D3363 194 GPQCQKWMQTCARERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1827 NV1D3365 195 GPQCQKWMQTCQRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1857 NV1D3366 196 GPQCQKWMQTCSRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1823 NV1D3367 197 GPQCQKWMQTCDRQRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1818 NV1D3368 198 GPQCQKWMQTCDRTRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1811 NV1D3369 199 GPQCQKWMQTCDREKKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1853 NV1D3370 200 GPQCQKWMQTCDRETKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1817 NV1D3371 201 GPQCQKWMQTCDREAKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1814 NV1D3372 202 GPQCQKWMQTCDREDKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1831 NV1D3374 203 GPQCQKWMQTCDREQKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1819 NV1D3375 204 GPQCQKWMQTCDRESKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1859 NV1D3376 205 GPQCQKWMQTCDRERRCCE
    GFVCTLWCRKKLW-COOH
    NV1G1825 NV1D3377 206 GPQCQKWMQTCDRERTCCE
    GFVCTLWCRKKLW-COOH
    NV1G1821 NV1D3378 207 GPQCQKWMQTCDRERACCE
    GFVCTLWCRKKLW-COOH
    NV1G1835 NV1D3379 208 GPQCQDWMQTCDRERDCCE
    GFVCTLWCRKKLW-COOH
    NV1G1815 NV1D3380 209 GPQCQEWMQTCDRERECCE
    GFVCTLWCRKKLW-COOH
    NV1G1833 NV1D3381 210 GPQCQKWMQTCDRERQCCE
    GFVCTLWCRKKLW-COOH
    NV1G1812 NV1D3382 211 GPQCQKWMQTCDRERSCCE
    GFVCTLWCRKKLW-COOH
    NV1G1782 NV1D3383 212 GPQCQKWMQTCDRERKCCR
    GFVCTLWCRKKLW-COOH
    NV1G1783 NV1D3384 213 GPQCQKWMQTCDRERKCCK
    GFVCTLWCRKKLW-COOH
    NV1G1785 NV1D3385 214 GPQCQKWMQTCDRERKCCT
    GFVCTLWCRKKLW-COOH
    NV1G1784 NV1D3386 215 GPQCQKWMQTCDRERKCCA
    GFVCTLWCRKKLW-COOH
    NV1G1780 NV1D3387 216 GPQCQKWMQTCDRERKCCD
    GFVCTLWCRKKLW-COOH
    NV1G1781 NV1D3388 217 GPQCQKWMQTCDRERKCCQ
    GFVCTLWCRKKLW-COOH
    NV1G1786 NV1D3389 218 GPQCQKWMQTCDRERKCCS
    GFVCTLWCRKKLW-COOH
    NV1G1851 NV1D3390 219 GPQCQKWMQTCDRERKCCE
    RFVCTLWCRKKLW-COOH
    NV1G1852 NV1D3391 220 GPQCQKWMQTCDRERKCCE
    KFVCTLWCRKKLW-COOH
    NV1G1854 NV1D3392 221 GPQCQKWMQTCDRERKCCE
    TFVCTLWCRKKLW-COOH
    NV1G1860 NV1D3393 222 GPQCQKWMQTCDRERKCCE
    AFVCTLWCRKKLW-COOH
    NV1G1789 NV1D3394 223 GPQCQKWMQTCDRERKCCE
    DFVCTLWCRKKLW-COOH
    NV1G1787 NV1D3396 224 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1856 NV1D3397 225 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1855 NV1D3398 226 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1788 NV1D3399 227 GPQCQKWMQTCDRERKCCE
    GFTCTLWCRKKLW-COOH
    NV1G1849 NV1D3400 228 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1795 NV1D3401 229 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRRKLW-COOH
    NV1G1803 NV1D3403 230 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRAKLW-COOH
    NV1G1807 NV1D3408 231 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKRLW-COOH
    NV1G1806 NV1D3409 232 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKTLW-COOH
    NV1G1805 NV1D3410 233 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKALW-COOH
    NV1G1809 NV1D3413 234 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKQLW-COOH
    NV1G1850 NV1D3414 235 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKSLW-COOH
    NV1G1793 NV1D3419 236 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLD-COOH
    NV1G1822 NV1D3423 237 GPQCQKWMQTCRRRRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1813 NV1D3424 238 GPQCQKWMQTCKRKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1840 NV1D3425 239 GPQCQKWMQTCRRRDKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1848 NV1D3426 240 GPQCQKWMQTCKRKDKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1841 NV1D3427 241 GPQCQKWMQTCRRREKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1844 NV1D3428 242 GPQCQKWMQTCKRKEKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1842 NV1D3430 243 GPQCQDWMQTCDRERKCCK
    GFVCTLWCRKKLW-COOH
    NV1G1846 NV1D3431 244 GPQCQEWMQTCDRERKCCK
    GFVCTLWCRKKLW-COOH
    NV1G1843 NV1D3432 245 GPQCQEWMQTCDRERKCCR
    GFVCTLWCRKKLW-COOH
    NV1G1892 NV1D3439 246 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLG-COOH
    NV1G1916 NV1D3465 247 GPQCQKFMQTCDRERKCCEG
    FVCTLWCRKKLW-COOH
    NV1G1922 NV1D3466 248 GPQCQKWMQTCDEERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1915 NV1D3467 249 GPQCQKWMQTCDRERKCCG
    GFVCTLWCRKKLW-COOH
    NV1G1924 NV1D3470 250 GPQCQKWMQTCDRERKCCE
    GLVCTLWCRKKLW-COOH
    NV1G1709 NV1D3510 251 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPASP
    GARAF-COOH
    NV1G1681 NV1D3511 252 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWSPGARAF-
    COOH
    NV1G1693 NV1D3512 253 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAPA
    PAPDGPWRKM-COOH
    NV1G1705 NV1D3513 254 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPADG
    PWRKM-COOH
    NV1G1689 NV1D3514 255 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWDGPWRK
    M-COOH
    NV1G1711 NV1D3515 256 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAPA
    PAPFGQKASS-COOH
    NV1G1685 NV1D3516 257 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAFG
    QKASS-COOH
    NV1G1697 NV1D3517 258 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWFGQKASS-
    COOH
    NV1G1695 NV1D3518 259 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAPA
    PAPQRFVTGHFGGLYPANG-
    COOH
    NV1G1701 NV1D3519 260 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAQR
    FVTGHFGGLYPANG-COOH
    NV1G1691 NV1D3520 261 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWQRFVTGH
    FGGLYPANG-COOH
    NV1G1679 NV1D3521 262 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAPA
    PAPRRRRRRRRRRR-COOH
    NV1G1683 NV1D3523 263 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWRRRRRRR
    RRRR-COOH
    NV1G1707 NV1D3524 264 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAPA
    PAPYGRKKRRQRRR-COOH
    NV1G1713 NV1D3525 265 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAYG
    RKKRRQRRR-COOH
    NV1G1687 NV1D3526 266 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWYGRKKRR
    QRRR-COOH
    NV1G1699 NV1D3527 267 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPAPA
    PAP-COOH
    NV1G1675 NV1D3528 268 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPA-
    COOH
    NV1G1754 NV1D3529 269 GPRCQKWMQTCDAKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1748 NV1D3530 270 GPSCQKWMQTCDAKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1747 NV1D3531 271 GPYCQKWMQTCDAKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1752 NV1D3532 272 GPACQKWMQTCDAKRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1722 NV1D3533 273 GPQCQKWMQTCDAKRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1744 NV1D3534 274 GPRCQKWMQTCDAKRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1742 NV1D3535 275 GPSCQKWMQTCDAKRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1723 NV1D3536 276 GPYCQKWMQTCDAKRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1745 NV1D3537 277 GPACQKWMQTCDAKRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1757 NV1D3538 278 GPRCQKWMQTCDRNRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1762 NV1D3539 279 GPSCQKWMQTCDRNRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1763 NV1D3540 280 GPYCQKWMQTCDRNRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1728 NV1D3541 281 GPACQKWMQTCDRNRKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1730 NV1D3542 282 GPQCQKWMQTCDRNRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1760 NV1D3543 283 GPRCQKWMQTCDRNRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1727 NV1D3544 284 GPSCQKWMQTCDRNRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1729 NV1D3545 285 GPYCQKWMQTCDRNRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1867 NV1D3546 286 GPACQKWMQTCDRNRKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1759 NV1D3547 287 GPRCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1758 NV1D3548 288 GPSCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1766 NV1D3549 289 GPYCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1761 NV1D3550 290 GPACQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1726 NV1D3551 291 GPRCQKWMQTCDRERKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1721 NV1D3552 292 GPSCQKWMQTCDRERKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1765 NV1D3553 293 GPYCQKWMQTCDRERKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1764 NV1D3554 294 GPACQKWMQTCDRERKCCE
    GFSCTLWCRKKLW-COOH
    NV1G1732 NV1D3555 295 GPRCQKWMQTCDAERKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1862 NV1D3556 296 GPYCQKWMQTCDAERKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1751 NV1D3558 297 GPRCQKWMQTCDANRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1866 NV1D3559 298 GPSCQKWMQTCDANRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1865 NV1D3560 299 GPYCQKWMQTCDANRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1716 NV1D3561 300 GPACQKWMQTCDANRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1724 NV1D3562 301 GPRCQKWMQTCDARRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1717 NV1D3563 302 GPSCQKWMQTCDARRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1743 NV1D3564 303 GPYCQKWMQTCDARRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1720 NV1D3565 304 GPACQKWMQTCDARRKCCE
    GFSCTLWCKKKLW-COOH
    NV1G1735 NV1D3566 305 GPRCQKWMQTCDAERKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1734 NV1D3568 306 GPACQKWMQTCDAERKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1741 NV1D3569 307 GPRCQKWMQTCDARRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1719 NV1D3570 308 GPSCQKWMQTCDARRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1718 NV1D3571 309 GPYCQKWMQTCDARRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1725 NV1D3572 310 GPACQKWMQTCDARRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1869 NV1D3573 311 GPRCQKWMQTCDANRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1755 NV1D3574 312 GPSCQKWMQTCDANRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1756 NV1D3575 313 GPYCQKWMQTCDANRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1746 NV1D3576 314 GPACQKWMQTCDANRKCCE
    GFVCTLWCKKKLW-COOH
    NV1G1733 NV1D3577 315 GPRCQKWMQTCDAERKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1738 NV1D3578 316 GPYCQKWMQTCDAERKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1737 NV1D3579 317 GPACQKWMQTCDAERKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1740 NV1D3580 318 GPRCQKWMQTCDARRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1864 NV1D3581 319 GPSCQKWMQTCDARRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1739 NV1D3582 320 GPYCQKWMQTCDARRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1870 NV1D3583 321 GPACQKWMQTCDARRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1715 NV1D3584 322 GPRCQKWMQTCDANRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1753 NV1D3585 323 GPSCQKWMQTCDANRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1750 NV1D3586 324 GPYCQKWMQTCDANRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1750-NH2 NV1D3586-NH2 325 GPYCQKWMQTCDANRKCCE
    GFSCRLWCKKKLW-NH2
    NV1G1749 NV1D3587 326 GPACQKWMQTCDANRKCCE
    GFSCRLWCKKKLW-COOH
    NV1G1871 NV1D3772 327 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWSHSNTQT
    LAKAPEHTG-COOH
    NV1G1839 NV1D3774 328 GPSHSNTQTLAKAPEHTGAPA
    PAPAPAPAPAPAPAPAPQCQK
    WMQTCDRERKCCEGFVCTLW
    CRKKLW-COOH
    NV1G1877 NV1D3775 329 GPSHSNTQTLAKAPEHTGAPA
    PAPAPAPQCQKWMQTCDRER
    KCCEGFVCTLWCRKKLW-
    COOH
    NV1G1872 NV1D3777 330 GPSHSNTQTLAKAPEHTGQC
    QKWMQTCDRERKCCEGFVCT
    LWCRKKLW-COOH
    NV1G1941 NV1D3782 331 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKAW-COOH
    NV1G1990 NV1D3788 332 GPAAAAAQCQKWMQTCDRE
    RKCCEGFVCTLWCRKKLW-
    COOH
    NV1G1991 NV1D3789 333 GPAPAPAQCQKWMQTCDRE
    RKCCEGFVCTLWCRKKLW-
    COOH
    NV1G1989 NV1D3791 334 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAAAAA-
    COOH
    NV1G1993 NV1D3792 335 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWGGGGG-
    COOH
    NV1G1967 NV1D3793 336 GPCCNCSSKWCRDHSRCCG
    RGSAPAPAPAPAPAPAPAPAP
    APGSQCQKWMQTCDRERKC
    CEGFVCTLWCRKKLW-COOH
    NV1G1969 NV1D3795 337 GPCCNCSSKWCRDHSRCCG
    SAPAPAPAPAPAPAPAPAPAP
    GSQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLW-COOH
    NV1G1974 NV1D3796 338 GPCCNCSSKWCRDHSRCCG
    SAPAPAPAPAPGSQCQKWMQ
    TCDRERKCCEGFVCTLWCRK
    KLW-COOH
    NV1G1950 NV1D3797 339 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWGSAPAPA
    PAPAPAPAPAPAPAPGSCCNC
    SSKWCRDHSRCC-COOH
    NV1G1948 NV1D3798 340 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWGSAPAPA
    PAPAPAPAPAPAPAPGSCCNC
    SSKWCRDHSRCCGR-COOH
    NV1G2057 NV1D3799 341 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWGSAPAPA
    PAPAPGSCCNCSSKWCRDHS
    RCC-COOH
    NV1G1954 NV1D3800 342 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWGSAPAPA
    PAPAPGSCCNCSSKWCRDHS
    RCCGR-COOH
    NV1G1956 NV1D3801 343 GPSPGARAFAPAPAPAPAPQC
    QKWMQTCDRERKCCEGFVCT
    LWCRKKLW-COOH
    NV1G1961 NV1D3802 344 GPSPGARAFAPAPAQCQKWM
    QTCDRERKCCEGFVCTLWCR
    KKLW-COOH
    NV1G1960 NV1D3803 345 GPSPGARAFQCQKWMQTCD
    RERKCCEGFVCTLWCRKKLW-
    COOH
    NV1G1977 NV1D3804 346 GPDGPWRKMAPAPAPAPAPQ
    CQKWMQTCDRERKCCEGFV
    CTLWCRKKLW-COOH
    NV1G1982 NV1D3805 347 GPDGPWRKMAPAPAQCQKW
    MQTCDRERKCCEGFVCTLWC
    RKKLW-COOH
    NV1G1984 NV1D3806 348 GPDGPWRKMQCQKWMQTCD
    RERKCCEGFVCTLWCRKKLW-
    COOH
    NV1G1985 NV1D3808 349 GPFGQKASSAPAPAQCQKWM
    QTCDRERKCCEGFVCTLWCR
    KKLW-COOH
    NV1G1983 NV1D3809 350 GPFGQKASSQCQKWMQTCD
    RERKCCEGFVCTLWCRKKLW-
    COOH
    NV1G1973 NV1D3810 351 GPQRFVTGHFGGLYPANGAP
    APAPAPAPQCQKWMQTCDRE
    RKCCEGFVCTLWCRKKLW-
    COOH
    NV1G1976 NV1D3811 352 GPQRFVTGHFGGLYPANGAP
    APAQCQKWMQTCDRERKCC
    EGFVCTLWCRKKLW-COOH
    NV1G1980 NV1D3812 353 GPQRFVTGHFGGLYPANGQC
    QKWMQTCDRERKCCEGFVCT
    LWCRKKLW-COOH
    NV1G1952 NV1D3813 354 GPRRRRRRRRRRRAPAPAPA
    PAPQCQKWMQTCDRERKCC
    EGFVCTLWCRKKLW-COOH
    NV1G1957 NV1D3814 355 GPRRRRRRRRRRRAPAPAQC
    QKWMQTCDRERKCCEGFVCT
    LWCRKKLW-COOH
    NV1G1981 NV1D3815 356 GPRRRRRRRRRRRQCQKWM
    QTCDRERKCCEGFVCTLWCR
    KKLW-COOH
    NV1G1959 NV1D3818 357 GPYGRKKRRQRRRQCQKWM
    QTCDRERKCCEGFVCTLWCR
    KKLW-COOH
    NV1G1986 NV1D3819 358 GPAPAPAPAPAPQCQKWMQT
    CDRERKCCEGFVCTLWCRKK
    LW-COOH
    NV1G1968 NV1D3822 359 GPGWCGDPGATCGKLRLYCCSGFCDSYTKTCK
    DKSSAGGGGSAPAPAPAPAPAPAPAPAPAPAP
    APAPAPAPGGGGSQCQKWMQTCDRERKCCEG
    FVCTLWCRKKLW-COOH
    NV1G1945 NV1D3823 360 GPQCQKWMQTCDRERKCCEGFVCTLWC
    RKKLWGGGGSAPAPAPAPAPAPAPAPAPA
    PAPAPAPAPAPGGGGSGWCGDPGATCG
    KLRLYCCSGFCDSYTKTCKDKSSA-COOH
    NV1G1972 NV1D3824 361 GPGWCGDPGATCGKLRLYCCSGFCD
    AYTKTCKDKSSAGGGGSAPAPAPAP
    APAPAPAPAPAPAPAPAPAPAPGGG
    GSQCQKWMQTCDRERKCCEGFVCT
    LWCRKKLW-COOH
    NV1G1946 NV1D3825 362 GPQCQKWMQTCDRERKCCEGFVCT
    LWCRKKLWGGGGSAPAPAPAPAPA
    PAPAPAPAPAPAPAPAPAPGGGGSG
    WCGDPGATCGKLRLYCCSGFCDAYT
    KTCKDKSSA-COOH
    NV1G1970 NV1D3826 363 GPGWCGDPGATCGKLRLYCCSGFCD
    CYTKTCKDKSSAGGGGSAPAPAPAP
    APAPAPAPAPAPAPAPAPAPAPGGG
    GSQCQKWMQTCDRERKCCEGFVCT
    LWCRKKLW-COOH
    NV1G1949 NV1D3828 364 GPQCQKWMQTCDRERKCCEGFVCT
    LWCRKKLWGSGGGGSAPAPAPAPA
    PAPAPAPAPAPGGGGSGSCCNCSSK
    WCRDHSRCCGR-COOH
    NV1G1951 NV1D3829 365 GPQCQKWMQTCDRERKCCEGFVCT
    LWCRKKLWGSGGGGSAPAPAPAPA
    PAPAPAPAPAPGGGGSGSCCNCSSK
    WCRDHSRCC-COOH
    NV1G1971 NV1D3830 366 GPCCNCSSKWCRDHSRCCGRGSGG
    GGSAPAPAPAPAPAPAPAPAPAPGG
    GGSGSQCQKWMQTCDRERKCCEGF
    VCTLWCRKKLW-COOH
    NV1G1975 NV1D3832 367 GPCRTIGPSVCAPAPAPAPAPAPAPA
    PAPAPQCQKWMQTCDRERKCCEGF
    VCTLWCRKKLW-COOH
    NV1G1978 NV1D3833 368 GPCRTIGPSVCAPAPAPAPAP
    QCQKWMQTCDRERKCCEGF
    VCTLWCRKKLW-COOH
    NV1G1979 NV1D3834 369 GPCRTIGPSVCAPAPAQCQK
    WMQTCDRERKCCEGFVCTLW
    CRKKLW-COOH
    NV1G2043 NV1D3835 370 GPCRTIGPSVCQCQKWMQTC
    DRERKCCEGFVCTLWCRKKL
    W-COOH
    NV1G1955 NV1D3838 371 GPQCQKWMQTCDRERKCCE
    GFVCTLWCRKKLWAPAPACR
    TIGPSVC-COOH
  • In some embodiments, the isolated Protoxin-II variant inhibits human Nav1.7 activity with an IC50 value of about 3×10−9 M or less.
  • In some embodiments, the isolated Protoxin-II variant inhibits human Nav1.7 activity with an IC50 value of between about 3×10−9 M to about 1×10−9 M.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the amino acid sequence GPQCX1X2WX3QX4CX5X6X7X3X9CCX10X11FX12CX13LWCX14KKLL (SEQ ID NO: 433), wherein
  • X1 is Q, R, K, A or S;
  • X2 is K, S, Q or R;
  • X3 is M or F;
  • X4 is T, S, R, K or Q;
  • X5 is D or T;
  • X6 is S, A or R;
  • X7 is E, R, N, K, T or Q;
  • X8 is R or K;
  • X9 is K, Q, S or A;
  • X10 is E, Q or D;
  • X11 is G or Q;
  • X12 is V or S;
  • X13 is R or T; and
  • X14 is K or R.
  • Exemplary Protoxin-II variants that inhibit human Nav1.7 activity with an ICs value of about 30×10−9 M or less are variants comprising the amino acid sequences of SEQ ID NOs: 56, 78, 111, 114, 117, 118, 119, 122, 123, 129, 130, 131, 132, 133, 134, 135, 136, 138, 139, 140, 141, 142, 145, 146, 147, 149, 150, 151, 152, 153, 154, 156, 158, 159, 165, 172, 173, 175, 177, 178, 183, 184, 185, 186, 189, 190, 193, 197, 199, 207, 210, 211, 216, 217, 224, 266, 273, 282, 335, 408, 409, 410, 422, 424, 425, 426, 427, and 428.
  • In some embodiments, the isolated Protoxin-II variant selectively inhibits human Nav1.7. The Protoxin-II variants of the invention may be more selective towards Nav1.7 when compared to the recombinant Protoxin-II (SEQ ID NO: 2). In the QPatch electrophysiology assay, recombinant Protoxin-II has an IC50 of about 2.2×10−9 M for Nav1.7 and an IC50 of about 62×10−9 M for Nav1.6, and therefore the ratio of IC50 for Nav1.6 to IC50 for Nav1.7 about 28 fold. “Selectivity” or “selective” or “more selective” or “selectively blocks” or “selectively inhibits” when used herein refers to a Protoxin-II variant that has a ratio of IC50 for Nav1.6 to IC50 for Nav1.7 (IC50(Nav1.6)/IC50(Nav1.7)) equal or over about 30. IC50 for Nav1.6 may be assayed in a QPatch electrophysiology assay using cell lines stably expressing Nav1.6 using similar methods to those described for Nav1.7.
  • Residue positions in Protoxin-II that can be mutagenized to improve selectivity include residues 7, 11, 12, 14, 17, 18 and 19, and optionally residues 1, 20, 22 and 26 (residue numbering according to SEQ ID NO: 1). Exemplary substitutions to improve selectivity are Y1Q, W7Q, S11R, S11A, E12T, M19F, V20S, R22T, and K26R. Exemplary Protoxin-II variants with improved selectivity are variants of SEQ ID NOs: 56, 59, 65, 78, 111, 114, 117, 118, 119, 121, 122, 123, 129, 130, 133, 150, 190, 217, 281, 324, 325 or 326.
  • Another embodiment of the invention is an isolated Protoxin-II variant comprising the sequence GPX1CQKWMQX2CDX3X4RKCCX5GFX6CX7LWCX8KKLW (SEQ ID NO: 405); wherein
  • X1 is Y, Q, A, S or R;
  • X2 is T or S;
  • X3 is S, R or A;
  • X4 is E, T or N;
  • X5 is E or Q;
  • X6 is V or S;
  • X7 is R or T; and
  • X8 is K or R;
  • wherein the Protoxin-II variant inhibits human Nav1.7 activity with an IC50 value of about 3×10−8 M or less, and selectively inhibits human Nav1.7.
  • In some embodiments, the isolated Protoxin-II variant comprises the sequence GPQCQKWMQX1CDX2X3RKCCX4GFX5CX6LWCX8KKLW (SEQ ID NO: 406); wherein
  • X1 is Tor S;
  • X2 is S, R or A;
  • X3 is E, T or N;
  • X4 is E or Q;
  • X5 is V or S;
  • X6 is R or T; and
  • X7 is K or R.
  • Another embodiment is an isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKWMQTCDSERKCCEGMVCRLWCKKKLL-COOH); wherein
  • the amino acid sequence has Q at position 7 and L at position 30, when residue numbering is according to SEQ ID NO: 1; and
    the polypeptide inhibits human Nav1.7 activity with an ICs value of about 30×10−9 M or less, wherein the ICs value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • Protoxin-II variants having substitutions W7Q and W30L have improved folding, yield and selectivity when compared to the wild type Protoxin-II.
  • Another embodiment is an isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 78 (GPQCQKWMQTCDRERKCCEGFVCTLWCRKKLW-COOH); wherein
  • the amino acid sequence has Q at position 1, Q at position 7 and F at position 19, when residue numbering is according to SEQ ID NO: 1;
    the polypeptide inhibits human Nav1.7 activity with an IC50 value of about 30×10−9 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and
    the polypeptide selectively inhibits Nav1.7.
  • In some embodiments, the isolated Protoxin-II variant has a free C-terminal carboxylic acid, amide, methylamide or butylamide group, which are generated via routine synthetic methods.
  • Another embodiment of the invention is an isolated fusion protein comprising the Protoxin-II variant of SEQ ID NOs: 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 109, 110, 112, 113, 114, 115, 116, 117, 118, 119, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, or 431. Such second polypeptides may be well known leader or secretory signal sequences, or synthetic sequences resulting for example from cloning steps, or tags such as hexahistidine tag (SEQ ID NO: 108). Such second polypeptide may be a half-life extending moiety. In one embodiment, the isolated fusion protein comprises the Protoxin-II variant of the invention conjugated to a half-life extending moiety.
  • Exemplary half-life extending moieties that can be used include well known human serum albumin, transthyretin (TTR), a thyroxine-binding globulin (TGB), albumin-binding domains, or an Fc or fragments thereof. Biologically suitable polymers or copolymers may also be used, for example ethylene glycol or polyethylene glycol (PEG) molecules, such as PEG5000 or PEG20000, dextran, polylysine, fatty acids and fatty acid esters of different chain lengths, for example laurate, myristate, stearate, arachidate, behenate, oleate, arachidonate, octanedioic acid, tetradecanedioic acid, octadecanedioic acid, docosanedioic acid, and the like, octane, or carbohydrates (dextran, cellulose, oligo- or polysaccharides). These moieties may be direct fusions with the Protoxin-II variant polypeptides and may be generated by standard cloning and expression techniques. Alternatively, well known chemical coupling methods may be used to attach the moieties to recombinantly produced Protoxin-II variants of the invention.
  • In another embodiment, the half-life extending moiety of the fusion protein of the invention is human serum albumin, albumin binding domain (ABD), or polyethylene glycol (PEG).
  • In another embodiment, the half-life extending moiety of is conjugated to the Protoxin-II variant via a linker. Suitable linkers are well known and include linkers having the sequence shown in SEQ ID NOs: 80 or 81.
  • Exemplary fusion proteins incorporating Protoxin-II variants of the invention are those having the polypeptide sequence of SEQ ID NOs: 83, 85, 87, 89, 91, 93, 95, 97, 99, 101 or 103.
  • Protoxin-II variants of the invention incorporating additional moieties may be compared for functionality by several well-known assays. For example, pharmacokinetic properties of Protoxin-II variants coupled to PEG may be evaluated in well known in vivo models.
  • Additional Protoxin-II variants and Protoxin-II variant fusion proteins are within the scope of the invention. Additional substitutions to the Protoxin-II variants of the invention can be made as long as the resulting variant or the fusion protein retains similar characteristics when compared to the parent peptide. Exemplary modifications are for example conservative substitutions that will result in Protoxin-II variants with similar characteristics to those of the parent molecules. Conservative replacements are those that take place within a family of amino acids that are related in their side chains. Genetically encoded amino acids can be divided into four families: (1) acidic (aspartate, glutamate); (2) basic (lysine, arginine, histidine); (3) nonpolar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan); and (4) uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine). Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. Alternatively, the amino acid repertoire can be grouped as (1) acidic (aspartate, glutamate); (2) basic (lysine, arginine histidine), (3) aliphatic (glycine, alanine, valine, leucine, isoleucine, serine, threonine), with serine and threonine optionally grouped separately as aliphatic-hydroxyl; (4) aromatic (phenylalanine, tyrosine, tryptophan); (5) amide (asparagine, glutamine); and (6) sulfur-containing (cysteine and methionine) (Stryer (ed.), Biochemistry, 2nd ed, WH Freeman and Co., 1981). Non-conservative substitutions can be made to the Protoxin-II variants that involve substitutions of amino acid residues between different classes of amino acids to improve properties of the Protoxin-II variants and Protoxin-II variant fusion proteins. Whether a change in the amino acid sequence of a polypeptide or fragment thereof results in a functional homolog can be readily determined by assessing the ability of the modified polypeptide or fragment to produce a response in a fashion similar to the unmodified polypeptide or fragment using the assays described herein. Peptides, polypeptides or proteins in which more than one replacement takes place can readily be tested in the same manner.
  • Another embodiment of the invention is an isolated synthetic polynucleotide comprising a polynucleotide encoding the Protoxin-II variant of the invention.
  • Certain exemplary synthetic polynucleotides are disclosed herein, however, other synthetic polynucleotides which, given the degeneracy of the genetic code or codon preferences in a given expression system, encode the Protoxin-II variants and Protoxin-II variant fusion proteins of the invention are also within the scope of the invention. Exemplary synthetic polynucleotides are for example polynucleotide sequences shown in SEQ ID NOs: 84, 86, 88, 90, 92, 94, 96, 98, 100, 102 and 104, which encode the Protoxin-II variant fusion proteins of the invention. Those skilled in the art can readily identify the polynucleotide segments in the fusion proteins that encode the Protoxin-II variant itself. The synthetic polynucleotide sequences encoding the Protoxin-II variants or fusion proteins of the invention can be operably linked to one or more regulatory elements, such as a promoter and enhancer, that allow expression of the nucleotide sequence in the intended host cell. The synthetic polynucleotide may be a cDNA.
  • The polynucleotides of the invention may be produced by chemical synthesis such as solid phase polynucleotide synthesis on an automated polynucleotide synthesizer. Alternatively, the polynucleotides of the invention may be produced by other techniques such as PCR based duplication, vector based duplication, or restriction enzyme based DNA manipulation techniques. Techniques for producing or obtaining polynucleotides of known sequences are well known.
  • The polynucleotides of the invention may also comprise at least one non-coding sequence, such as transcribed but not translated sequences, termination signals, ribosome binding sites, mRNA stabilizing sequences, introns and polyadenylation signals. The polynucleotide sequences may also comprise additional sequences encoding additional amino acids. These additional polynucleotide sequences may, for example, encode a marker or well-known tag sequences such as a hexa-histidine (SEQ ID NO: 108) or a HA tag which facilitate the purification of fused polypeptides.
  • Another embodiment of the invention is a vector comprising the polynucleotide of the invention. Such vectors may be plasmid vectors, viral vectors, vectors for baculovirus expression, transposon based vectors or any other vector suitable for introduction of the polynucleotide of the invention into a given organism or genetic background by any means. For example, polynucleotides encoding the Protoxin-II variants or the Protoxin-II variant fusion proteins of the invention are inserted into an expression vector and may be operably linked to control sequences in the expression vector to ensure efficient expression, such as signal sequences, promoters (e.g. naturally associated or heterologous promoters), enhancer elements, and transcription termination sequences, and are chosen to be compatible with the host cell chosen to express the Protoxin-II variant or the Protoxin-II variant fusion protein of the invention. Once the vector has been incorporated into the appropriate host, the host is maintained under conditions suitable for high level expression of the proteins encoded by the incorporated polynucleotides.
  • Suitable expression vectors are typically replicable in the host organisms either as episomes or as an integral part of the host chromosomal DNA. Commonly, expression vectors contain selection markers such as ampicillin-resistance, hygromycin-resistance, tetracycline resistance, kanamycin resistance or neomycin resistance to permit detection of those cells transformed with the desired DNA sequences.
  • Suitable promoter and enhancer elements are known in the art. For expression in a bacterial cell, suitable promoters include, but are not limited to, lacI, lacZ, T3, T7, gpt, lambda P and trc. For expression in a eukaryotic cell, suitable promoters include, but are not limited to, light and/or heavy chain immunoglobulin gene promoter and enhancer elements; cytomegalovirus immediate early promoter; herpes simplex virus thymidine kinase promoter; early and late SV40 promoters; promoter present in long terminal repeats from a retrovirus; mouse metallothionein-I promoter; and various art-known tissue specific promoters. For expression in a yeast cell, a suitable promoter is a constitutive promoter such as an ADH1 PGK1, ENO or PYK1 promoter and the like, or a regulatable promoter such as a GALL or GAL10 promoter. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • Large numbers of suitable vectors and promoters are known to those of skill in the art; many are commercially available for generating recombinant constructs. The following vectors are provided by way of example. Bacterial: pBs, phagescript, PsiX174, pBluescript SK, pBs KS, pNH8a, pNH16a, pNH18a, pNH46a (Stratagene, La Jolla, Calif., USA); pTrc99A, pKK223-3, pKK233-3, pDR540, and pRIT5 (Pharmacia, Uppsala, Sweden). Eukaryotic: pWLneo, pSV2cat, pOG44, PXR1, pSG (Stratagene) pSVK3, pBPV, pMSG and pSVL (Pharmacia).
  • An exemplary vector for expression of the Protoxin-II variants or Protoxin-II variant fusion proteins is a vector having ampicillin-resistance selection marker, CMV promoter, CMV intron, signal peptide, neomycin resistance, f1 origin of replication, SV40 polyadenylation signal, and cDNA encoding the Protoxin-II variant or the Protoxin-II variant fusion protein of the invention.
  • Another embodiment of the invention is a host cell comprising the vector of the invention. The term “host cell” refers to a cell into which a vector has been introduced. It is understood that the term host cell is intended to refer not only to the particular subject cell but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not be identical to the parent cell, but are still included within the scope of the term “host cell” as used herein. Such host cells may be eukaryotic cells, prokaryotic cells, plant cells or archaeal cells.
  • Escherichia coli, bacilli, such as Bacillus subtilis, and other enterobacteriaceae, such as Salmonella, Serratia, and various Pseudomonas species, are examples of prokaryotic host cells. Other microbes, such as yeast, are also useful for expression. Saccharomyces (e.g., S. cerevisiae) and Pichia are examples of suitable yeast host cells. Exemplary eukaryotic cells may be of mammalian, insect, avian or other animal origins. Mammalian eukaryotic cells include immortalized cell lines such as hybridomas or myeloma cell lines such as SP2/0 (American Type Culture Collection (ATCC), Manassas, Va., CRL-1581), NSO (European Collection of Cell Cultures (ECACC), Salisbury, Wiltshire, UK, ECACC No. 85110503), FO (ATCC CRL-1646) and Ag653 (ATCC CRL-1580) murine cell lines. An exemplary human myeloma cell line is U266 (ATTC CRL-TIB-196). Other useful cell lines include those derived from Chinese Hamster Ovary (CHO) cells such as CHO-K1SV (Lonza Biologics, Walkersville, Md.), CHO-K1 (ATCC CRL-61) or DG44.
  • Introduction of a polynucleotide, such as a vector, into a host cell can be effected by methods well known to those skilled in the art. Exemplary methods are calcium phosphate transfection, DEAE-Dextran mediated transfection, microinjection, cationic lipid-mediated transfection and electroporation.
  • Another embodiment of the invention is a method for producing the Protoxin-II variant of the invention comprising the steps of providing a host cell of the invention; and culturing the host cell under conditions sufficient for the expression of at least one Protoxin-II variant of the invention.
  • Host cells can be cultured under any conditions suitable for maintaining or propagating a given type of host cell and sufficient for expressing a polypeptide. Culture conditions, media, and related methods sufficient for the expression of polypeptides are well known in the art. For example, many mammalian cell types can be aerobically cultured at 37° C. using appropriately buffered DMEM media while bacterial, yeast and other cell types may be cultured at 37° C. under appropriate atmospheric conditions in LB media.
  • In the methods of the invention, the expression of the Protoxin-II variant can be confirmed using a variety of well-known methods. For example, expression of a polypeptide can be confirmed using detection reagents, such as using SDS-PAGE or HPLC.
  • Another aspect of the invention is a method of modulating the activity of Nav1.7 in a biological tissue, the method comprising contacting the biological tissue expressing Nav1.7 with a Nav1.7-modulating amount of the Protoxin-II variant of the invention.
  • Methods of Treatment
  • Protoxin-II variants of the invention may be utilized in any therapy where it is desired to treat, reduce or alleviate symptoms of pain or other disorders of sensory or sympathetic neuron dysfunction.
  • Pain treated with the Protoxin-II variants of the invention may be any type of pain, such as chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, pain associated with inflammatory conditions, post-operative pain, thermal pain or pain associated with disease and degeneration.
  • Pain treated with the Protoxin-II variants of the invention may be Nav1.7-mediated pain.
  • Nav1.7-mediated pain as used herein refers to pain resulting at least partially from increased Nav1.7 channel activity.
  • The methods of the invention may be used to treat an animal patient belonging to any classification. Examples of such animals include mammals such as humans, rodents, dogs, cats and farm animals.
  • The pain and/or Nav1.7-mediated pain may result from one or more causes, such as peripheral neuropathy, central neuropathy, nerve compression or entrapment syndromes such as carpal tunnel syndrome, tarsus tunnel syndrome, ulnar nerve entrapment, compression radiculopathy, lumbar spinal stenosis, sciatic nerve compression, spinal root compression, intercostal neuralgia, compression radiculopathy and radicular lower back pain, spinal root lesions, neuritis, autoimmune diseases, general inflammation, chronic inflammatory conditions, arthritis, rheumatic diseases, lupus, osteoarthritis, general gastrointestinal disorders, colitis, gastric ulceration, duodenal ulcers, inflammatory bowel disorders, irritable bowel syndrome, pain associated with diarrhea, inflammatory eye disorders, inflammatory or unstable bladder disorders, psoriasis, skin complaints with inflammatory components, sunburn, carditis, dermatitis, myositis, neuritis, collagen vascular diseases, inflammatory pain and associated hyperalgesia and allodynia, neuropathic pain and associated hyperalgesia and allodynia, multiple sclerosis, demyelinating diseases, diabetes, diabetic neuropathy pain, causalgia, pain resulting from amputation or abscess, phantom limb pain, fracture pain, bone injury, direct trauma, HIV infection, acquired immune deficiency syndrome (“AIDS”), smallpox infection, herpes infection, exposure to toxins or other foreign particles or molecules, invasive cancer, cancer, chemotherapy, radiotherapy, hormonal therapy, burns, congenital defect, dental pain, gout pain, fibromyalgias, encephalitis, chronic alcoholism, hypothyroidism, uremia and vitamin deficiencies, trigeminal neuralgia, stroke, thalamic pain syndrome, general headache, migraine, cluster headache, tension headache, mixed-vascular and non-vascular syndromes, sympathetically maintained pain, deafferentation syndromes, asthma, epithelial tissue damage or dysfunction, disturbances of visceral motility at respiratory, genitourinary, gastrointestinal or vascular regions, wounds, burns, allergic skin reactions, pruritis, vasomotor or allergic rhinitis, or bronchial disorders, dysmenorrhea, pain during labor and delivery, dyspepsia, gastroesophageal reflux, pancreatitis, and visceralgia.
  • Other disorders of sensory or sympathetic neuron dysfunction that may be alleviated by the Protoxin-II variants of the invention include itch, cough and asthma. In mice, global deletion of the SCN9A gene leads to complete insensitivity to histamine-induced itch (Gingras et al., American Pain Society Meeting Abstract 2013 and U.S. Pat. Publ. No. 2012/0185956). This finding suggests that peptide Nav1.7 blockers may have utility in the treatment of itch, which may arise from various sources, such as dermatological or inflammatory disorders; or inflammatory disorders such as renal or hepatobiliary disorders, immunological disorders, medication reactions and unknown/idiopathic conditions, including dermatitis, psoriasis, eczema, insect sting or bite. Nav1.7 is also expressed in sensory nerves innervating the airways (Muroi et al., J Physiol. 2011 Dec. 1; 589(Pt 23):5663-76; Muroi et al., Am J Physiol Regul Integr Comp Physiol. 2013 Apr. 10), suggesting that peptide Nav1.7 blockers may be beneficial in the treatment of cough e.g., acute or chronic cough, or cough caused by irritation from gastroesophageal reflux disease, and inflammatory diseases of the airways such as asthma and allergy-related immune responses, bronchospasm, chronic obstructive pulmonary disease, chronic bronchitis, emphysema, and hiccups (hiccoughs, singultus). Silencing Nav1.7 in vivo in nodose ganglia of guinea pigs using shRNA nearly abolished the cough reflex induced by mechanical probing (Muroi et al., Am J Physiol Regul Integr Comp Physiol. 2013 Apr. 10).
  • One aspect of the invention is a method of alleviating or treating itch, cough or asthma in a subject by administering a therapeutically effective amount of the Protoxin-II variant of the invention to a subject in need thereof for a time sufficient to alleviate the itch, cough or asthma.
  • Another aspect of the invention is a method of alleviating or treating Nav1.7-mediated itch, Nav1.7-mediated cough or Nav1.7-mediated asthma in a subject by administering a therapeutically effective amount of the Protoxin-II variant of the invention to a subject in need thereof for a time sufficient to alleviate the itch, cough or asthma. Nav1.7-mediated itch as used herein refers to itch resulting at least partially from increased Nav1.7 channel activity.
  • Nav1.7-mediated cough as used herein refers to cough resulting at least partially from increased Nav1.7 channel activity.
  • Nav1.7-mediated asthma as used herein refers to asthma resulting at least partially from increased Nav1.7 channel activity.
  • Protoxin-II variants of the invention may be tested for their effect in reducing or alleviating pain and/or Nav1.7-mediated pain using animal models described herein, and models such as the rat spinal nerve ligation (SNL) model of neuropathic pain, carrageenan induced allodynia model, the Freund's complete adjuvant (CFA)-induced allodynia model, the thermal injury model, the formalin model and the Bennett Model, and other models as described in U.S. Pat. Appl. No. 2011/0124711 and U.S. Pat. No. 7,998,980. Carrageenan induced allodynia and CFA-induced allodynia are models of inflammatory pain. The Bennett model provides an animal model for chronic pain including post-operative pain, complex regional pain syndrome, and reflex sympathetic dystrophy.
  • Any of the foregoing animal models may be used to evaluate the efficacy of Protoxin-II variants of the invention inhibitor in treating pain and/or Nav1.7-mediated pain. The efficacy of the Protoxin-II variants of the invention may be compared to a no treatment or placebo control. Additionally or alternatively, efficacy may be evaluated in comparison to one or more known pain-relieving medicaments.
  • The present invention provides methods of treating Nav1.7-mediated pain using the Protoxin-II variants of the invention. It has been discovered in the pending application by the inventors (U.S. Patent Application No. 61/781,276) that administration of Nav1.7 blocking peptides are efficacious in treating and/or alleviating pain in various animal models of pain, contrary to what was disclosed and suggested in the literature. While peptide inhibitors of Nav1.7 have been shown to be potent and/or selective towards Nav1.7 in in vitro cell culture models using overexpressed Nav1.7 or on isolated neurons in which the blood-nerve barrier is subverted through desheathing or hypertonic saline injection, they have so far proven non-efficacious in in vivo animal models of pain, where the lack of efficacy has been reported to result from the inability of the peptides to pass the blood-nerve barrier. Several publications describe lack of efficacy of Nav1.7 blocking peptides in animal models of pain or in isolated nerves. For example Hackel et al., Proc Natl Acad Sci 109:E2018-27, 2012, describes the inability of ProTx-II to inhibit action potential firing in isolated nerves unless the perineural barrier, which provides a diffusion barrier in this model, is compromised. ProTx-II was found non-efficacious in rodent models of acute and inflammatory pain; a likely explanation stated the inability of ProTx-II to cross the blood-nerve barrier (Schmalhofer et al., Mol Pharmacol 74:1476-1484, 2008). It has been proposed that Nav1.7 peptide toxin blockers have poor oral bioavailability and they are difficult to deliver to nerve endings, implying that their use as therapeutic agents remain limited (Dib-Hajj et al., Nature Rev Neuroscience 14, 49-62, 2013).
  • Nav1.7 is expressed in the peripheral nervous system e.g., in nociceptive dorsal root ganglions (DRG), most notably in nociceptive small-diameter DRG neurons, in particular in peripheral terminals in the skin, with little representation in the brain. Nav1.7 distribution (e.g. sensory ending) and physiology predispose it to a major role in transmitting painful stimuli.
  • One embodiment of the invention is a method of treating Nav1.7-mediated pain by administering a therapeutically effective amount of the Protoxin-II variant of the invention to a subject in need thereof for a time sufficient to treat the Nav1.7-mediated pain.
  • The Protoxin-II variants of the invention Nav1.7 may be utilized in any therapy where it is desired to treat Nav1.7mediated pain or other disorders of sensory or sympathetic neuron dysfunction. “Treat” or “treatment” of pain is meant to include partially or completely to prevent, stop, inhibit, reduce, or delay the perception of pain.
  • In some embodiments, the Nav1.7-mediated pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, post-operative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis, or pain associated with disease and degeneration.
  • Neuropathic pain includes for example painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN) or trigeminal neuralgia (TN). Other causes of neuropathic pain include spinal cord injuries, multiple sclerosis, phantom limb pain, post-stroke pain and HIV-associated pain. Conditions such as chronic back pain, osteoarthritis and cancer may also result in the generation of neuropathic-related pain and thus are potentially suitable for treatment with the Protoxin-II variants of the invention.
  • In another embodiment, the Nav1.7-mediated pain is associated with primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis or fibromyalgia.
  • In the methods of the invention, the Protoxin-II variants of the invention may be conjugated to a second polypeptide to form a fusion protein. Such fusion proteins are for example the well-known Fc fusions or fusions to human serum albumin to extend half-life of the peptide inhibitors. The conjugation may be a direct conjugation via a linker, such as a glycine-serine rich linker. Such linkers are well known in the art. The Protoxin-II variants of the invention incorporating additional moieties may be compared for their Nav1.7 blocking ability and efficacy in treatment or reducing pain using well known methods and those described herein.
  • Other disorders of sensory or sympathetic neuron dysfunction that can be treated with the Protoxin-II variants of the invention, including asthma, cough, heart-burn, itch, dermatitis, bladder instability, and Reynaud's disease.
  • Pharmaceutical Compositions
  • The Protoxin-II variants of the invention may be formulated in a pharmaceutically acceptable vehicle or carrier. One embodiment of the invention is a pharmaceutical composition comprising the isolated Protoxin-II variant of the invention and a pharmaceutically acceptable excipient.
  • A suitable vehicle or carrier may be water for injection, physiological saline solution or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. These solutions are sterile and generally free of particulate matter, and may be sterilized by conventional, well-known sterilization techniques (e.g., filtration). The compositions may contain pharmaceutically acceptable excipients as required to approximate physiological conditions, such as pH adjusting and buffering agents, stabilizing, thickening, lubricating and coloring agents, etc. Suitable vehicles and their formulation and packaging are described, for example, in Remington: The Science and Practice of Pharmacy (21st ed., Troy, D. ed., Lippincott Williams & Wilkins, Baltimore, Md. (2005) Chapters 40 and 41).
  • In the methods of the invention, the Protoxin-II variants of the invention may be administered by peripheral administration. “Peripheral administration” or “administered peripherally” means introducing an agent into a subject outside of the central nervous system. Peripheral administration encompasses any route of administration other than direct administration to the spine or brain.
  • Peripheral administration can be local or systemic. Local administration may be used to concentrate the therapeutic to the site of action, such as local administration to joints, spinal cord, surgical wounds, sites of injury/trauma, peripheral nerve fibers, various organs GI, urogenital, etc.) or inflamed tissues. Systemic administration results in delivery of a pharmaceutical composition to essentially the entire peripheral nervous system of the subject and may also result in delivery to the central nervous system depending on the properties of the composition.
  • Routes of peripheral administration encompass, without limitation, topical administration, intravenous or other injection, and implanted mini-pumps or other extended release devices or formulations.
  • Pharmaceutical compositions of the invention include formulations involving the Protoxin-II variants of the invention in sustained- or controlled-delivery formulations. These formulations may be achieved through use of for example injectable microspheres, bio-erodible particles, microemulsions, nanoparticles, nanocapsules, macroemulsions, polymeric compounds (such as polyesters, polyamino acids, hydrogels, poly(lactic acid), polyglycolic acid or ethylene vinylacetate copolymers), beads or liposomes, hyaluronic acid or implantable drug delivery devices.
  • The Protoxin-II variants of the invention may be prepared for use for parenteral (subcutaneous, intramuscular or intravenous), intracerebral (intra-parenchymal), intracerebroventricular, intramuscular, intra-ocular, intra-arterial, intraportal, or intralesional routes; by sustained release systems or by implantation devices, or any other administration, particularly in the form of liquid solutions or suspensions; for buccal or sublingual administration such as in the form of tablets or capsules; or intranasally such as in form of powders, nasal drops or aerosols or certain agents; transdermally in a form of a gel, ointment, lotion, cream or dusting powder, suspension or patch delivery system with chemical enhancers to either modify the skin structure or to increase the drug concentration in the transdermal patch, or with agents that enable the application of formulations containing proteins and peptides onto the skin (Int. Pat. Publ. No. WO98/53847), or applications of electric fields to create transient transport pathways such as electroporation, or to increase the mobility of charged drugs through the skin such as iontophoresis, or application of ultrasound such as sonophoresis (U.S. Pat. Nos. 4,309,989 and 4,767,402). The composition also may be administered locally via implantation of a membrane, sponge or another appropriate material onto which the desired molecule has been absorbed or encapsulated.
  • In certain embodiments, where an implantation device is used, the device may be implanted into any suitable tissue or organ, and delivery of the desired molecule may be via diffusion, timed-release bolus, or continuous administration.
  • The concentration of the Protoxin-II variants of the invention or other peptide inhibitors of Nav1.7 in such pharmaceutical formulation can vary widely, for example from about 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1.0%, 1.1%, 1.2%, 1.3%, 1.4%, 1.5%, 1.6%, 1.7%, 1.8%, 1.9%, 2%, or between 2% to 5%, up to as much as 15%, 20%, 30%, 40%, 50%, 60% or 70% by weight and will be selected primarily based on fluid volumes, viscosities and other factors, according to the particular mode of administration selected. The Protoxin-II variants of the invention can be lyophilized for storage and reconstituted in a suitable vehicle prior to use. This technique has been shown to be effective with conventional protein preparations. Lyophilization and reconstitution techniques are well known in the art.
  • An exemplary pharmaceutical composition of the present invention may comprise Tris buffer of about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, and may further include sorbitol, sucrose, Tween-20 and/or a suitable substitute thereof.
  • The appropriate therapeutically effective dose may be determined readily by those skilled in the art. An effective dose refers to an amount or dosage sufficient to produce a desired result, i.e. to partially or completely prevent, stop, inhibit, reduce, or delay the perception of pain associated with any painful medical condition. The effective amount may vary depending on the specific vehicle and the Protoxin-II variants of the invention selected, and is also dependent on a variety of factors and conditions related to the subject to be treated and the severity of the pain. For example, factors such as age, weight and health of the subject to be administered with the pharmaceutical compositions of the invention as well as dose response curves and toxicity data obtained in preclinical animal work could be among those considered. A determined dose may, if necessary, be repeated at appropriate time intervals selected as appropriate by a physician or other person skilled in the relevant art (e.g. nurse, veterinarian, or veterinary technician) during the treatment period. The determination of an effective amount or a therapeutically effective amount for a given agent is well within the ability of those skilled in the art.
  • Thus, a pharmaceutical composition of the invention for intramuscular injection could be prepared to contain 1 mL sterile buffered water, and between about 1 ng to about 100 mg, about 50 ng to about 30 mg or about 5 mg to about 25 mg of a Protoxin-II variant of the invention. Similarly, a pharmaceutical composition of the invention for intravenous infusion could be made up to contain about 250 ml of sterile Ringer's solution, and about 1 mg to about 30 mg or about 5 mg to about 25 mg of the Protoxin-II variants of the invention. Actual methods for preparing parenterally administrable compositions are well known and are described in more detail in, for example, “Remington's Pharmaceutical Science,” 15th ed., Mack Publishing Company, Easton, Pa.
  • Further Embodiments of the Invention
  • Set out below are certain further embodiments of the invention according to the disclosures elsewhere herein. Features from embodiments of the invention set out above described as relating to the invention disclosed herein also relate to each and every one of these further numbered embodiments.
  • 1) An isolated Protoxin-II variant comprising the sequence X1X2X3CX4X5WX6QX7CX8X9X10X11X12CCX13X14FX15CX16LWCX17KKLW (SEQ ID NO: 403), wherein
  • X1 is G, P, A or deleted;
  • X2 is P, A or deleted;
  • X3 is S, Q, A, R or Y;
  • X4 is Q, R, K, A or S;
  • X5 is K, S, Q or R;
  • X6 is M or F;
  • X7 is T, S, R, K or Q;
  • X8 is D or T;
  • X9 is S, A or R;
  • X10 is E, R, N, K, T or Q;
  • X11 is R or K;
  • X12 is K, Q, S or A;
  • X13 is E, Q or D;
  • X14 is G or Q;
  • X15 is V or S;
  • X16 is R or T; and
  • X17 is K or R;
  • optionally having an N-terminal extension or a C-terminal extension,
    wherein the polypeptide inhibits human Nav1.7 activity with an IC50 value of about 1×10−7 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
  • 2) The Protoxin-II variant as described above, wherein the N-terminal extension comprises the amino acid sequence of SEQ ID NOs: 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384 or 385.
  • 3) The Protoxin-II variant as described above, wherein the C-terminal extension comprises the amino acid sequence of SEQ ID NOs: 374, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396 or 397.
  • 4) The Protoxin-II variant as described above, wherein the N-terminal and/or the C-terminal extension is conjugated to the Protoxin-II variant via a linker.
  • 5) The Protoxin-II variant as described above, wherein the linker comprises the amino acid sequence of SEQ ID NOs: 383, 392, 398, 399, 400, 401 or 402.
  • 6) The isolated Protoxin-II variant as described above, comprising the amino acid sequence of SEQ ID NOs: 30, 40, 44, 52, 56, 59, 65, 78, 109, 110, 111, 114, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 162, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 177, 178, 179, 180, 182, 183, 184, 185, 186, 189, 190, 193, 195, 197, 199, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 224, 226, 227, 231, 232, 243, 244, 245, 247, 249, 252, 255, 258, 261, 263, 264, 265, 266, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 332, 334, 335, 336, 337, 339, 340, 341, 342, 346, 351, 358, 359, 364, 366, 367, or 368.
  • 7) The isolated Protoxin-II variant as described above that inhibits human Nav1.7 activity with an IC50 value of about 3×10−8 M or less.
  • 8) The isolated Protoxin-II variant as described above that inhibits human Nav1.7 activity with an IC50 value of between about 3×10−8 M to about 1×10−9 M.
  • 9) The isolated Protoxin-II variant as described above comprising the amino acid sequence GPQCX1X2WX3QX4CX5X6X7X8X9CCX10X11FX12CX13LWCX14KKLW (SEQ ID NO: 404), wherein
  • X1 is Q, R, K, A or S;
  • X2 is K, S, Q or R;
  • X3 is M or F;
  • X4 is T, S, R, K or Q;
  • X5 is D or T;
  • X6 is 5, A or R;
  • X7 is E, R, N, K, T or Q;
  • X8 is R or K;
  • X9 is K, Q, S or A;
  • X10 is E, Q or D;
  • X11 is G or Q;
  • X12 is V or S;
  • X13 is R or T; and
  • X14 is K or R.
  • 10) The isolated Protoxin-II variant as described above, comprising the amino acid sequence of SEQ ID NOs: 56, 78, 111, 114, 117, 118, 119, 122, 123, 129, 130, 131, 132, 133, 134, 135, 136, 138, 139, 140, 141, 142, 145, 146, 147, 149, 150, 151, 152, 153, 154, 156, 158, 159, 165, 172, 173, 175, 177, 178, 183, 184, 185, 186, 189, 190, 193, 197, 199, 207, 210, 211, 216, 217, 224, 266, 273, 282 or 335.
  • 11) The isolated Protoxin-II variant as described above, wherein the variant selectively inhibits human Nav1.7.
  • 12) The isolated Protoxin-II variant as described above, comprising the sequence GPX1CQKWMQX2CDX3X4RKCCX5GFX6CX7LWCX8KKLW (SEQ ID NO: 405); wherein
  • X1 is Y, Q, A, S or R;
  • X2 is T or S;
  • X3 is S, R or A;
  • X4 is E, T or N;
  • X5 is E or Q;
  • X6 is V or S;
  • X7 is R or T; and
  • X8 is K or R.
  • 13) The isolated Protoxin-II variant as described above, comprising the amino acid sequence of SEQ ID NOs: 56, 59, 65, 78, 111, 114, 117, 118, 119, 121, 122, 123, 129, 130, 133, 150, 190, 217, 281, 324, 325 or 326.
  • 14) The isolated Protoxin-II variant as described above, comprising the sequence GPQCQKWMQX1CDX2X3RKCCX4GFX5CX6LWCX8KKLW (SEQ ID NO: 406); wherein
  • X1 is Tor S;
  • X2 is S, R or A;
  • X3 is E, T or N;
  • X4 is E or Q;
  • X5 is V or S;
  • X6 is R or T; and
  • X7 is K or R.
  • 15) An isolated Protoxin-II variant comprising the amino acid sequence that is 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO: 78 (GPQCQKWMQTCDRERKCCEGFVCTLWCRKKLW-COH), wherein
      • a) the amino acid sequence has Q at position 1, Q at position 7 and F at position 19, when residue numbering is according to SEQ ID NO: 1;
      • b) the polypeptide inhibits human Nav1.7 activity with an IC50 value of about 30×10−9 M or less, wherein the IC50 value is measured using a FLIPR® Tetra membrane depolarization assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−9 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and
      • c) the polypeptide selectively inhibits Nav1.7.
  • 16) The isolated Protoxin-II variant as described above having a free C-terminal carboxylic acid, amide, methylamide or butylamide group.
  • 17) An isolated fusion protein comprising the Protoxin-II variant as described above conjugated to a half-life extending moiety.
  • 18) The fusion protein of claim 17, wherein the half-life extending moiety is human serum albumin (HSA), albumin binding domain (ABD), Fc or polyethylene glycol (PEG).
  • 19) An isolated polynucleotide encoding a Protoxin-II variant as described above.
  • 20) A vector comprising the isolated polynucleotide as described above.
  • 21) A host cell comprising the vector as described above.
  • 22) A method of producing a isolated Protoxin-II variant, comprising culturing the host cell of as described above and recovering the Protoxin-II variant produced by the host cell.
  • 23) A pharmaceutical composition comprising an isolated Protoxin-II variant as described above and a pharmaceutically acceptable excipient.
  • 24) A method of treating Nav1.7-mediated pain in a subject, comprising administering to a subject in need thereof an effective amount of the Protoxin-II variant as described above to treat the pain.
  • 25) The method as described above, wherein the pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, postoperative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis.
  • 26) The method as described above, wherein the Protoxin-II variant is administered peripherally.
  • 27) The method as described above, wherein the Protoxin-II variant is administered locally to a joint, spinal cord, surgical wound, sites of injury or trauma, peripheral nerve fibers, urogenital organs, or inflamed tissues.
  • 28) The method as described above, wherein the subject is a human.
  • 29) The Protoxin-II variant as described above for use in treating pain in a subject in need thereof.
  • 30) The Protoxin-II variant for use as described above, wherein pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, postoperative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis.
  • 31) The Protoxin-II variant for use as described above, wherein the Protoxin-II variant is administered peripherally.
  • 32) The Protoxin-II variant for use as described above, wherein the Protoxin-II variant is administered locally to a joint, spinal cord, surgical wound, sites of injury or trauma, peripheral nerve fibers, urogenital organs, or inflamed tissues.
  • The present invention will now be described with reference to the following specific, non-limiting examples.
  • Example 1: Design and Generation of Protoxin-II Variants
  • Protoxin-II single position limited amino acid scanning library substitution was designed to assess to what degree selectivity, peptide yield, and homogeneity can be improved.
  • Protoxin-II variants were designed as HRV3C protease cleavable HSA fusion proteins in the following format from N- to C-terminus: 6×His-HSA-linker-HRV3C cleavable peptide-Protoxin-II variant (“6×His” disclosed as SEQ ID NO: 108); linker being (GGGGSGGGGSGGGGSGGGGS; SEQ ID NO: 80, HSA having the sequence of SEQ ID NO: 106, HRV3C cleavable peptide having the sequence of SEQ ID NO: 82). Each Protoxin-II variant, after cleavage from HSA had a residual N-terminal GP from the cleavage site.
  • The variants were characterized in membrane depolarization assays using FLIPR® Tetra as described in Example 3 FLIPR® Tetra membrane depolarization assay, and in whole cell patch clamp experiments using the QPatch assay as described in Example 3.
  • Combinatorial libraries were designed to test for additive effects of select single position hits in an attempt to generate Nav1.7 antagonists with further improved potency and selectivity profile compared to the native peptide.
  • Construction of the Expression Vectors
  • The designed Protoxin-II variant genes were generated using synthetic gene assembly technology as described in U.S. Pat. No. 6,521,427. The amino acid sequences of the designed peptide variants were back-translated to DNA sequences using human high-frequency codons. The DNA sequence of each variant gene, together with a portion of vector DNA including the DNA cloning sites, was synthesized as multiple oligonucleotides, some of which contained degenerate codons, and assembled into full-length DNA fragments. The assembled DNA fragments were amplified by PCR and PCR products were subsequently cloned as a pool. Pooled PCR products were digested with the appropriate restriction enzymes and cloned into the designed expression vector in such a manner as to fuse each toxin variant gene to the signal peptide and the fusion partner (6×His-HSA-linker-HRV3C cleavable peptide (“6×His” disclosed as SEQ ID NO: 108) contained in the vector. Standard molecular biology techniques were used to identify a positive clone for each designed variant. The plasmid DNA from these positive clones was purified and sequence confirmed before expressing the Protoxin-II peptide variant fusion proteins using standard methods.
  • Protein Expression
  • HEK 293-F cells were maintained in 293 Freestyle™ media (Invitrogen Cat #12338) and split when the cell concentration was between 1.5 and 2.0×106 cells per mL. The cells were grown in suspension, shaking at 125 RPM in a humidified incubator set at 37° C. and 8% CO2. HEK 293F cells were transiently transfected using a DNA/lipid complex after they were diluted to 1.0×106 cells per mL. To generate the complex, 1.25 μg DNA per mL of transfection was diluted in 1.0 ml of OptiPro media (Invitrogen Cat #12309) and 1.25 ml of Freestyle™ Max transfection reagent (Invitrogen Cat #16447) was diluted in 1.0 mL of OptiPro media. The DNA and Max transfection reagent were mixed together and incubated for 10 minutes at room temperature before adding to the cells. Transfected cells were placed in a humidified incubator set at 37° C. and 8% CO2 for 4 days shaking at 125 RPM. The supernatant was separated from the cells by centrifugation at 5,000×g for 10 minutes and filtered through a 0.2 μm filter (Corning; Cat #431153), then concentrated 10 and 50 fold using an Amicon Ultra Concentrator 10K (Cat #UFC901096), and centrifuging for approximately 10 minutes at 3,750×g.
  • Example 2: Purification of Protoxin-II Variants
  • Protoxin-II variants were expressed as HSA fusion proteins as indicated in Example 1 and the Protoxin-II variant peptides were cleaved with HRV3C protease prior to purification. Two methodologies were tested for efficient purification of the Protoxin-II variants.
  • Protein Purification Purification of Protoxin-II Variants by RP-HPLC
  • The secreted proteins were purified from the expression supernatants via IMAC using 1 ml HisTrap HP columns (GE Healthcare Cat #17-5247-01). The chromatography method was run using an AKTA Xpress and protein was eluted from the column using a step gradient of imidazole. Peak fractions were pooled and digested overnight with HRV 3C protease (1 μg protease/150 μg fusion).
  • Cleaved peptide-fusion pools were further purified using a Dionex HPLC system with a reverse phase Phenomenex Luna 5 μm C18(2) column (Cat #00B-4252-PO-AX). Samples were eluted from the column with a 0-68% Acetonitrile (0.05% TFA) linear gradient. Elution fractions were pooled, lyophilized overnight and reconstituted in HEPES buffered saline, pH 7.4 (10 mM HEPES, 137 mM NaC1, 5.4 mM KC1, 5 mM glucose, 2 mM CaC12, 1 mM MgC12).
  • Table 4 shows yields of Protoxin-II variants purified by RP-HPLC. The average mg yield/L was 0.01615.
  • TABLE 4
    Protoxin-II Variant yield Protoxin-II Variant yield
    Peptide ID (mg) Peptide ID (mg)
    NV1D816 0.0008 NV1D2496 0.0006
    NV1D2511 0.0009 NV1D2503 0.0030
    NV1D2513 0.0034 NV1D766 0.0054
    NV1D2504 0.0071 NV1D770 0.0040
    NV1D2260 0.0129 NV1D772 0.0015
    NV1D2498 0.0079 NV1D792 0.0016
    NV1D2499 0.0076 NV1D815 0.0008
    IW1D2512 0.0061 NV1D768 0.0060
    NV1D2267 0.0095 NV1D2508 0.0017
    NV1D2507 0.0000 NV1D2501 0.0008
    NV1D2509 0.0000 NV1D2296 0.0018
    NV1D2305 0.0001 NV1D2292 0.0059
    NV1D815 0.0021 NV1D750 0.0023
    NV1D2506 0.0001 NV1D748 0.0036
    NV1D2505 0.0006 NV1D774 0.0050
    NV1D812 0.0001 NV1D786 0.0036
    NV1D2510 0.0009 NV1D855 0.0008
    NV1D769 0.0031 NV1D2312 0.0011
    NV1D2497 0.0038 NV1D1410 0.0074
    NV1D2500 0.0004 NV1D1415 0.0128
    NV1D767 0.0004 NV1D751 0.0033
    NV1D2502 0.0002
  • Purification of Protoxin-II Variants by Solid Phase Extraction (SPE)
  • The secreted proteins were purified from the expression supernatants via IMAC using 1 mL HisTrap HP columns (GE Healthcare Cat #17-5247-01). The chromatography method was run using an AKTA Xpress and protein was eluted from the column using a step gradient of imidazole. Peak fractions were pooled and digested overnight with HRV3C protease (1 μg protease/150 μg fusion). The cleaved sample was loaded into a 50 kDa molecular weight cut off centrifugal filter unit (Millipore UFC805096) and cleaved peptide collected in the filtrate fraction.
  • Peptide pools were loaded onto a 96-well solid phase extraction block (Agilent Bond Elut Plexa A3969030) for further purification, desalting, and concentration. Blocks were used in conjunction with a vacuum manifold (Whatman). Peptide samples were loaded and washed in 0.05% TFA in water and eluted with a step gradient of acetonitrile with 0.05% TFA in water. Elution fractions were then lyophilized overnight and reconstituted in HEPES buffered saline, pH 7.4 (10 mM HEPES, 137 mM NaC1, 5.4 mM KC1, 5 mM glucose, 2 mM CaC12, 1 mM MgCl2).
  • Peptides were reconstituted in supplemented HEPES buffered saline, pH 7.4 (10 mM HEPES, 137 mM NaC1, 5.4 mM KC1, 5 mM glucose, 2 mM CaC12, 1 mM MgC12) and absorbance was measured at 280 nm. Concentration values were then calculated using each sample's extinction coefficient. 2 μg of each peptide were loaded onto an Invitrogen NuPAGE® Novex® Bis-Tris Gel 15 well gel and run in MES buffer non-reduced.
  • Samples were analyzed on Agilent 1100 HPLC using 4-80% acetonitrile in 0.05% TFA linear gradient with a Phenomenex Luna C18(2) analytical column (Cat #00A-4041-B0). Concentrations of all peptides were normalized and 10 μL of each were injected for a total of 1.3 μg per sample. Absorbance at 220 nm was monitored and chromatograms analyzed were using Chromeleon software.
  • Table 5 shows yields (mg) of Protoxin-II variants purified by SPE. The average mg yield/L was 0.05353.
  • The benefits of the SPE purification process are ease and throughput of purification since samples are processed in parallel in a 96-well block rather than serially on RP-HPLC, and improvement in yield. There was, on average, more than 3-fold higher yield (mg/L) for variants purified by SPE versus RP-HPLC.
  • TABLE 5
    Protoxin-II Variant yield Protoxin-II Variant yield
    Peptide ID (mg) Peptide ID (mg)
    NV1D12 0.0054 NV1D2734 0.0602
    NV1D2659 0.0234 NV1D2772 0.2050
    NV1D2664 0.0060 NV1D2775 0.2225
    NV1D2666 0.0225 NV1D2738 0.0512
    NV1D2708 0.0721 NV1D2740 0.0373
    NV1D2725 0.0144 NV1D2733 0.1913
    NV1D2739 0.0053 NV1D788 0.0000
    NV1D2765 0.0097 NV1D757 0.0021
    NV1D2748 0.0995 NV1D791 0.0007
    NV1D2771 0.0103 NV1D2310 0.0011
    NV1D2770 0.0121 NV1D2308 0.0014
    NV1D2778 0.0644 NV1D778 0.0019
    NV1D2782 0.0202 NV1D2294 0.0000
    NV1D2756 0.0466 NV1D856 0.0047
    NV1D2759 0.0218 NV1D2309 0.0023
    NV1D2712 0.0558 NV1D846 0.0020
    NV1D12 0.0127 NV1D2896 0.0504
    NV1D2673 0.0625 NV1D2913 0.0203
    NV1D2662 0.0433 NV1D2910 0.0253
    NV1D2669 0.2661 NV1D2893 0.0569
    NV1D2665 0.0389 NV1D2909 0.0195
    NV1D2731 0.2547 NV1D2917 0.0339
    NV1D2767 0.0238 NV1D2914 0.0201
    NV1D2730 0.2566 NV1D2922 0.0554
    NV1D2766 0.0198 NV1D2902 0.0061
    NV1D2667 0.0050 NV1D2889 0.0022
    NV1D2769 0.0142 NV1D2887 0.0025
    NV1D2719 0.0675 NV1D2878 0.0272
    NV1D2776 0.0633 NV1D2877 0.0129
    NV1D2663 0.0344 NV1D2851 0.0029
    NV1D2709 0.1841 NV1D2850 0.0026
    NV1D2720 0.0538 NV1D2820 0.0020
    NV1D12 0.0095 NV1D2819 0.0015
    NV1D2773 0.1921 NV1D2814 0.0163
    NV1D2810 0.0086 NV1D2918 0.0256
    NV1D2732 0.0262 NV1D2921 0.0533
    NV1D757 0.0026 NV1D2905 0.0126
    NV1D791 0.0206 NV1D2906 0.0189
    NV1D2310 0.0085 NV1D2881 0.0207
    NV1D2308 0.0179 NV1D2882 0.0223
    NV1D778 0.0094 NV1D2869 0.0038
    NV1D856 0.0247 NV1D2870 0.0187
    NV1D2309 0.0035 NV1D2867 0.0147
    NV1D846 0.0043 NV1D2888 0.0045
    NV1D2889 0.0107 NV1D2816 0.0133
    NV1D2887 0.0061 NV1D2885 0.0025
    NV1D2861 0.0469 NV1D2974 0.0418
    NV1D2729 0.1101 NV1D2972 0.1089
    NV1D2890 0.0088 NV1D2971 0.0407
    NV1D2899 0.0402 NV1D2970 0.0557
    NV1D2804 0.0044 NV1D2969 0.0799
  • Example 3: Characterization of Protoxin-II Variants
  • Select Protoxin-II variants were characterized in membrane depolarization and whole cell patch clamp assays to assess their potency and selectivity towards Nav1.7.
  • FLIPR® Tetra Membrane Depolarization Assay
  • The ability of the generated peptides to inhibit membrane depolarization induced by Nav1.7 agonist veratridine (3-veratroylveracevine; Biomol, Catalog #NA125) was measured with a FRET (fluorescence resonance energy transfer) assay on FLIPR® Tetra using DISBAC2(3) (Invitrogen, K1018) as an electron acceptor and PTS18 (Trisodium 8-octadecyloxypyrene-1,3,6-trisulfonate) (Sigma) as a donor by exciting the donor at 390-420 nm and measuring FRET at 515-575 nm.
  • HEK293 cells stably expressing human Nav1.7 were cultured in DMEM/F-12 media (1:1), supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 400 μg/mL geneticin and 100 μM NEAAs (all reagents from Invitrogen). 50 μL of harvested cells were plated at 25,000 cells/well into poly-lysine coated 384-well black clear bottom plates. The plates were incubated at room temperature (RT) for 15 min followed by an overnight incubation at 37° C. All incubations were done in the dark unless otherwise stated. The next day, the wells were washed 4 times with assay buffer (137 mM NaC1, 4 mM KC1, 2 mM MgC12, 2 mM CaC12, 5 mM Glucose, 10 mM HEPES, pH 7.4), and resuspended in 25 μL of assay buffer. 2× stock (6 μM) of the PTS18 dye was prepared by suspending the dye in 10% Pluronic F127 in DMSO at 1:1 (v/v ratio). 25 μL of the 2×PTS18 stock was added into the wells and the cells were stained for 30 min at RT, after which the dye was washed off with the assay buffer. Peptides were suspended at 3× their final concentration in the assay buffer containing 10 μM DISBAC2(3) and 400 μM VABSC-1 to suppress background fluorescence (Sigma, cat #201987). 25 μL/well of the suspended peptides were added into each well, and incubated for 60 minutes at RT. Depolarization was induced by 25 μM final concentration of veratridine (by adding 25 μL/well of 75 μM (3×) stock solution), and the reduction in the mean intensity of FRET dye fluorescence was measured 30-100 seconds after adding the agonist. A 1.3× dilution of each measured peptide occurred after adding veratridine by convention, the concentration at the beginning of the FLIPR® Tetra assay is reported.
  • Concentration-response curves of synthetic Protoxin-II (Peptide International) were constructed in each experimental series and were used as controls. Fluorescence counts for each well were converted to % response by normalizing the signal to the difference between negative control (response to agonist veratridine alone) and positive control (response to veratridine in the presence of 10 μM tetracaine) values. For measurements, “spatial uniformity correction” (all fluorescence traces are normalized to the average initial starting intensity) and “subtract bias value” (subtract the initial starting intensity from each trace) were turned on in FLIPR® Tetra. Each data point represented the response in an individual well. All individual data points were used in a non-linear least-squares fitting procedure to find the best fit to a Hill function using Origin (Microcal). IC50 values were extracted from the resultant fitted curve. The mean responses of the positive (P) and negative (N) controls were used to calculate the % response in a well as follows: % response=100*(N−R)/(N−P).
  • Assay plates were accepted if the potency of control antagonists for that day were within ±0.5 log units of their historical mean.
  • QPatch Assay
  • HEK293 cells stably expressing human Nav1.5 (SEQ ID NO: 105), Nav1.7 (SEQ ID NO: 79) or Nav1.6 (SEQ ID NO: 407) were cultured in DMEM/F-12 media (1:1), supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 400 μg/mL Geneticin and 100 μM NEAAs (all reagents from Invitrogen). Cells were maintained at 37° C. and in 5% CO2 and assayed upon reaching ˜50-90% confluency. CHO cells stably expressing human Nav1.6 in a tetracycline-inducible manner (SEQ ID NO: 407) were cultured in HAMs F12, supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin, 10 μg/mL Blasticidin and 400 μg/mL Zeocin. Cells were maintained at 37° C. and in 5% CO2, and assayed upon reaching ˜50-90% confluency. Nav1.6 expression was induced with 1 μg/ml of tetracycline, 24-48 h prior to an experiment.
  • Before testing in QPatch HT (Sophion), cells were first dissociated using 0.05% trypsin (5 min at 37° C.), resuspended in CHO-S-SFM media (Life Technologies) and gently triturated to break up cell clumps. Cell density was adjusted to 1-2×106/mL with the same media and cells were the transferred to a cell “hotel” in QPatch HT and used in experiments for several hours. For giga-ohm seal formation and whole-cell patch clamp recording, the extracellular solution contained 137 mM NaC1, 5.4 mM KC1, 1 mM MgC12, 2 mM CaC12, 5 mM glucose, and 10 mM HEPES, pH=7.4 and osmolarity=315 mOsm. The intracellular solution contained 135 mM CsF, 10 mM CsCl, 5 mM EGTA, 5 mM NaC1 and 10 mM HEPES, pH=7.3 and osmolarity=290 mOsm. The voltage protocol used in the assay was as follows. From a holding potential of −75 mV (Nav1.7), −60 mV (Nav1.6), or −105 mV (Nav1.5) cells were first hyperpolarized to −120 mV for 2 sec and then depolarized to 0 mV for 5 ms before returning to the holding potential. This protocol was repeated once every 60 sec during liquid applications (see below). Cells were otherwise held at the holding potential when the above voltage protocol was not executed. Upon establishment of the whole-cell recording configuration, a total of five applications of the extracellular solution (all containing 0.1% bovine serum albumin (BSA) with or without test compound, except for the last application, which contained 1 μM TTX or 10 mM lidocaine as a positive control) were made on to cells being recorded. The first liquid application contained only the control buffer (5 μL). The voltage protocol was executed 10 times (for a total duration of 10 min) five seconds after the application. The next three liquid applications (5 μL each) contained a test compound (same compound at the same concentration for all three applications) or control buffer (for control cells only). Five seconds after each of these applications, the voltage protocol was again executed 10 times (also once per minute). The last application contained positive (composed of three 10 μL sub-applications, each separated by 2 seconds), five seconds after which the same voltage protocol was executed twice to obtain the baseline current. Currents were sampled at 25 kHz and filtered at 5 kHz with an 8-pole Bessel filter. The series resistance compensation level was set at 80%. For each cell, the peak current amplitude at 0 mV for each current trace in the first four liquid applications was first subtracted from that of the last trace in the presence of positive control and then normalized to that of the last trace in the first (control buffer) application as % inhibition. To control for current rundown, this (% inhibition) value for each cell in the presence of a test compound was further normalized to the average % inhibition value for control (typically 5-6) cells in the same experiment. The mean of the last two such values in the last compound application (i.e., the corrected % inhibition value for each concentration of a test compound) were taken as the % inhibition value for each cell at the particular compound concentration tested. The % inhibition values for all cells tested at each compound concentration were averaged and used in concentration response calculations. All experiments were performed at room temperature (−22° C.). Data are expressed as mean±SE. Wild type Protoxin-II was included in each experiment as a positive control. Data were accepted only if the potency of Protoxin-II was within ±0.5 log units of its historical mean.
  • IC50 values for Nav1.7 for select Protoxin-II variants obtained using the FLIPR® Tetra assay are shown in Table 6.
  • TABLE 6
    Protoxin-II
    Protoxin-II variant hNav1.7
    Variant Peptide TETRA
    Protein ID Peptide ID SEQ ID NO: IC50 (nM)
    NV1D12 5 NV1D12 2 4.1 ± 3.6
    NV1G1045 NV1D791 11 4.8 ± 0.4
    NV1D1332 1 NV1D1332 12 6.7 ± 0.5
    NV1D1336 1 NV1D1336 14 10.5 ± 1.2 
    NV1D1337 1 NV1D1337 15 10.3 ± 1.0 
    NV1G1049 NV1D2308 16 4.5 ± 0.4
    NV1G953 NV1D2670 17 22.2 ± 3.3 
    NV1G951 NV1D2674 18 4.0 ± 0.2
    NV1G963 NV1D2671 20 31.5 ± 6.4 
    NV1G949 NV1D2675 21 4.3 ± 0.3
    NV1G977 NV1D2665 22 4.9 ± 0.4
    NV1G957 NV1D2668 23 17.5 ± 2.6 
    NV1G965 NV1D2672 24 4.5 ± 0.3
    NV1G973 NV1D2662 25 4.0 ± 0.4
    NV1G975 NV1D2669 26 18.4 ± 5.7 
    NV1G971 NV1D2673 27 4.3 ± 0.5
    NV1G995 NV1D2663 28 4.2 ± 0.4
    NV1G961 NV1D2676 29 26.5 ± 2.9 
    NV1G911 NV1D2666 30 66.5 ± 36.7
    NV1G1133 NV1D2816 31  667 ± 93.6
    NV1G905 NV1D2735 32 60.0 ± 16.2
    NV1G979 NV1D2731 34 20.7 ± 7.2 
    NV1G1097 NV1D2810 35  339 ± 5750
    NV1G1099 NV1D2732 36  126 ± 26.9
    NV1G1011 NV1D2740 37 3.6 ± 9.9
    NV1G1105 NV1D2729 39 8.0 ± 0.9
    NV1G1013 NV1D2733 40 7.5 ± 2.9
    NV1G1095 NV1D2814 41  754 ± 51.3
    NV1G983 NV1D2730 43 25.5 ± 4.3 
    NV1G1003 NV1D2734 44 13.4 ± 0.8 
    NV1G1009 NV1D2738 45 2.6 ± 0.2
    NV1G1129 NV1D2867 49 >1000
    NV1G1121 NV1D2881 50  488 ± 72.2
    NV1G1123 NV1D2882 51  857 ± 65.7
    NV1G899 NV1D2774 52 50.5 ± 15.2
    NV1G1103 NV1D2861 54 >1000
    NV1G1127 NV1D2870 55  784 ± 84.8
    NV1G1007 NV1D2775 56 25.4 ± 2.0 
    NV1G1067 NV1D2893 57 75.5 ± 10.5
    NV1G1005 NV1D2772 59 15.6 ± 1.8 
    NV1G1061 NV1D2896 60 80.3 ± 7.1 
    NV1G1085 NV1D2877 61  441 ± 73.3
    NV1G1083 NV1D2878 62  680 ± 40.7
    NV1G1079 NV1D2889 64 12.1 ± 1.5 
    NV1G1001 NV1D2773 65 18.8 ± 1.5 
    NV1G1107 NV1D2890 66 25.8 ± 4.2 
    NV1G1109 NV1D2899 67 33.3 ± 6.7 
    NV1G1117 NV1D2905 68  713 ± 87.3
    NV1G1119 NV1D2906 69  940 ± 86.7
    NV1G1115 NV1D2921 70  586 ± 71.7
    NV1G1075 NV1D2922 71  204 ± 45.7
    NV1G1069 NV1D2909 72 97.1 ± 10.1
    NV1G1065 NV1D2910 73  441 ± 41.7
    NV1G1063 NV1D2913 74 79.7 ± 9.3 
    NV1G1073 NV1D2914 75 135 ± 7.8 
    NV1G1071 NV1D2917 76  197 ± 48.3
    NV1G1113 NV1D2918 77  983 ± 98.7
    NV1G1153 NV1D3034 78 10.3 ± 2.1 
  • Select Protoxin-II variants were tested for selectivity against human Nav1.5 using QPatch. IC50 values for both Nav1.7 and Nav1.5 for select peptides obtained using QPatch are shown in Table 7.
  • TABLE 7
    Protoxin-II
    Protoxin-11 variant hNav1.7 hNav1.5
    Variant Peptide QPatch
    Protein ID Peptide ID SEQ ID NO: IC50 (nM) IC50 (nM)
    NV1D12 5 NV1D12 2 2.2 ± 1.3 >1000
    NV1G899 NV1D2774 52 18.7 ± 13.6 >3000
    NV1G1007 NV1D2775 56 4.0 ± 8.9 >3000
    NV1G1005 NV1D2772 59 6.2 ± 3.2 >3000
    NV1G1001 NV1D2773 65 4.3 ± 3.3 >3000
    NV1G1153 NV1D3034 78 4.3 ± 4.3 >1000
  • Example 4: Generation and Characterization of Combinatorial Protoxin-II Variants
  • Combinatorial libraries were designed to test for additive effects of select single position hits in an attempt to generate Nav1.7 antagonists with further improved potency and selectivity profile compared to the native peptide using several approaches.
  • A limited amino acid scan was conducted at all noncysteine Protoxin-II positions using A, D, Q, R, K and S for diversification. In these experiments, Protoxin-II was expressed and tested as monovalent Fc fusion protein as described in Example 1. From this scan, substitutions Y1Q, W7Q, S11A, were identified that improved potency and/or selectivity of the resulting variants.
  • A full amino acid scan (excluding Cys and Trp) at positions M6 and M19 was also conducted. M19F substitution was identified from this scan that improved potency and/or selectivity of the resulting variants.
  • Protoxin-II/Huwentoxin-IV single position chimeras were designed bidirectionally. The purpose of this library was to obtain Protoxin-II variants that retained potency and selectivity profile of the wild type Protoxin-II and would achieve beneficial refolding properties associated with Huwentoxin-IV. Substitutions R22T and E12N were identified from this scan.
  • Peptide NV1G1153 was further engineered by diversifying position Y1 by a limited amino acid scan using R, K, T, A, D, E, Q and S, and by charge cluster engineering, where all sets of charged residues in the three-dimensional structure of the peptide (D10/E12, K4/E17, D10/E12/R13) were mutated.
  • N- and C-terminal extensions were introduced to select peptides, including NV1 G1153 with the purpose of improving peptide distribution to the site of action and of improving half-life of the peptides without significantly increasing the molecular weight of the resulting peptide. The N- and C-terminal extensions that were used are shown in Table 8 and 9, respectively, and are described in Oi et al., Neuroscience Letters 434, 266-272, 2008; Whitney et al., Nature Biotechnology 2011 29:4, 352-356; Sockolosky et al., (2012) 109:40, 16095-16100. Cell penetrating peptides HIV Tat and polyarginine were also used. Various linkers were used to couple the Protoxin-II variant to the N- and/or C-terminal extensions. The linkers used are shown in Table 10.
  • Protoxin-II variants from each campaign were tested for their potency and selectivity for Nav1.7 using methods described in Example 3. The amino acid sequences of the variants that inhibited Nav1.7 with an IC50 value of 200 nM or less are shown in Table 3. Table 11 shows the amino acid substitutions in select variant when compared to the wild type Protoxin-II, and the IC50 values for Nav1.7 inhibition in the FLIPR Tetra assay.
  • TABLE 8
    N-terminal extension
    Amino acid sequence SEQ ID NO:
    GPAAAAA 372
    GPAPAPA 373
    GGGGG 374
    GPCCNCSSKWCRDHSRCC 375
    GPSPGARAF 376
    GPDGPWRKM 377
    GPFGQKASS 378
    GPCRTIGPSVC 379
    GPSHSNTQTLAKAPEHTG 380
    GPQRFVTGHFGGLYPANG 381
    GPGWCGDPGATCGKLRLYCCSGFCDSYTKTCKDKSSA 382
    APAPAPAPAP 383
    GPYGRKKRRQRRR 384
    GPRRRRRRRRRRR 385
  • TABLE 9
    C-terminal extensions
    Amino acid sequence SEQ ID NO:
    CRTIGPSVC 386
    YGRKKRRQRRR 387
    GGGGG 374
    DGPWRKM 388
    CCNCSSKWCRDHSRCC 389
    RRRRRRRRRRR 390
    SHSNTQTLAKAPEHTG 391
    APAPA 392
    AAAAA 393
    FGQKASS 394
    QRFVTGHFGGLYPANG 395
    SPGARAF 396
    GPGWCGDPGATCGKLRLYCCSGFCDAYTKTCKDKSSA 397
  • TABLE 10
    Linkers
    Amino acid sequence SEQ ID No:
    GSAPAPAPAPAPGS 398
    GSAPAPAPAPAPAPAPAPAPAPGS 399
    GGGGSAPAPAPAPAPAPAPAPAPAPAPAPA 400
    PAPAPGGGGS
    APAPA 392
    GSGGGGAPAPAPAPAPAPAPAPAPAPGGGGSGS 401
    APAPAPAPAP 383
    APAPAPAPAPAPAPAPAPAP 402
  • TABLE 11
    Protoxin-II Nav1.7
    Protein variant Protein IC50
    name peptide name SEQ ID NO: Substitutions (nM) SE
    NV1G1728 NV1D3541 281 Y1A, W7Q, 9.4 1.2
    S11R, E12N, M19F
    NV1G1870 NV1D3583 321 Y1A, W7Q, S11A, E12R, 13.1 1.57
    M19F, V20S
    NV1G1752 NV1D3532 272 Y1A, W7Q, S11A, E12K, 17.3 2
    M19F, R22T, K26R
    NV1G1749 NV1D3587 326 Y1A, W7Q, S11A, 18.3 2.6
    E12N, M19F, V20S
    NV1G1725 NV1D3572 310 Y1A, W7Q, S11A, E12R, 19.8 2.2
    M19F, R22T
    NV1G1745 NV1D3537 277 Y1A, W7Q, S11A, E12K, 21.4 4.1
    M19F, V20S, R22T, K26R
    NV1G1720 NV1D3565 304 Y1A, W7Q, S11A, E12R, 23 2.8
    M19F, V20S, R22T
    NV1G1761 NV1D3550 290 Y1A, W7Q, S11R, M19F, 25.8 2.7
    R22T, K26R
    NV1G1746 NV1D3576 314 Y1A, W7Q, S11A, E12N, 26.7 5.2
    M19F, R22T
    NV1G979 NV1D2731 34 Y1A, W7Q, S11A 20.7 7.2
    NV1G953 NV1D2670 17 Y1A, W7Q 22.2 3.3
    NV1G1519 NV1D3006 133 Y1Q, W7Q, S11A, 4.03 1.05
    E12R, M19F
    NV1G1007- NV1D2775- 111 Y1Q, W7Q, S11A, M19F 5.06 0.473
    NH2 NH2
    NV1G1517 NV1D3004 131 Y1Q, W7Q, S11R, M19F 6.23 1.56
    (-GP) N—Ac— (-GP) N—Ac— 114 Y1Q, W7Q, S11A, M19F, 6.43 1.06
    NV1G1137- NV1D2974- V20S, R22T
    NH2 NH2
    NV1G1776 NV1D3339 172 Y1Q, Q3R, W7Q, S11R, 6.57 0.675
    M19F, R22T, K26R
    NV1G1153- NV1D3034- 119 Y1Q, W7Q, S11R, M19F, 7.1 0.9
    NH-methyl NH-methyl R22T, K26R
    (-GP) N—Ac— (-GP) N—Ac— 121 Y1Q, W7Q, S11R, M19F, 7.63 1.04
    NV1G1153- NV1D3034- R22T, K26R
    NH2 NH2
    NV1G1523 NV1D3012 135 Y1Q, W7Q, S11R, E12N, 7.74 0.904
    M19F
    NV1G1515 NV1D3005 132 Y1Q, W7Q, S11A, E12N, 7.83 1.38
    M19F
    NV1G1187 NV1D3015 138 Y1Q, W7Q, S11R, M19F, 8.86 2.28
    K26R
    NV1G1521 NV1D3018 141 Y1Q, W7Q, S11A, E12N, 9.79 2.91
    M19F, K26R
    NV1G1267 NV1D3044 150 Y1Q, W7Q, S11R, E12N, 9.8 0.849
    M19F, R22T, K26R
    NV1G1153 NV1D3034 78 Y1Q, W7Q, S11R, M19F, 10.3 2.14
    R22T, K26R
    NV1G1836 NV1D3359 190 Y1Q, W7Q, T8S, S11R, M19F, 10.5 0.739
    R22T, K26R
    NV1G1593 NV1D3050 153 Y1Q, W7Q, S11R, E12K, 10.8 1.3
    M19F
    NV1G1215 NV1D3048 152 Y1Q, W7Q, S11A, E12K, M19F 11.1 1.05
    NV1G1868 NV1D3353 185 Y1Q, W7Q, T8R, S11R, M19F, 11.2 1.25
    R22T, K26R
    NV1G1525 NV1D3013 136 Y1Q, W7Q, S11R, E12R, 11.3 1.83
    M19F
    NV1G1775 NV1D3340 173 Y1Q, Q3K, W7Q, S11R, M19F, 11.5 0.798
    R22T, K26R
    NV1G1833 NV1D3381 210 Y1Q, W7Q, S11R, K14Q, 12.2 1.56
    M19F, R22T, K26R
    NV1G1153- NV1D3034- 117 Y1Q, W7Q, S11R, M19F, 12.2 1
    NH2 NH2 R22T, K26R
    NV1G1777 NV1D3342 175 Y1Q, Q3A, W7Q, S11R, M19F, 12.8 2.67
    R22T, K26R
    NV1G1259 NV1D3058 158 Y1Q, W7Q, S11A, E12K, 12.9 1.29
    M19F, R22T, K26R
    NV1G1511 NV1D3032 146 Y1Q, W7Q, S11R, E12N, 13 203
    M19F, K26R
    NV1G1527 NV1D3031 145 Y1Q, W7Q, S11R, E12R, 13 1.36
    M19F, R22T
    NV1G1265 NV1D3062 159 Y1Q, W7Q, S11R, E12K, 13.2 1.43
    M19F, R22T, K26R
    NV1G1781 NV1D3388 217 Y1Q, W7Q, S11RE17Q, 13.5 1.14
    M19F, R22T, K26R
    NV1G1824 NV1D3354 186 Y1Q, W7Q, T8K, S11R, M19F, 13.9 1.12
    R22T, K26R
    NV1G1772 NV1D3352 184 Y1Q, K4S, W7Q, S11R, M19F, 14.2 2.01
    R22T, K26R
    NV1G1509 NV1D3033 147 Y1Q, W7Q, S11R, E12R, 14.5 2.18
    M19F, K26R
    NV1G1779 NV1D3351 183 Y1Q, K4Q, W7Q, S11R, M19F, 15.3 2.39
    R22T, K26R
    NV1G1687 NV1D3526 266 Y1Q, W7Q, S11R, M19F, 15.4
    R22T, K26R
    NV1G1269 NV1D3045 151 Y1Q, W7Q, S11R, E12R, 15.6 1.39
    M19F, R22T, K26R
    NV1G1623 NV1D3056 156 Y1Q, W7Q, S11R, E12K, 16.2 2.99
    M19F, R22T
    NV1G1859 NV1D3376 205 Y1Q, W7Q, S11R, K14R, 16.3 2.53
    M19F, R22T, K26R
    NV1G1153- NV1D3034- 118 Y1Q, W7Q, S11R, M19F, 16.6 1.4
    NH-butyl NH-butyl R22T, K26R
    NV1G1211 NV1D3036 149 Y1Q, W7Q, S11A, E12R, 17.2 1.55
    M19F, R22T, K26R
    NV1G1885 NV1D3254 165 Y1Q, W7Q, S11A, M19F 17.5 2.45
    NV1G1730 NV1D3542 282 Y1Q, W7Q, S11R, E12N, 17.7 2.5
    M19F, V20S, R22T, K26R
    NV1G1263 NV1D3051 154 Y1Q, W7Q, S11A, E12K, 17.9 1.78
    M19F, R22T
    NV1G1818 NV1D3368 122 Y1Q, W7Q, S11R, E12T, 17.9 1.89
    M19F, R22T, K26R
    NV1G1153 NV1D3034 116 Y1Q, W7Q, S11R, M19F, 18 2.5
    (synthetic) R22T, K26R
    NV1G1823 NV1D3367 197 Y1Q, W7Q, S11R, E12Q, 18.6 2.17
    M19F, R22T, K26R
    NV1G1820 NV1D3362 193 Y1Q, W7Q, D1OT, S11R, 20.1 2.32
    M19F, R22T, K26R
    NV1G1811 NV1D3369 199 Y1Q, W7Q, S11R, R13K, 20.4 2.44
    M19F, R22T, K26R
    NV1G1810 NV1D3358 189 Y1Q, W7Q, T8Q, S11R, M19F, 20.5 2.11
    R22T, K26R
    NV1G1818- NV1D3368- 123 Y1Q, W7Q, S11R, E12T, 20.5 2.8
    NH2 NH2 M19F, R22T, K26R
    NV1G1137 NV1D2974 129 Y1Q, W7Q, S11A, M19F, 21.6 1.34
    (synthetic) V20S, R22T
    NV1G1221 NV1D3017 140 Y1Q, W7Q, S11A, E12R, 21.9 2.48
    M19F, R22T
    NV1G1722 NV1D3533 273 Y1Q, W7Q, S11A, E12K, 22.4 3.5
    M19F, V20S, R22T, K26R
    NV1G1767 NV1D3345 177 Y1Q, Q3S, W7Q, S11R, M19F, 22.4 2.52
    R22T, K26R
    NV1G1769 NV1D3346 178 Y1Q, K4R, W7Q, S11R, M19F, 23.2 3.39
    R22T, K26R
    NV1G1780 NV1D3387 216 Y1Q, W7Q, S11R, E17D, 23.7 2.85
    M19F, R22T, K26R
    NV1G1886 NV1D3249 162 Y1Q, W7Q, S11A, M19F 24.1 11.5
    NV1G1812 NV1D3382 211 Y1Q, W7Q, S11R, K14S, M19, 24.3 2.14
    R22T, K26R
    NV1G1857 NV1D3366 196 Y1Q, W7Q, D105, S11R, 24.6 3.8
    M19F, R22T, K26R
    NV1G1821 NV1D3378 207 Y1Q, W7Q, S11R, K14A, 24.8 2.66
    M19F, R22T, K26R
    NV1G1993 NV1D3792 335 Y1Q, W7Q, S11R, M19F, 25.3 2.8
    R22T, K26R
    NV1G1007 NV1D2775 56 Y1Q, W7Q, S11A, M19F 25.4 2
    NV1G1787 NV1D3396 224 Y1Q, W7Q, S11R, G18Q, 26.4 3.17
    M19F, R22T, K26R
    NV1G1257 NV1D3016 139 Y1Q, W7Q, S11A, E12N, 26.6 3.1
    M19F, R22T
    NV1G1153 NV1D3034 116 Y1Q, W7Q, S11R, M19F, 27.3 2.02
    (synthetic) R22T, K26R
    NV1G1803 NV1D3403 230 Y1Q, W7Q, S11R, M19F, 28.3 1.97
    R22T, K26R, K27A
    (-GP) N—Ac— N—Ac— 115 Y1Q, W7Q, S11A, M19F, 28.6 2.23
    NV1G1137 NV1D2974 V20S, R22T
    NV1G1531 NV1D3019 142 Y1Q, W7Q, S11A, E12R, 28.7 4.78
    M19F, K26R
    NV1G1513 NV1D3007 134 Y1Q, W7Q, S11A, M19F, 29.6 9.17
    K26R
    NV1G1991 NV1D3789 333 Y1Q, W7Q, S11R, M19F, 29.9 5.19
    R22T, K26R
    NV1G1013 NV1D2733 40 Y1R, W7Q, M19F 7.54 2.9
    NV1G1740 NV1D3580 318 Y1R, W7Q, S11A, E12R, 8.4 1.5
    M19F, V20S
    NV1G1757 NV1D3538 278 Y1R, W7Q, S11R, E12N, 11.6 1.4
    M19F, R22T, K26R
    NV1G1741 NV1D3569 307 Y1R, W7Q, S11A, E12R, 11.9 0.8
    M19F, R22T
    NV1G1715 NV1D3584 322 Y1R, W7Q, S11A, E12N, 13.9 1.4
    M19F, V20S
    NV1G1754 NV1D3529 269 Y1R, W7Q, S11A, E12K, 14.6 1.7
    M19F, R22T, K26R
    NV1G1005 NV1D2772 59 Y1R, W7Q, S11A, M19F 15.6 1.8
    NV1G1733 NV1D3577 315 Y1R, W7Q, S11A, M19F, V20S 18.8 2.2
    NV1G1744 NV1D3534 274 Y1R, W7Q, S11A, E12K, 20.6 2.2
    M19F, V20S, R22T, K26R
    NV1G1724 NV1D3562 301 Y1R, W7Q, S11A, E12R, 23.6 2.7
    M19F, V20S, R22T
    NV1G1735 NV1D3566 305 Y1R, W7Q, S11A, M19F, R22T 23.7 2.5
    NV1G1760 NV1D3543 283 Y1R, W7Q, S11R, E12N, 23.8 1.9
    M19F, V20S, R22T, K26R
    NV1G1759 NV1D3547 287 Y1R, W7Q, S11R, M19F, 26.5 2.1
    R22T, K26R
    NV1G1751 NV1D3558 297 Y1R, W7Q, S11A, E12N, 26.7 3.4
    M19F, V20S, R22T
    NV1G1726 NV1D3551 291 Y1R, W7Q, S11R, M19F, 29.3 3.8
    V2OS, R22T, K26R
    NV1G1105 NV1D2729 39 Y1R, W7Q, S11A 8 8.85E-01
    NV1G957 NV1D2668 23 Y1R, W7Q 17.5 2.6
    (-GP)NV1G1001 (-GP)NV1D2773 109 Y1S, W7Q, S11A, M19F 9.47 1.28
    (-GP)NV1G1001 (-GP)NV1D2773 110 Y1S, W7Q, S11A, M19F 11.5 0.61
    —NH-methyl —NH-methyl
    NV1G1003 NV1D2734 44 Y1S, W7Q, M19F 13.4 0.8
    NV1G1864 NV1D3581 319 Y1S, W7Q, S11A, E12R, 14.6 1.7
    M19F, V20S
    NV1G1748 NV1D3530 270 Y1S, W7Q, S11A, E12K, 15.6 2.2
    M19F, R22T, K26R
    NV1G1758 NV1D3548 288 Y1S, W7Q, S11R, M19F, 17.6 1.9
    R22T, K26R
    NV1G1727 NV1D3544 284 Y1S, W7Q, S11R, E12N, 17.8 2.2
    M19F, V20S, R22T, K26R
    NV1G1719 NV1D3570 308 Y1S, W7Q, S11A, E12R, 18.1 1.5
    M19F, R22T
    NV1G1742 NV1D3535 275 Y1S, W7Q, S11A, E12K, 18.7 2.8
    M19F, V20S, R22T, K26R
    NV1G1001 NV1D2773 65 Y1S, W7Q, S11A, M19F 18.8 1.5
    NV1G1753 NV1D3585 323 Y1S, W7Q, S11A, E12N, 19.4 2.1
    M19F, V20S
    NV1G1762 NV1D3539 279 Y1S, W7Q, S11R, E12N, 19.4 1.8
    M19F, R22T, K26R
    NV1G1755 NV1D3574 312 Y1S, W7Q, S11A, E12N, 22.3 2.7
    M19F, R22T
    NV1G1717 NV1D3563 302 Y1S, W7Q, S11A, E12R, 22.4 2.4
    M19F, V20S, R22T
    NV1G1866 NV1D3559 298 Y1S, W7Q, S11A, E12N, 26.5 5.02
    M19F, V20S, R22T
    NV1G1721 NV1D3552 292 Y1S, W7Q, S11R, M19F, 28.1 3.7
    V20S, R22T, K26R
    NV1G975 NV1D2669 26 Y1S, W7Q 18.4 5.7
    NV1G983 NV1D2730 43 Y1S, W7Q, S11A 25.5 4.3
    NV1G1750- NV1D3586- 325 W7Q, S11A, E12N, M19F, 4.23 0.33
    NH2 NH2 V2OS
    NV1G1747 NV1D3531 271 W7Q, S11A, E12K, M19F, 13 2.1
    R22T, K26R
    NV1G1763 NV1D3540 280 W7Q, S11R, E12N, M19F, 16 1.5
    R22T, K26R
    NV1G1739 NV1D3582 320 W7Q, S11A, E12R, M19F, 17.8 2.2
    V20
    NV1G1750 NV1D3586 324 W7Q, S11A, E12N, M19F, 20.5 2.2
    V20S
    NV1G1718 NV1D3571 309 W7Q, S11A, E12R, M19F, 21 2.3
    R22
    NV1G1865 NV1D3560 299 W7Q, S11A, E12N, M19F, 27.2 3.42
    V20S, R22T
    NV1G1766 NV1D3549 289 W7Q, S11R, M19F, R22T, 27.5 3.2
    K26R
    NV1G961 NV1D2676 29 W7Q, S11A 26.5 2.9
    NV1G951 NV1D2674 18 Y1A, S11A 4.03 0.2
    NV1G1011 NV1D2740 37 Y1Q, S11A, M19F 3.62 9.9
    NV1G977 NV1D2665 22 Y1Q, M19F 4.9 0.4
    NV1G949 NV1D2675 21 Y1Q, S11A 4.33 0.3
    NV1G973 NV1D2662 25 Y1R, M19F 4.03 0.4
    NV1G965 NV1D2672 24 Y1R, S11A 4.5 0.3
    NV1G1009 NV1D2738 45 Y1S, S11A, M19F 2.57 0.2
    NV1G995 NV1D2663 28 Y1S, M19F 4.19 0.4
    NV1G1107- NV1D2890- 112 Y1S, M6F, S11A, M19L 9.12 1.17
    NH2 NH2
    NV1G971 NV1D2673 27 Y1S, S11A 4.31 0.5
    NV1G1782 NV1D3383 212 Y1Q, W7Q, S11R, E17R, 30.3 4.06
    M19F, R22T, K26R
    NV1G1990 NV1D3788 332 Y1Q, W7Q, S11R, M19F, 30.3 4.78
    R22T, K26R
    (-GP) N—Ac— (-GP) N—Ac— 120 Y1Q, W7Q, S11R, M19F, 30.4 2.96
    NV1G1153- NV1D3034 R22T, K26R
    NV1G1786 NV1D3389 218 Y1Q, W7Q, S11R, E17S, 30.8 4.48
    M19F, R22T, K26R
    NV1G1147 NV1D2969 124 Y1S, W7Q, S11A, M19F, 31 6.15
    V20S
    NV1G1764 NV1D3554 294 Y1A, W7Q, S11R, M19F, 31.4 3.3
    V20S, R22T, K26R
    NV1G963 NV1D2671 20 Y1Q, W7Q 31.5 6.4
    NV1G1835 NV1D3379 208 Y1Q, K4D, W7Q, S11R, 31.6 2.88
    M19F, R22T, K26R
    NV1G1231 NV1D3035 148 Y1Q, W7Q, S11A, E12N, 32 4.9
    M19F, R22T, K26R
    NV1G1743 NV1D3564 303 W7Q, S11A, E12R, M19F, 32.3 3.1
    V20S, R22T
    NV1G1960 NV1D3803 345 Y1Q, W7Q, S11R, M19F, 32.3 5.33
    R22T, K26R
    NV1G1924 NV1D3470 250 Y1Q, W7Q, S11R, M19L, 32.5 403
    R22T, K26R
    NV1G1756 NV1D3575 313 W7Q, S11A, E12N, M19F, 33.2 3.9
    R22T
    NV1G1109 NV1D2899 67 Y1S, W7Q, S11A, M19L 33.3 6.7
    NV1G1818 NV1D3368 122 Y1Q, W7Q, S11R, E12T, 33.5 10.7
    M19F, R22T, K26R
    NV1G1784 NV1D3386 215 Y1Q, W7Q, S11R, E17A, 33.6 4.71
    M19F, R22T, K26R
    NV1G1141 NV1D2972 127 Y1Q, W7Q, S11A, M19F, 34.1 6.2
    V20S
    NV1G1774 NV1D3347 179 Y1Q, K4T, W7Q, S11R, 34.2 5.99
    M19F, R22T, K26R
    NV1G1881 NV1D3257 167 Y1Q, W7Q, S11A, M19F 34.2 2.81
    NV1G1915 NV1D3467 249 Y1Q, W7Q, S11R, E17G, 34.5 4
    M19F, R22T, K26R
    NV1G1984 NV1D3806 348 Y1Q, W7Q, S11R, M19F, 35.1 4.56
    R22T, K26R
    NV1G1716 NV1D3561 300 Y1A, W7Q, S11A, E12N, 35.6 5
    M19F, V20S, R22T
    NV1G1255 NV1D3014 137 Y1Q, W7Q, S11R, M19F, 36.1 5.37
    R22T
    NV1G1959 NV1D3818 357 Y1Q, W7Q, S11R, M19F, 36.3 204
    R22T, K26R
    NV1G1825 NV1D3377 206 Y1Q, W7Q, S11R, K14T, 36.4 4.83
    M19F, R22T, K26R
    NV1G1723 NV1D3536 276 W7Q, S11A, E12K, M19F, 37 5.4
    V20S, R22T, K26R
    NV1G1732 NV1D3555 295 Y1R, W7Q, S11A, M19F, 37.4 4.3
    V20S, R22T
    NV1G1983 NV1D3809 350 Y1Q, W7Q, S11R, M19F, 38.9 4.81
    R22T, K26R
    NV1G1982 NV1D3805 347 Y1Q, W7Q, S11R, M19F, 41.2 5.44
    R22T, K26R
    NV1G1785 NV1D3385 214 Y1Q, W7Q, S11R, E17T, 41.5 6.5
    M19F, R22T, K26R
    NV1G1583 NV1D3030 144 Y1Q, W7Q, S11R, E12N, 41.9 5.15
    M19F, R22T
    NV1G1729 NV1D3545 285 W7Q, S11R, E12N, M19F, 42.8 4.6
    V20S, R22T, K26R
    NV1G1007 NV1D2775 56 Y1Q, W7Q, S11A, M19F 42.9 6.7
    NV1G1734 NV1D3568 306 Q1A, W7Q, S11A, M19F, 44 8.3
    R22T
    NV1G1683 NV1D3523 263 Y1Q, W7Q, S11R, M19F, 44.7
    R22T, K26R
    NV1G1834 NV1D3360 191 Y1Q, W7Q, D10R, S11R, 45.2 3.79
    M19F, R22T, K26R
    NV1G1795 NV1D3401 229 Y1Q, W7Q, S11R, M19F, 45.5 6.58
    R22T, K26R, K27R
    NV1G1689 NV1D3514 255 Y1Q, W7Q, S11R, M19F, 46.4
    R22T, K26R
    NV1G2043 NV1D3835 370 Y1Q, W7Q, S11R, M19F, 46.4 4.09
    R22T, K26R
    NV1G1783 NV1D3384 213 Y1Q, W7Q, S11R, E17K, 46.8 7.39
    M19F, R22T, K26R
    NV1G1239 NV1D3020 143 Y1Q, W7Q, S11A, M19F, 47.2 7.84
    R22T, K26R
    NV1G1788 NV1D3399 227 Y1Q, W7Q, S11R, M19F, 47.3 6.36
    V20T, R22T, K26R
    NV1G899 NV1D2774 52 Y1A, W7Q, S11A, M19F 50.5 15.2
    NV1G2057 NV1D3799 341 Y1Q, W7Q, S11R, M19F, 50.6 6.33
    R22T, K26R
    NV1G1738 NV1D3578 316 W7Q, S11A, M19F, V20S, 50.7 5.7
    NV1G1713 NV1D3525 265 Y1Q, W7Q, S11R, M19F, 52.3
    R22T, K26R
    NV1G1765 NV1D3553 293 W7Q, S11R, M19F, V20S, 52.4 10
    R22T, K26R
    NV1G1916 NV1D3465 247 Y1Q, W5F, W7Q, S11R, 52.8 10.3
    M19F, R22T, K26R
    NV1G1977 NV1D3804 346 Y1Q, W7Q, S11R, M19F, 53.6 6.27
    R22T, K26R
    NV1G1879 NV1D3259 168 Y1Q, W7Q, S11A, M19F 54.9 7.62
    NV1G1884 NV1D3256 166 Y1Q, W7Q, S11A, M19F 55.7 10.5
    NV1G1986 NV1D3819 358 Y1Q, W7Q, S11R, M19F, 56 6.57
    R22T, K26R
    NV1G1633 NV1D3251 163 Y1Q, W7Q, S11A, M19F 56.1 13.9
    NV1G1880 NV1D3261 170 Y1Q, W7Q, S11A, M19F 57 6.25
    NV1G1985 NV1D3808 349 Y1Q, W7Q, S11R, M19F, 57 6.74
    R22T, K26R
    NV1G1849 NV1D3400 228 Y1Q, W7Q, S11R, M19F, 57.3 9.52
    V20Q, R22T, K26R
    NV1G1883 NV1D3260 169 Y1Q, W7Q, S11A, M19F 57.6 6.91
    NV1G1145 NV1D2970 125 Y1S, W7Q, S11A, M19F, 58 18.8
    R22T
    NV1G1697 NV1D3517 258 Y1Q, W7Q, S11R, M19F, 58.5
    R22T, K26R
    NV1G1737 NV1D3579 317 Y1A, W7Q, S11A, M19F, 59.9 9.6
    V20S
    NV1G1978 NV1D3833 368 Y1Q, W7Q, S11R, M19F, 60.3 9.57
    R22T, K26R
    NV1G1954 NV1D3800 342 Y1Q, W7Q, S11R, M19F, 60.9 6.43
    R22T, K26R
    NV1G1989 NV1D3791 334 Y1Q, W7Q, S11R, M19F, 61.8 8.66
    R22T, K26R
    NV1G1815 NV1D3380 209 Y1Q, K4E, W7Q, S11R, 64 10.5
    M19F, R22T, K26R
    NV1G1967 NV1D3793 336 Y1Q, W7Q, S11R, M19F, 64.6 8.19
    R22T, K26R
    NV1G1869 NV1D3573 311 Y1R, W7Q, S11A, E12N, 64.7 50.7
    M19F, R22T
    NV1G1872 NV1D3777 330 Y1Q, W7Q, S11R, M19F, 64.9 15.3
    R22T, K26R
    NV1G1979 NV1D3834 369 Y1Q, W7Q, S11R, M19F, 65.5 7.59
    R22T, K26R
    NV1G1827 NV1D3365 195 Y1Q, W7Q, D10Q, S11R, 66.1 10.1
    M19F, R22T, K26R
    NV1G1768 NV1D3341 174 Y1Q, Q3T, W7Q, S11R, 66.2 9.32
    M19F, R22T, K26R
    NV1G911 NV1D2666 30 W7Q, M19F 66.5 36.7
    NV1G1856 NV1D3397 225 Y1Q, W7Q, S11R, G18S, 66.7 7.31
    M19F, R22T, K26R
    NV1G1973 NV1D3810 351 Y1Q, W7Q, S11R, M19F, 66.9 7.04
    R22T, K26R
    NV1G1855 NV1D3398 226 Y1Q, W7Q, S11R, M19F, 67.3 11
    V20S, R22T, K26R
    NV1G1961 NV1D3802 344 Y1Q, W7Q, S11R, M19F, 68 8.23
    R22T, K26R
    NV1G1846 NV1D3431 244 Y1Q, K4E, W7Q, S11R, 68.6 13.9
    E17K, M19F, R22T, K26R
    NV1G1771 NV1D3348 180 Y1Q, K4A, W7Q, S11R, 70.6 15.9
    M19F, R22T, K26R
    NV1G1691 NV1D3520 261 Y1Q, W7Q, S11R, M19F, 71.4
    R22T, K26R
    NV1G1681 NV1D3511 252 Y1Q, W7Q, S11R, M19F, 71.5
    R22T, K26R
    NV1G1968 NV1D3822 359 Y1Q, W7Q, S11R, M19F, 74.2 11.1
    R22T, K26R
    NV1G1813 NV1D3424 238 Y1Q, W7Q, D10K, S11R, 75.2 12.2
    E12K, M19F, R22T, K26R
    NV1G1067 NV1D2893 57 Y1Q, W7Q, S11A, M19L 75.5 10.5
    NV1G1867 NV1D3546 286 Y1A, W7Q, S11R, E12N, 76 17.6
    M19F, V20S, R22T, K26R
    NV1G1143 NV1D2971 126 Y1S, W7Q, S11A, M19F, 77.5 22.1
    V20S, R22T
    NV1G1806 NV1D3409 232 Y1Q, W7Q, S11R, M19F, 79.1 11.3
    R22T, K26R, K28T
    NV1G1061 NV1D2896 60 Y1R, W7Q, S11A, M19L 80.3 7.13
    NV1G1793 NV1D3419 236 Y1Q, W7Q, S11R, M19F, 80.9 11.9
    R22T, K26R, W30D
    NV1G1613 NV1D3057 157 Y1Q, W7Q, S11R, E12K, 83.4 16.6
    M19F, K26R
    NV1G1585 NV1D3052 155 Y1Q, W7Q, S11A, E12K, 84.8 28.8
    M19F, K26R
    NV1G1707 NV1D3524 264 Y1Q, W7Q, S11R, M19F, 84.9
    R22T, K26R
    NV1G1773 NV1D3350 182 Y1Q, K4E, W7Q, S11R, 85.6 14.4
    M19F, R22T, K26R
    NV1G1949 NV1D3828 364 Y1Q, W7Q, S11R, M19F, 87.5 11
    R22T, K26R
    NV1G1976 NV1D3811 352 Y1Q, W7Q, S11R, M19F, 87.7 15.7
    R22T, K26R
    NV1G1956 NV1D3801 343 Y1Q, W7Q, S11R, M19F, 88.1 11.4
    R22T, K26R
    NV1G1975 NV1D3832 367 Y1Q, W7Q, S11R, M19F, 88.4 12.3
    R22T, K26R
    NV1G1839 NV1D3774 328 Y1Q, W7Q, S11R, M19F, 88.6 19.6
    R22T, K26R
    NV1G1971 NV1D3830 366 Y1Q, W7Q, S11R, M19F, 88.6 9.88
    R22T, K26R
    NV1G1882 NV1D3262 171 Y1Q, W7Q, S11A, M19F 89.2 8.32
    NV1G1950 NV1D3797 339 Y1Q, W7Q, S11R, M19F, 91.1 13.5
    R22T, K26R
    NV1G1828 NV1D3363 194 Y1Q, W7Q, D10A, S11R, 93.1 15.3
    M19F, R22T, K26R
    NV1G1139 NV1D2973 128 Y1Q, W7Q, S11A, M19F, 93.9 19.5
    R22T
    NV1G1842 NV1D3430 243 Y1Q, K4D, W7Q, S11R, 93.9 14.1
    E17K, M19F, R22T, K26R
    NV1G1948 NV1D3798 340 Y1Q, W7Q, S11R, M19F, 94.5 17.8
    R22T, K26R
    NV1G1807 NV1D3408 231 Y1Q, W7Q, S11R, M19F, 94.8 17.8
    R22T, K26R, K28R
    NV1G1137 NV1D2974 129 Y1Q, W7Q, S11A, M19F, 95.7 16.2
    V20S, R22T
    NV1G1843 NV1D3432 245 Y1Q, K4E, W7Q, S11R, 95.9 10.4
    E17R, M19F, R22T, K26R
    NV1G1822 NV1D3423 237 Y1Q, W7Q, D10R, S11R, 99.5 9.45
    E12R, M19F, R22T, K26R
    NV1G1862 NV1D3556 296 W7Q, S11A, M19F, V20S, 100 18.5
    R22T
    NV1G1969 NV1D3795 337 Y1Q, W7Q, S11R, M19F, 100 14.5
    R22T, K26R
    NV1G1980 NV1D3812 353 Y1Q, W7Q, S11R, M19F, 101 23.6
    R22T, K26R
    NV1G1850 NV1D3414 235 Y1Q, W7Q, S11R, M19F, 102 19.4
    R22T, K26R, K28S
    NV1G1981 NV1D3815 356 Y1Q, W7Q, S11R, M19F, 102 13.5
    R22T, K26R
    NV1G1851 NV1D3390 219 Y1Q, W7Q, S11R, G18R, 108 15.5
    M19F, R22T, K26R
    NV1G1922 NV1D3466 248 Y1Q, W7Q, S11E, M19F, 108 922
    R22T, K26R
    NV1G1778 NV1D3349 181 Y1Q, K4D, W7Q, S11R, 109 16
    M19F, R22T, K26R
    NV1G1972 NV1D3824 361 Y1Q, W7Q, S11R, M19F, 110 16.1
    R22T, K26R
    NV1G1974 NV1D3796 338 Y1Q, W7Q, S11R, M19F, 110 19.6
    R22T, K26R
    NV1G1826 NV1D3357 188 Y1Q, W7Q, T8E, S11R, 111 15.1
    M19F, R22T, K26R
    NV1G1892 NV1D3439 246 Y1Q, W7Q, S11R, M19F, 112 13.2
    R22T, K26R, W30G
    NV1G1819 NV1D3375 204 Y1Q, W7Q, S11R, R13S, 113 1270
    M19F, R22T, K26R
    NV1G1805 NV1D3410 233 Y1Q, W7Q, S11R, M19F, 114 21.5
    R22T, K26R, K28A
    NV1G1831 NV1D3374 203 Y1Q, W7Q, S11R, R13Q, 114 1600
    M19F, R22T, K26R
    NV1G1693 NV1D3512 253 Y1Q, W7Q, S11R, M19F, 115.6
    R22T, K26R
    NV1G1854 NV1D3392 221 Y1Q, W7Q, S11R, G18T, 117 21.8
    M19F, R22T, K26R
    NV1G1951 NV1D3829 365 Y1Q, W7Q, S11R, M19F, 122 13.3
    R22T, K26R
    NV1G1860 NV1D3393 222 Y1Q, W7Q, S11R, G18A, 125 24.8
    M19F, R22T, K26R
    NV1G1099 NV1D2732 36 Y1Q, W7Q, S11A 126 26.9
    NV1G1705 NV1D3513 254 Y1Q, W7Q, S11R, M19F, 131.2
    R22T, K26R
    NV1G1848 NV1D3426 240 Y1Q, W7Q, D10K, S11R, 135 39.9
    E12K
    NV1G1952 NV1D3813 354 Y1Q, W7Q, S11R, M19F, 139 30.1
    R22T, K26R
    NV1G1631 NV1D3252 164 Y1Q, W7Q, S11A, M19F 145 53
    NV1G1817 NV1D3371 201 Y1Q, W7Q, S11R, R13A, 151 33.7
    M19F, R22T, K26R
    NV1G1789 NV1D3394 223 Y1Q, W7Q, S11R, G18D, 155 41.4
    M19F, R22T, K26R
    NV1G1852 NV1D3391 220 Y1Q, W7Q, S11R, G18K, 157 23.1
    M19F, R22T, K26R
    NV1G1709 NV1D3510 251 Y1Q, W7Q, S11R, M19F, 159
    R22T, K26R
    NV1G1840 NV1D3425 239 Y1Q, W7Q, D10R, S11R, 161 27.9
    E12R
    NV1G1809 NV1D3413 234 Y1Q, W7Q, S11R, M19F, 164 43.7
    R22T, K26R, K28Q
    NV1G1863 NV1D3356 187 Y1Q, W7Q, T8D, S11R, 167 32.2
    M19F, R22T, K26R
    NV1G1699 NV1D3527 267 Y1Q, W7Q, S11R, M19F, 169.1
    R22T, K26R
    NV1G1844 NV1D3428 242 Y1Q, W7Q, D10K, S11R, E12 180 52.4
    NV1G1853 NV1D3370 200 Y1Q, W7Q, S11R, R13T, 181 25.1
    M19F, R22T, K26R
    NV1G1946 NV1D3825 362 Y1Q, W7Q, S11R, M19F, 194 28.4
    R22T, K26R
  • The wild-type Protoxin-II inhibits Nav1.7 with an IC50 value of about 4 nM in FLIPR assay as described in Example 3. Variants retaining significant Nav1.7 potency were characterized further. FIG. 1 shows the sequence genus of generated Protoxin-II variants that inhibit Nav1.7 with an IC50 value of 30 nM or less.
  • Select Protoxin-II variants were tested for their inhibition of Nav1.7 and for their selectivity against human Nav1.6 using QPatch. IC50 values for both Nav1.7 and Nav1.6 for select peptides obtained using QPatch are shown in FIG. 2. These peptides inhibited Nav1.7 with an ICs of 30 nM or less, and were at least 30-fold selective over Nav1.7 when compared to Nav1.6.
  • The amino acid sequences of the peptides shown in FIG. 2 are shown in FIG. 3. All these peptides had W7Q and M19F substitutions when compared to the wild type Protoxin-II.
  • The Protoxin-II variants were expressed and purified as described in Example 1, or synthesized by standard solid phase synthesis methods. The yields of the recombinant or synthetic peptides were compared to the yields of the wild-type protoxin. Table 12 shows that the yields of the select Protoxin-II variants were significantly higher than that of Protoxin-II, indicating improved folding properties of the variants. The scale of the solid-phase synthesis was 0.5 mmol.
  • TABLE 12
    Solid phase synthesis Recombinant
    Total Yield Yield expression
    Peptide yield from Crude From Linear % active isomer
    Protoxin-II 52 mg 2.7% 7.3% 54.0%
    NV1D2775 84 mg 4.5% 18.7% 89.1%
    NV1D3034 149 mg  8.0% 21.0% 85.2%
    NV1D3368 83 mg 4.0% 24.0% 93.8%
  • Example 5: Protoxin-II Variants are Efficient in In Vivo Models of Pain Materials and Methods Animals
  • Male C57Bl/6 mice (24-26 g), ordered from Charles River and housed individually, were used for this study.
  • Behavioral Tests
  • Von Frey Test: Mechanical (tactile) threshold was assessed by Von Frey Hairs following the Up-Down method (Dixon, 1980, Chaplan et al., 1994). 7 graded stimuli (von Frey filaments: 0.03, 0.07, 0.16, 0.4, 0.6, 1, 2 g; Stoelting, Wood Dale, Ill.) were used. Von Frey hairs were presented perpendicularly against the center plantar area (between toris) on a hindpaw. Sufficient force was applied to bend the filament slightly and held for 3 seconds. Per the Chaplan paper, a positive response can be either 1) a sharp withdrawal or 2) immediate flinching upon removal of the filament. See Chaplan et al. for more details. Mice were acclimated to the wire mesh in the testing chamber for 30-60 minutes prior to testing.
  • Hargreaves Test: A modified Hargreaves box was used to measure thermal paw withdrawal latency (PWL) (Hargreaves et al., 1988, Pain, 32:77-88; Dirig et al., 1997, J Neurosci. Methods, 76:183-191). This box consists of a chamber with a raised glass floor maintained at a constant temperature (27° C.). The thermal nociceptive stimulus originates from a projection bulb light beam below the glass surface. The light beam is aimed at the area between toris (center plantar). The “start” button will turn on the light and start the timer. Movements (such as a sudden withdrawal) of the stimulated paw will trigger the switch to turn off the light and stop the timer. The latency in seconds is displayed. If no movement occurs, the bulb will be turned off after 20 seconds (cutoff) to prevent tissue injury. The animals were allowed to habituate on the glass surface for 30-60 minutes before PWL measurement. Constant amperage was used throughout the study, which resulted in Pre-test paw withdrawal latencies between 8-12 seconds when averaged over 3 to 6 read-outs taken at least 5 minutes apart.
  • MPE % Calculation: Percent maximum possible effect (MPE %)=(T1−To)/(Tc−Td×100%. To: threshold on day( ) (post-CFA, pre-pump); To: threshold on day 1 post pump implantation; Tc: cut-off of the test (20 s for the Hargreaves test and 2 g for the Von Frey test).
  • Hotplate Test: Animals were placed on a 10″×10″ metal plate surrounded by 4 Plexiglas walls (15 inches high). The plate was maintained at a temperature of either 50 or 55° C. The response latency (time when the animal first flinches or licks its hind paw, jumps, or vocalizes) was measured and the animal removed from the plate. Animals showing no response were removed from the plate after 40 s (50° C.) or 20 s (55° C.) to prevent any possible tissue damage. This trial was repeated 2-5 times every 15-60 minutes in a day.
  • Inflammatory Pain Models
  • CFA Model: Animals were anesthetized with isoflurane (4% induction and 2% maintenance) and 20 μL of 100% Complete Freund's Adjuvant (CFA; Sigma-Aldrich; Saint Louis, Mo.) was injected into the center plantar area on one hind paw using a 27gauge needle attached to a 50-μL Hamilton syringe. Carrageenan model: Animals were anesthetized with isoflurane (4% induction and 2% maintenance) and 25 μL of 2% A-carrageenan (Sigma-Aldrich; Saint Louis, Mo.) dissolved in normal saline was injected into the center plantar area on hind paws using an insulin syringe (BD; Franklin Lakes, N.J.).
  • Implantation of Mini-Pumps
  • Alzet micro-osmotic mini pumps (Durect Corporation Model 1003D and 2001 D) were filled and primed per manufacturer's guide. Mice were anesthetized with isoflurane (5% induction; 2% maintenance). Their backs were shaved, wiped down with isopropyl alcohol and povidone iodine, and a small incision was made between the scapulae. Using a pair of forceps or hemostat, a small pocket was formed by spreading the subcutaneous connective tissues apart. The pump was inserted into the pocket with the flow moderator pointing away from the incision. The skin incision was then closed using 7-mm staples and the animals were allowed to recover in their home cages.
  • Data Analysis
  • Data are represented as mean±s.e.m. Prism (Graphpad Software Inc., La Jolla, Calif.) was used for graphing and statistical analysis. For comparison of threshold values over time, a two-way ANOVA followed by Bonferroni's multiple comparison test was used with a significance level of p<0.05. Hotplate and MPE % data were analyzed by one-way ANOVA followed by Bonferroni's multiple comparison test.
  • Results
  • Efficacy of variants NV1D3034-OH (NV1D3034-COOH), NV1 D3368-OH (NV1 D3368-COOH) and NV1 D2775-OH (NV1 D2775-COOH) was studied in the CFA model, a commonly used model of inflammatory pain. The injection of CFA in the hindpaw induced paw edema (not shown) and hypersensitivity to thermal stimuli (thermal hyperalgesia), as indicated by the lowered thermal latency in the injected paw on day( ) (FIG. 6A). Thermal hyperalgesia was completely reversed by NV1D3034-OH at 684 and 1824 μg/day, when administered by a subcutaneous osmotic mini-pump (FIGS. 4A and 4B).
  • NV1 D3368-OH fully reversed CFA-induced thermal hyperalgesia at 684 and 1824 μg/day (FIGS. 5A and 5B). NV1 D2775-OH demonstrated strong efficacy in the CFA model. Thermal latencies reached values close to the cut-off following NV1 D2775 administration (FIGS. 6A and 6B, 1824 μg/day), suggesting a strong analgesia effect on top of the anti-hyperalgesia effect. In addition, NV1 D2775-OH reversed CFA-induced tactile allodynia (FIGS. 6C and 6D, 1824 μg/day). The anti-hyperalgesic effect of NV1 D2775-OH was seen as early as 4 hr post-pump implantation (FIG. 7A). The effect reached the maximum at 8 hr in both the thermal and tactile tests (FIGS. 7A and 7B), which was maintained at 24 hr. Thermal latency and tactile threshold returned the control level by 48 h post pump implantation (approximately 24 h after the pumps were predicted to be empty) (FIGS. 7A and 7B).
  • CFA-induced thermal hyperalgesia was readily reversed by two additional peptides, NV1 D3368-amide (NV1 D3368-NH2) and NV1D3034-N-methylamide (NV1D3034-NHMe). Thermal MPE % from the experiments is summarized in Table 13.
  • TABLE 13
    Dose (pg/day/mouse)
    Vehicle
    Peptide (PBS) 228 684 1824
    NV1D3034-OH 20 ± 7 (11) 22 ± 6 (6) 48 ± 10* (8) 50 ± 6* (8)
    NV1D3368-OH 13 ± 7 (8) 23 ± 8 (7) 42 ± 9* (7) 47 ± 6** (8)
    NV1D2775-OH 15 ± 4 (20) 35 ± 8 (8) 57 ± 12*** (8) 85 ± 6**** (12)
    NV1D3368-NH 2 15 ± 13 (6) 27 ± 4 (4) 46 ± 9 (4) 55 ± 15 (6)
    NV1D3034-NHMe 5 ± 25 (3) 49 ± 17 (6)
    *P < 0.05,
    **P < 0.01,
    ***P < 0 001 and
    ****P < 0.0001 vs. PBS, one-way ANOVA followed by Bonferroni's multiple comparison.
  • NV1 D2775-OH also exhibited strong, dose-dependent efficacy in the hotplate test (FIG. 8). Latencies at 50 and 55° C. reached values near cut-off following the administration of 1824 μg/day. At 228 μg/day, NV1 D2775-OH produced a modest yet significant increase in the thermal latency, compared to the PBS control.
  • The efficacy of NV1 D2775-OH was evaluated in another model of inflammatory pain, the carrageenan model. Animals were implanted with NV1 D2775-OH or PBS pumps. Thermal withdrawal latencies were measured pre- and on day 1 post-pump. A-carrageenan was injected into the hindpaws and thermal latencies were measured again on 2, 3 and 4 hr following carrageenan. NV1D2775-OH at 1824 μg/day produced significant analgesia (FIG. 9). Injection of A-carrageenan in the hindpaws induced inflammation (not shown) and lowered thermal paw withdrawal latency in the Hargreaves test over the 4 hr test-period (FIG. 9, PBS group). Animals pretreated with NV1 D2775-OH at 1824 μg/day were fully protected from carrageenan-induced hyperalgesia.
  • Example 6: Generation and Characterization of Combinatorial Protoxin-II Variants
  • An amino acid scanning library was generated for Protoxin-II. At every non-cysteine position in Protoxin-II (Tyr1, Gln3, Lys4, Trp5, Met6, Trp7, Thr8, Asp10, Ser11, Glu12, Arg13, Lys14, Glu17, Gly18, Met19, Val20, Arg22, Leu23, Trp24, Lys26, Lys27, Lys28, Leu29 and Trp30) the following residues were substituted in place of the native residue: Ala, Asp, Glu, Phe, Gly, His, Ile, Lys, Leu, Asn, Pro, Gln, Arg, Ser, Thr, Val, and Tyr.
  • Mutant peptides were expressed as recombinant fusions to human serum albumin and site-specifically enzymatically cleaved using HRV3C to generate Protoxin-II variants as described in Example 1. Each Protoxin-II variant, after cleavage from HSA had a residual N-terminal GP from the cleavage site. For each Protoxin-II variant, IC50 values against human Nav1.7 were measured using FLIPR Tetra or Qpatch according to the protocols described in Example 3. Variants demonstrating IC50<100 nM for human Nav1.7 were counter-screened for selectivity against additional hNav channels using Qpatch electrophysiology. Selective hits were identified and used in the design of combinatorial peptide libraries which were produced using both recombinant expression and solid-phase peptide synthesis. Combinatorial variants were screened using the same strategy as detailed above.
  • Based on the results, positions that can be mutated to improve selectivity include Gln3, Ser11, Glu12, Lys14, Glu17, Gly18, Leu29 and Trp30 (residues numbering according to SEQ ID NO: 1).
  • The solution structure of Protoxin-II was determined by NMR and is shown in FIG. 10 as a surface representation. The left hand side of the Figure shows the previously described (Park et al., J. Med. Chem. 2014, 57:6623-6631) ring of Trp residues, W5/W7/W24, surrounding M6. On the opposite side of the molecule, using both mutagenesis and the NMR structure, a selectivity face was identified in this study on Protoxin-II consisting of multiple amino acid positions which can be mutated to improve selectivity for hNav1.7 over other sodium channel isoforms. The residues residing on the selectivity face include residues Ser11, Glu12, Lys14, Glu17, Gly18, Leu29 and Trp30 (residue numbering according to SEQ ID NO: 1). The identification of the selectivity face and multiple positions within responsible for selectivity towards Nav1.7 has not been described earlier.
  • Improved selectivity of Protoxin II variants with substitution at Ser11 is unexpected as it has been earlier demonstrated that mutation of Ser11 affect activity on multiple Nav channels, and therefore the residue was concluded not to play a role in Protoxin-II Nav1.7 selectivity (Park et al., J. Med. Chem. 2014, 57:6623-6631).
  • A key step in the synthetic production of Protoxin-II variants is the oxidative refolding of the linear peptide, where the disulfide pairings are formed. The RP-HPLC trace for native Protoxin-II purification following refolding revealed multiple peaks at differing retention times that were of correct mass but demonstrated differing levels of activity, indicative of improper folding of the peptide.
  • The relative abundance of the RP-HPLC major peak, and therefore the relative abundance of correctly folded peptide could be improved by making substitutions at various Protoxin-II positions. Mutation of Trp7 or Trp30 improved folding of the resulting Protoxin-II variant. Mutation of both Trp7 and Trp30 in combination further improved folding of the resulting Protoxin-II variant, and could rescue folding of difficult-to-refold Protoxin-II variants.
  • Production of combinatorial mutant peptides having one or more substitutions that improved selectivity (Gln3, Ser11, Glu12, Lys14, Glu17, Gly18, and Leu29) as well as mutations at Trp7 and Trp30 resulted in peptides with both improved selectivity and improved refolding properties. Protoxin-II belongs to a family 3 of inhibitory cysteine knot peptides (Klint et al., Toxicon 60:478-491, 2012). Trp7 is conserved in all family 3 members, and substitutions at this position as well as at Trp5 and Met6 in Jingzhaotoxin-V, another family 3 inhibitory cysteine knot peptide, resulted in loss in potency, indicating that hydrophobic residues at positions 5, 6 and 7 in Jingzhaotoxin-V are essential to Jingzhaotoxin-V Nav1.7 inhibitory potency (Int. Pat. Publ. No. 2014/165277). Trp5/Met6/Trp7 is also conserved in Protoxin-II, and therefore it was unexpected that polar substitutions at Trp7 can be made without loss of Protoxin-II activity with significantly improved refolding properties. Substitutions at Trp30 were shown to simultaneously improve Nav1.7 selectivity and refolding properties of the variant peptide and were unexpected since individual advantageous substitutions typically only improve a single parameter.
  • Table 14 shows the amino acid sequences of the select generated Protoxin-II variants.
  • TABLE 14
    Protein
    Protein Substitution SEQ ID Amino acid sequence
    NV1G2232 W30L 408 GPYCQKWMWTCDSERKCCEGM
    VCRLWCKKKLL-COOH
    NV1G2182 W30F 409 GPYCQKWMWTCDSERKCCEGM
    VCRLWCKKKLF-COOH
    NV1G2319 W30Y 410 GPYCQKWMWTCDSERKCCEGM
    VCRLWCKKKLY-COOH
    NV1G2329 W30G 411 GPYCQKWMWTCDSERKCCEGM
    VCRLWCKKKLG-COOH
    NV1G2129 W30I 412 GPYCQKWMWTCDSERKCCEGM
    VCRLWCKKKLI-COOH
    NV1G2291 W30V 413 GPYCQKWMWTCDSERKCCEGM
    VCRLWCKKKLV-COOH
    NV1G2156 W7Y 414 GPYCQKWMYTCDSERKCCEGM
    VCRLWCKKKLW-COOH
    NV1G2082 W7E 415 GPYCQKWMETCDSERKCCEGM
    VCRLWCKKKLW-COOH
    63930841 W7Q 416 GPYCQKWMQTCDSERKCCEGM
    VCRLWCKKKLW-COOH
    64087946 (-GP) W7Q, S11A, 417 YCQKWMQTCDAERKCCEGFSC-
    M19F, V20S, (N-Me-Arg)-LWCKKKLL-COOH
    R22Me, W30L
    64053366 (-GP) 418 YCQKWMQTCDDERKCCEGMVC
    W7Q, S11D, W30L RLWCKKKLL-COOH
    64053340 (-GP) W7Q, K14F, 419 YCQKWMQTCDSERFCCEGMVC
    W30L RLWCKKKLL-COOH
    64053236 W7Q, K14F, W30L 420 GPYCQKWMQTCDSERFCCEGM
    VCRLWCKKKLL-COOH
    64053223 W7Q, S11I, W30L 421 GPYCQKWMQTCDIERKCCEGMV
    CRLWCKKKLL-COOH
    63955918 W7Q W30L 422 GPYCQKWMQTCDSERKCCEGM
    VCRLWCKKKLL-COOH
    64053210 W7Q, E17N, W30L 423 GPYCQKWMQTCDSERKCCNGM
    VCRLWCKKKLL-COOH
    64087907 (-GP) W7Q 424 YCQKWMQTCDSERKCCEGMVC
    RLWCKKKLW-COOH
    64032488 (-GP) W7Q, W30L 425 YCQKWMQTCDSERKCCEGMVC
    RLWCKKKLL-COOH
    64053301 W7Q S11V, W30L 426 GPYCQKWMQTCDVERKCCEGM
    VCRLWCKKKLL-COOH
    64053275 W7Q, E17L, W30L 427 GPYCQKWMQTCDSERKCCLGM
    VCRLWCKKKLL-COOH
    64053327 (-GP) 428 YCQKWMQTCDSERKCCNGMVC
    W7Q, E17N, W30L RLWCKKKLL-COOH
    NV1G2324 E17Y 429 GPYCQKWMWTCDSERKCCYGM
    VCRLWCKKKLW-COOH
    NV1G2094 E17I 430 GPYCQKWMWTCDSERKCCIGM
    VCRLWCKKKLW-COOH
    NVG1996 E17L 431 GPYCQKWMWTCDSERKCCLGM
    VCRLWCKKKLW-COOH
  • Select variants were characterized for their inhibition of Nav1.7 using FLIPR Tetra or Qpatch as described in Example 3. Table 15 shows the IC50 values obtained. For some variants, % inhibition at certain concentration was recorded for Qpatch results (% of Protoxin-11).
  • TABLE 15
    hNav1.7
    Protein TETRA QP
    Protein SEQ ID IC50 IC50)
    Name NO: (nM) se* (nM % blk** se*
    NV1G223 408 16.7 1.32 5.0 56.5% @ 10 nM 5.7
    NV1G218 409 17.3 1.37 3.8 54.2% @ 10 nM 5.4
    Figure US20210363204A1-20211125-P00899
    NV1G231 410 20.7 2.3 9.7 43.2% @ 10 nM 6.2
    Figure US20210363204A1-20211125-P00899
    NV1G232 411 38 2.43E+00
    Figure US20210363204A1-20211125-P00899
    NV1G212 412 47.3 3.81 −6.5% @ 10 nM 6.5
    Figure US20210363204A1-20211125-P00899
    NV1G229 413 63.3 14.9
    Figure US20210363204A1-20211125-P00899
    NV1G215 414 90.5 6.88
    Figure US20210363204A1-20211125-P00899
    NV1G208 415 90.8 11.4
    Figure US20210363204A1-20211125-P00899
    63930841 416 20.9
    64087946 417 23.8 20.7% @ 10 nM 10.9
    64053366 418 22.1% @ 10 nM 3.5
    64053340 419 26.8% @ 10 nM 3.7
    64053236 420 28.0% @ 10 nM 13.2
    64053223 421 33.0% @ 10 nM 5.8
    63955918 422 10.8 38.50% @ 10 nM 4.5
    64053210 423 41.7% @ 10 nM 6.2
    64087907 424 7.1 45.1% @ 10 nM 6.0
    64032488 425 6.5 45.6% @ 10 nM 4.6
    64053301 426 10.7 45.83% @ 10 nM 3.3
    64053275 427 2.9 48.22% @ 10 nM 5.2
    64053327 428 7.9 51.9% @ 10 nM 2.6
    NV1G232 429 57.5% @ 10 nM 3.9
    Figure US20210363204A1-20211125-P00899
    NV1G209 430 63.2%@ 30 nM 6.2
    Figure US20210363204A1-20211125-P00899
    NV1G199 431 0.5 76.9% @ 10 nM 2.3
    Figure US20210363204A1-20211125-P00899
    *se; standard error
    **% blk:
    QP: QPatch
    Figure US20210363204A1-20211125-P00899
    indicates data missing or illegible when filed
  • Selectivity of select variants were tested against various human Navl.x channels. Table 16 shows the results of those experiments. IC50 values for each channel were measured using QPatch.
  • TABLE 16
    Protein
    Protein SEQ ID IC50 (nM)
    Name Substitution NO: Nav1.1 Nav1.2 Nav1.4 Nav1.6
    NV1G2232 W30L 408 3847.0 562.7
    NV1G2182 W30F 409 239.6 732.2 253.1
    NV1G2319 W30Y 410 1704.0
    63930841 W7Q 416 543.1
    64087946 (-GP) 417 2586.0
    W7Q, S11A, M19F,
    V20S, R22Me, W30L
    63955918 W7Q W30L 422 1951.0 17000.0 1987.0
    64087907 (-GP) W7Q 424 1460.0
    64032488 (-GP) W7Q 425 1336.0 1842.0
    W30L
    64053301 W7Q SUV 426 15340.0 19350.0 2244.0
    W30L
    64053275 W7Q E17L 427 3868.0 136.7 2219.0
    W30L
    64053327 (-GP) W7Q 428 6391.0 6656.0 3867.0
    E17N W30L
  • Protoxin-II variants were expressed and purified as described in Example 1, or synthesized by standard solid phase synthesis methods. The yields of the recombinant or synthetic peptides were compared to the yields of the wild-type protoxin. Table 17 shows that the yields of the select Protoxin-II variants were significantly higher than that of Protoxin-II, indicating improved folding properties of the variants. The scale of the solid-phase synthesis was 0.1 mmol.
  • TABLE 17
    total
    Protein name Substitution yield(mg)
    NV1D12(Protoxin-II with 3.8
    N-terminal GP)
    63930841 W7Q 14.4
    NV1G2232 W30L 14.5
    63955918 W7Q, W30L 16.2
    NV1G1996 E17L 1.8
    64053275 E17L, W7Q, W30L 13.0
  • Example 7. Protoxin-II Variants are Efficient In Vivo Models of Pain Following Intrathecal Administration
  • Efficacy of select Protoxin-II variants in reducing pain after intrathecal administration was evaluated.
  • Peptides NV1D2775-OH, NV1D3034 and 63955918 were used in the studies. Animal models that measure acute thermal pain (tail flick and hot plate) and injury-induced pain (formalin flinching) were used.
  • Tail-flick test: The animals were placed on a tail-flick device (Ugo Basile). The device has a focal infrared light heating area (diameter-5 mm). The tail (⅓-½ way from distal end) of the animal was placed on the focal heating area. The temperature of the heat source was adjusted to elicit a tail-flick within 10 seconds in animals treated with vehicle. A 15 second cut-off time was used to prevent tissue damage, as is standard in the literature. The time elapsed between the start of the heat stimulus and any avoidance response was measured automatically and recorded for the test groups.
  • Hot plate test: The animal was placed on a 10″×10″ metal plate surrounded by 4 Plexiglas walls (15 inches high) and maintained at a temperature of 48-55° C. If the animal licked its hind paw, jumped, or vocalized, it was removed from the plate and the response latency was documented. If the animal did not show any response within 20-90 seconds (cut-off time), it was removed from the plate to prevent any possible tissue damage.
  • Formalin Flinching: Hindpaw injection of formalin-induced pain behavior (i.e. paw flinches) was measured using an automated “flinch response” measuring device UCSD. The device detects any sudden movement of a metal band glued onto one hind paw of the animal using a motion sensor installed underneath the device floor. One-half to one hour prior to formalin injection, a small metal band was attached to the plantar surface of one hind paw using a small drop of cyanoacrylate and the animal was placed in the testing chamber to be acclimatized. The attachment of the metal band did not appear to be irritating to the animal. Formalin (2.5%, 50 μL) was injected subcutaneously into the dorsum of the paw with the metal band. The animal was placed in the customized cylinder (25×10×20 cm, San Diego Instrument) immediately after intraplantar formalin injection. Paw flinches were recorded automatically.
  • In the acute thermal pain models, Protoxin-II variant 63955918 produced potent and prolonged analgesia as indicated by the elevated latency in the tail flick test (FIG. 11A and FIG. 11B) and hot plate test (FIG. 11C, FIG. 11D) after a single intrathecal administration. The significance and duration of the analgesia was dose-dependent.
  • Hindpaw formalin injection is a commonly used model for injury-induced pain. The injection induces a characteristic, bi-phasic flinching behavior, which indicates pain in test animals. As shown in FIG. 11E, animals pretreated with intrathecal injection of Protoxin-II variant 63955918 demonstrated fewer flinches in the formalin test, suggesting an inhibition of injury-induced pain.
  • Similarly, peptides NV1 D2775-OH and NV1D3034 demonstrated significant efficacy in the tail flick, hot plate and formalin test (FIG. 12A, FIG. 12B, FIG. 12C, FIG. 12D, FIG. 12E, FIG. 13A, FIG. 13B, FIG. 13C, FIG. 13D, FIG. 13E) following a single intrathecal administration.

Claims (26)

We claim:
1. An isolated Protoxin-II variant, comprising: a modified SEQ ID NO: 1, wherein amino acid numbering corresponds to amino acid numbering in SEQ ID NO: 1, comprising a W7Q substitution, a W30L substitution and a conserved M19 wherein the Protoxin-II variant is 30 or 32 amino acid residues and wherein, when the Protoxin-II variant is 32 amino acid residues, residues GP are at the amino-terminus of the Protoxin-II variant; and
wherein the variant possesses a potency comprising an IC50 value of 1×10−7 M or less, wherein the IC50 value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7.
2. The isolated Protoxin-II variant of claim 1, wherein the modified SEQ ID NO: 1 further comprises at least one of an N-terminal GP and a C-terminal extension.
3. The isolated Protoxin-II variant of claim 1, wherein the isolated Protoxin-II variant further comprises an N-terminal extension, wherein the N-terminal extension comprises at least one of SEQ ID NOs: 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384 and 385.
4. The isolated Protoxin-II variant of claim 2, wherein the modified SEQ ID NO: 1 further comprises a C-terminal extension, wherein the C-terminal extension is selected from the group consisting of: (i) at least one of SEQ ID NOs: 374, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396 and 397; and (ii) a C-terminal extension that is a sequence selected from the group consisting of SEQ ID NOs: 374, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, and 397 with at least one conservative amino acid substitution therein.
5. The isolated Protoxin-II variant of claim 1, further comprising:
an N-terminal extension; and
a linker conjugating the N-terminal to the modified SEQ ID NO: 1.
6. The isolated Protoxin-II variant of claim 5, wherein the linker comprises at least one of SEQ ID NOs: 383, 392, 398, 399, 400, 401 and 402.
7. The isolated Protoxin-II variant of claim 1, wherein the isolated Protoxin-II variant comprises the amino acid sequence of SEQ ID NOs: 56, 78, 111, 114, 117, 118, 119, 122, 123, 129, 130, 131, 132, 133, 134, 135, 136, 138, 139, 140, 141, 142, 145, 146, 147, 149, 150, 151, 152, 153, 154, 156, 158, 159, 165, 172, 173, 175, 177, 178, 183, 184, 185, 186, 189, 190, 193, 197, 199, 207, 210, 211, 216, 217, 224, 266, 273, 282, or 335.
8. The isolated Protoxin-II variant of claim 1 comprising the amino acid sequence GPQCX1X2WX3QX4CX5X6X7X8X9CCX10X11FX12CX13LWCX14KKLL (SEQ ID NO: 433), wherein
X1 is Q, R, K, A or S;
X2 is K, S, Q or R;
X3 is M or F;
X4 is T, S, R, K or Q;
X5 is D or T;
X6 is S, A or R;
X7 is E, R, N, K, T or Q;
X8 is R or K;
X9 is K, Q, S or A;
X10 is E, Q or D;
X11 is G or Q;
X12 is V or S;
X13 is R or T; and
X14 is K or R.
9. The isolated Protoxin-II variant of claim 1, further comprising at least one of a free C-terminal carboxylic acid, a free C-terminal amide, a free C-Terminal methylamide and a free C-terminal butylamide group.
10. An isolated fusion protein comprising the isolated Protoxin-II variant of claim 1 and a moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment.
11. An isolated conjugated polypeptide comprising the isolated Protoxin-II variant of claim 1 and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG).
12. An isolated polynucleotide encoding a Protoxin-II variant or fusion protein selected from the group consisting of:
(a) the isolated Protoxin-II variant of claim 1;
(b) an isolated fusion protein comprising the Protoxin-II variant of claim 10;
(c) an isolated Protoxin-II variant, comprising: a modified SEQ ID NO: 1, wherein amino acid numbering corresponds to amino acid numbering in SEQ ID NO: 1, comprising a W7Q substitution, a W30L substitution and a conserved M19 wherein the Protoxin-II variant is 30 or 32 amino acid residues and wherein, when the Protoxin-II variant is 32 amino acid residues, residues GP are at the amino-terminus of the Protoxin-II variant; and
wherein the variant possesses a potency comprising an IC50 value of 3×10−8 M or less, wherein the IC50 value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and
(c) an isolated Protoxin-II variant, comprising an amino acid sequence that is 90% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKVVMQTCDSERKCCEGMVCRLWCKKKLL-COOH) wherein the amino sequence comprises:
a Q or W at position 7, when residue numbering is according to SEQ ID NO: 1;
a L, F, Y, or W at position 30, when residue numbering is according to SEQ ID NO: 1, wherein the variant comprises 30 or 32 amino acids; and
a potency comprising an ICs value of about 30×10−9 M or less, wherein the ICs value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and wherein the conservative amino acid substitutions are selected from the group consisting of: (1) a substitution of one acidic amino acid for another acidic amino acid, wherein the acidic amino acids are aspartic acid and glutamic acid; (2) a substitution of one basic amino acid for another basic amino acid, wherein the basic amino acids are lysine, arginine, and histidine; (3) a substitution of one nonpolar amino acid for another nonpolar amino acid, wherein the nonpolar amino acids are alanine, valine, leucine, isoleucine, phenylalanine, methionine, and tryptophan; and (4) a substitution of one uncharged polar amino acid for another uncharged polar amino acid, wherein the uncharged polar amino acids are glycine, asparagine, glutamine, cysteine, serine, threonine, and tyrosine.
13. An isolated vector comprising the polynucleotide of claim 10.
14. An isolated host cell comprising the vector of claim 13.
15. A method of producing the isolated Protoxin-II variant, comprising culturing the host cell of claim 14 and recovering the Protoxin-II variant produced by the host cell.
16. A pharmaceutical composition comprising:
(a) a Protoxin-II variant, fusion protein, or conjugated polypeptide selected from the group consisting of:
(i) a Protoxin-II variant selected from the group consisting of:
(A) the isolated Protoxin-II variant of claim 1;
(B) an isolated Protoxin-II variant, comprising: a modified SEQ ID NO: 1, wherein amino acid numbering corresponds to amino acid numbering in SEQ ID NO: 1, comprising a W7Q substitution, a W30L substitution and a conserved M19 wherein the Protoxin-II variant is 30 or 32 amino acid residues and wherein, when the Protoxin-II variant is 32 amino acid residues, residues GP are at the amino-terminus of the Protoxin-II variant; and
wherein the variant possesses a potency comprising an IC50 value of 3×10−8 M or less, wherein the IC50 value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and
(C) an isolated Protoxin-II variant, comprising an amino acid sequence that is 90% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKVVMQTCDSERKCCEGMVCRLWCKKKLL-COOH) wherein the amino sequence comprises:
a Q or W at position 7, when residue numbering is according to SEQ ID NO: 1;
a L, F, Y, or W at position 30, when residue numbering is according to SEQ ID NO: 1, wherein the variant comprises 30 or 32 amino acids; and
a potency comprising an IC50 value of about 30×10−9 M or less, wherein the IC50 value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and wherein the conservative amino acid substitutions are selected from the group consisting of: (1) a substitution of one acidic amino acid for another acidic amino acid, wherein the acidic amino acids are aspartic acid and glutamic acid; (2) a substitution of one basic amino acid for another basic amino acid, wherein the basic amino acids are lysine, arginine, and histidine; (3) a substitution of one nonpolar amino acid for another nonpolar amino acid, wherein the nonpolar amino acids are alanine, valine, leucine, isoleucine, phenylalanine, methionine, and tryptophan; and (4) a substitution of one uncharged polar amino acid for another uncharged polar amino acid, wherein the uncharged polar amino acids are glycine, asparagine, glutamine, cysteine, serine, threonine, and tyrosine; or
(ii) a fusion protein selected from the group consisting of:
(A) a fusion protein comprising the isolated Protoxin-II variant of claim 1 and a moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment;
(B) a fusion protein comprising the isolated Protoxin-II variant of (a)(i)(B) and a moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment; and
(C) a fusion protein comprising the isolated Protoxin-II variant of (a)(i)(C) and a moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment; and
(iii) a conjugated polypeptide selected from the group consisting of:
(A) a conjugated polypeptide comprising the isolated Protoxin-II variant of claim 1 and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG);
(B) a conjugated polypeptide comprising the isolated Protoxin-II variant of (a)(i)(B) and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG); and
(C) a conjugated polypeptide comprising the isolated Protoxin-II variant of (a)(i)(C) and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG); and
(b) a pharmaceutically acceptable excipient.
17. A method of treating Nav1.7-mediated pain in a subject, comprising administering to a subject in need thereof an effective amount of a Protoxin-II variant, fusion protein, or conjugated polypeptide to treat the pain,
wherein the Protoxin-II variant is the isolated Protoxin-II variant of claim 1;
wherein the fusion protein is selected from the group consisting of:
(d) a fusion protein comprising the isolated Protoxin-II variant of claim 1 and a half-life extending moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment;
(e) a fusion protein comprising an isolated Protoxin-II variant, comprising: a modified SEQ ID NO: 1, wherein amino acid numbering corresponds to amino acid numbering in SEQ ID NO: 1, comprising a W7Q substitution, a W30L substitution and a conserved M19 wherein the Protoxin-II variant is 30 or 32 amino acid residues and wherein, when the Protoxin-II variant is 32 amino acid residues, residues GP are at the amino-terminus of the Protoxin-II variant; and
wherein the variant possesses a potency comprising an IC50 value of 3×108 M or less, wherein the IC50 value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7, and a moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment; and
(f) a fusion protein comprising an isolated Protoxin-II variant, comprising an amino acid sequence that is 90% identical to the amino acid sequence of SEQ ID NO: 422 (GPYCQKVVMQTCDSERKCCEGMVCRLWCKKKLL-COOH) wherein the amino sequence comprises:
a Q or W at position 7, when residue numbering is according to SEQ ID NO: 1;
a L, F, Y, or W at position 30, when residue numbering is according to SEQ ID NO: 1, wherein the variant comprises 30 or 32 amino acids; and
a potency comprising an IC50 value of about 30×10−9 M or less, wherein the IC50 value is measured using a veratridine-induced depolarization inhibition assay using fluorescence resonance energy transfer (FRET) in the presence of 25×10−6 M 3-veratroylveracevine in HEK293 cells stably expressing human Nav1.7; and wherein the conservative amino acid substitutions are selected from the group consisting of: (1) a substitution of one acidic amino acid for another acidic amino acid, wherein the acidic amino acids are aspartic acid and glutamic acid; (2) a substitution of one basic amino acid for another basic amino acid, wherein the basic amino acids are lysine, arginine, and histidine; (3) a substitution of one nonpolar amino acid for another nonpolar amino acid, wherein the nonpolar amino acids are alanine, valine, leucine, isoleucine, phenylalanine, methionine, and tryptophan; and (4) a substitution of one uncharged polar amino acid for another uncharged polar amino acid, wherein the uncharged polar amino acids are glycine, asparagine, glutamine, cysteine, serine, threonine, and tyrosine, and a moiety selected from the group consisting of a half-life extending moiety selected from the group consisting of human serum albumin (HSA), an albumin binding domain (ABD), and an Fc fragment; and
wherein the conjugated polypeptide is selected from the group consisting of:
(g) a conjugated polypeptide comprising the isolated Protoxin-II variant of claim 1 and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG);
(h) a conjugated polypeptide comprising the isolated Protoxin-II variant of (e) and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG); and
(i) a conjugated polypeptide comprising the isolated Protoxin-II variant of (f) and a half-life extending moiety wherein the half-life extending moiety is polyethylene glycol (PEG).
18. The method of claim 17, wherein the pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, postoperative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis.
19. The method of claim 17, wherein the Protoxin-II variant, fusion protein, or conjugated polypeptide is administered peripherally.
20. The method of claim 19, wherein the Protoxin-II variant, fusion protein, or conjugated polypeptide is administered locally to a joint, spinal cord, surgical wound, sites of injury or trauma, peripheral nerve fibers, urogenital organs, or inflamed tissues.
21. The method of claim 17, wherein the subject is a human.
22. A method of treating Nav1.7-mediated pain in a subject, comprising administering to a subject in need thereof an effective amount of the pharmaceutical composition of claim 16 to treat the pain.
23. The method of claim 22, wherein the pain is chronic pain, acute pain, neuropathic pain, nociceptive pain, visceral pain, back pain, postoperative pain, thermal pain, phantom limb pain, or pain associated with inflammatory conditions, primary erythemalgia (PE), paroxysmal extreme pain disorder (PEPD), osteoarthritis, rheumatoid arthritis, lumbar discectomy, pancreatitis, fibromyalgia, painful diabetic neuropathy (PDN), post-herpetic neuropathy (PHN), trigeminal neuralgia (TN), spinal cord injuries or multiple sclerosis.
24. The method of claim 22, wherein the pharmaceutical composition is administered peripherally.
25. The method of claim 24, wherein the pharmaceutical composition is administered locally to a joint, spinal cord, surgical wound, sites of injury or trauma, peripheral nerve fibers, urogenital organs, or inflamed tissues.
26. The method of claim 22, wherein the subject is a human.
US17/306,572 2015-03-03 2021-05-03 Protoxin-ii variants and methods of use Abandoned US20210363204A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/306,572 US20210363204A1 (en) 2015-03-03 2021-05-03 Protoxin-ii variants and methods of use

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201562127339P 2015-03-03 2015-03-03
US15/060,158 US20160257726A1 (en) 2015-03-03 2016-03-03 Protoxin-ii variants and methods of use
US15/583,793 US10995125B2 (en) 2015-03-03 2017-05-01 Protoxin-II variants and methods of use
US17/306,572 US20210363204A1 (en) 2015-03-03 2021-05-03 Protoxin-ii variants and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/583,793 Division US10995125B2 (en) 2015-03-03 2017-05-01 Protoxin-II variants and methods of use

Publications (1)

Publication Number Publication Date
US20210363204A1 true US20210363204A1 (en) 2021-11-25

Family

ID=56849056

Family Applications (3)

Application Number Title Priority Date Filing Date
US15/060,158 Abandoned US20160257726A1 (en) 2015-03-03 2016-03-03 Protoxin-ii variants and methods of use
US15/583,793 Active US10995125B2 (en) 2015-03-03 2017-05-01 Protoxin-II variants and methods of use
US17/306,572 Abandoned US20210363204A1 (en) 2015-03-03 2021-05-03 Protoxin-ii variants and methods of use

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US15/060,158 Abandoned US20160257726A1 (en) 2015-03-03 2016-03-03 Protoxin-ii variants and methods of use
US15/583,793 Active US10995125B2 (en) 2015-03-03 2017-05-01 Protoxin-II variants and methods of use

Country Status (15)

Country Link
US (3) US20160257726A1 (en)
EP (1) EP3265476A4 (en)
JP (2) JP2018512123A (en)
KR (1) KR20170120703A (en)
CN (1) CN107531769A (en)
AR (1) AR103838A1 (en)
AU (2) AU2016226443B2 (en)
BR (1) BR112017018834A2 (en)
CA (1) CA2978435A1 (en)
HK (1) HK1249118A1 (en)
IL (2) IL254273A0 (en)
MA (1) MA41642A (en)
MX (1) MX2017011223A (en)
TW (1) TW201706292A (en)
WO (1) WO2016140859A2 (en)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10414808B2 (en) 2012-05-18 2019-09-17 Janssen Biotech, Inc. Huwentoxin-IV variants and methods of use
WO2015051216A1 (en) 2013-10-03 2015-04-09 Janssen Biotech, Inc. Protoxin-ii variants and methods of use
CN107106280A (en) 2014-12-23 2017-08-29 奥特梅德私人有限公司 Conveying equipment, system and associated method
MA41642A (en) 2015-03-03 2018-01-09 Janssen Biotech Inc PROTOXIN II VARIANTS AND METHODS OF USE
EP3277304B1 (en) 2015-04-02 2021-08-04 Janssen Biotech, Inc. Protoxin-ii variants and methods of use
CN109369785B (en) * 2018-11-14 2019-08-30 青海芬陀利华生物科技有限公司 Ccotx1 toxin and its application
CN109369784B (en) * 2018-11-14 2019-08-30 青海芬陀利华生物科技有限公司 PaTx-1 toxin and its application
US11957542B2 (en) * 2020-04-30 2024-04-16 Automed Patent Holdco, Llc Sensing complete injection for animal injection device
CN114957431B (en) * 2022-06-27 2023-06-20 四川丽妍工坊生物科技有限公司 Skin anti-wrinkle polypeptide Cj2a2, preparation method and application

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4309989A (en) 1976-02-09 1982-01-12 The Curators Of The University Of Missouri Topical application of medication by ultrasound with coupling agent
US4767402A (en) 1986-07-08 1988-08-30 Massachusetts Institute Of Technology Ultrasound enhancement of transdermal drug delivery
CA2285658C (en) 1997-04-16 2011-10-04 Nozar M. Mehta Direct expression of peptides into culture media
IL120943A (en) 1997-05-29 2004-03-28 Univ Ben Gurion Transdermal delivery system
US6670127B2 (en) 1997-09-16 2003-12-30 Egea Biosciences, Inc. Method for assembly of a polynucleotide encoding a target polypeptide
ATE462004T1 (en) 1997-09-16 2010-04-15 Centocor Inc METHODS FOR COMPLETE CHEMICAL SYNTHESIS AND ASSEMBLY OF GENES AND GENOMES
EP1348466A3 (en) 2002-02-01 2003-10-29 Inspire Pharmaceuticals, Inc. Method for treating pain with adenosine-tetraphosphates
US7659082B2 (en) 2002-02-19 2010-02-09 Xenon Pharmaceuticals Inc. Methods for identifying analgesic agents
US7833979B2 (en) 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
EP1942948A4 (en) 2005-11-04 2010-03-03 Alnylam Pharmaceuticals Inc Compositions and methods for inhibiting expression of nav1.8 gene
WO2007109324A2 (en) 2006-03-21 2007-09-27 Xenon Pharmaceuticals, Inc. Potent and selective nav 1.7 sodium channel blockers
CA2662453A1 (en) 2006-09-08 2008-03-13 Mayo Foundation For Medical Education And Research Aquaretic and natriuretic polypeptides lacking vasodilatory activity
WO2008033564A1 (en) 2006-09-15 2008-03-20 Hydra Biosciences Inc. Compounds for modulating trpv3 function
US7803769B2 (en) 2006-10-25 2010-09-28 Amgen Inc. OSK1 peptide analogs and pharmaceutical compositions
CN102202677A (en) 2008-09-03 2011-09-28 阿尔伯维塔公司 Agents and methods for treatment of pain
WO2010104115A1 (en) 2009-03-10 2010-09-16 独立行政法人産業技術総合研究所 Method for preparing polypeptide specifically recognizing membrane protein
CN102573884B (en) 2009-09-15 2015-01-14 阿洛莫恩临床有限公司 Novel peptides isolated from spider venom, and uses thereof
WO2012004664A2 (en) * 2010-07-07 2012-01-12 Purdue Pharma L.P. Analogs of sodium channel peptide toxin
CN101979411A (en) 2010-10-21 2011-02-23 湖南师范大学 Ornithoctonus huwena protease inhibitor
KR20140009311A (en) 2011-01-18 2014-01-22 암젠 인크 Nav1.7 knockout mice and uses thereof
JP6771282B2 (en) 2012-05-18 2020-10-21 ヤンセン バイオテツク,インコーポレーテツド Fentoxin IV mutant and how to use it
US10414808B2 (en) 2012-05-18 2019-09-17 Janssen Biotech, Inc. Huwentoxin-IV variants and methods of use
US9102751B2 (en) * 2012-05-18 2015-08-11 Janssen Biotech, Inc. Huwentoxin-IV variants and methods of use
US9718865B2 (en) 2012-07-27 2017-08-01 Purdue Pharma L.P. Sodium channel blocking peptides and the use thereof
US10344060B2 (en) 2013-03-12 2019-07-09 Amgen Inc. Potent and selective inhibitors of Nav1.7
CN104065367B (en) 2013-03-20 2017-11-07 江苏多维科技有限公司 A kind of low-watt consumption magnetic resistance switch sensor
WO2015051216A1 (en) 2013-10-03 2015-04-09 Janssen Biotech, Inc. Protoxin-ii variants and methods of use
MA41642A (en) 2015-03-03 2018-01-09 Janssen Biotech Inc PROTOXIN II VARIANTS AND METHODS OF USE
EP3277304B1 (en) 2015-04-02 2021-08-04 Janssen Biotech, Inc. Protoxin-ii variants and methods of use

Also Published As

Publication number Publication date
EP3265476A2 (en) 2018-01-10
MA41642A (en) 2018-01-09
HK1249118A1 (en) 2018-10-26
WO2016140859A3 (en) 2016-10-27
KR20170120703A (en) 2017-10-31
IL282482A (en) 2021-06-30
AR103838A1 (en) 2017-06-07
US20160257726A1 (en) 2016-09-08
CN107531769A (en) 2018-01-02
MX2017011223A (en) 2018-06-20
WO2016140859A2 (en) 2016-09-09
AU2021200400A1 (en) 2021-03-18
US10995125B2 (en) 2021-05-04
EP3265476A4 (en) 2018-08-22
BR112017018834A2 (en) 2018-04-17
CA2978435A1 (en) 2016-09-09
JP2022071050A (en) 2022-05-13
JP2018512123A (en) 2018-05-17
AU2016226443B2 (en) 2020-10-22
AU2016226443A1 (en) 2017-09-21
TW201706292A (en) 2017-02-16
US20170334959A1 (en) 2017-11-23
IL254273A0 (en) 2017-10-31

Similar Documents

Publication Publication Date Title
US20210363204A1 (en) Protoxin-ii variants and methods of use
US10975129B2 (en) Protoxin-II variants and methods of use
US11083776B2 (en) Protoxin-II variants and methods of use

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

AS Assignment

Owner name: JANSSEN BIOTECH, INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:FLINSPACH, MACK;WICKENDEN, ALAN;SIGNING DATES FROM 20160307 TO 20160308;REEL/FRAME:057466/0611

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION