Introduction to Enteris BioPharma, Inc

Introduction to Enteris BioPharma, Inc. Enteris BioPharma • Privately held, New Jersey based biotech company • Owned by Victory Park Capital, a ...
Author: Peter Chandler
9 downloads 3 Views 3MB Size
Introduction to Enteris BioPharma, Inc.

Enteris BioPharma



Privately held, New Jersey based biotech company



Owned by Victory Park Capital, a Chicago based investment firm



Clinically validated oral formulation technology • for peptides and challenging small molecules



Extensive scientific know-how and R&D experience



Proven GMP tablet manufacturing capabilities

2

Enteris BioPharma



Enteris has effectively addressed both permeability and solubility challenges with a simple, elegant and scalable solution



Demonstrated a track record of clinical success across a range of compounds and therapeutic indications



Enteris offers robust IP protection, regulatory CMC support and finished, solid dosage formulations for preclinical and early phase clinical studies

3

Clinically Validated Oral Delivery Technology •

Clinically validated oral peptide delivery technology • Positive Phase 3 oral Calcitonin: Osteoporosis(1)

• Positive Phase 2 oral PTH: Osteoporosis(2) • Positive Phase 2 oral Calcitonin: Osteopenia(3)

• Positive Phase 1 oral CR845: Neuropathic Pain(4) •

• • • •

Sponsored preclinical peptide programs • 15 ongoing or completed formulation programs

(1) Tarsa Therapeutics, Inc. (JBMR 27, No.8, 2012, 1821-1829) (2) Unigene Laboratories, Inc. (Bone 53, 2013, 160-166) (Clin Pharm 52, No. 6, 2013) (3) Tarsa Therapeutics, Inc. (ASBMR, 2012) (4) Cara Therapeutics, Inc. (data on file)

4

Actual Bioavailability Data Peptides and Small Molecules 25

Absolute Bioavailability (%)

proprietary peptide

20

15

zanamivir tigecycline CR-845 kanamycin tobramycin

10 proprietary peptides proprietary peptide

5

octreotide *GLP-1 analog

0

0 Studies in beagle dogs Except *Rat Study

1000

2000

calcitonin

PTH 1-34

3000

4000

insulin

5000

6000

Molecular Weight (Da) 5

Criteria for Selection of Peptides

ENTERIS PEPTIDE ASSESSMENT MATRIX Molecular Weight (Da) • Sequence of peptide • Hydrodynamic radius (Å) • Cyclic peptide?

Solubility • Acid

• Water • Buffers/salts

• FaSSIF/FeSSIF

Chemical Stability • x-S-S-x Resistant to Proteolysis?

Modified? Physical Stability • Aggregation rate Projected Dose (mg)

Injectable Dose (mg)

Net Charge • Anionic charges

Bioavailability (%) if known

• Cationic charges

• Rat

• pI

• Dog 6

Criteria for Selection of Small Molecules

ENTERIS SMALL MOLECULE ASSESSMENT MATRIX Molecular Weight (Da)

Moisture sensitive/hygroscopic

BCS Class, if known

Is the API plastic, elastic or brittle?

Describe the solid (salt form, hydrate, solvate, etc.

Any known excipient incompatibilities?

Solubility

Primary mechanism(s) of chemical degradation



pH/Solubility curve available?



Water



Buffers/Solvents/Emulsions



If insoluble, what is the most likely reason? (hydrophobicity/crystal surface energy/etc.)

Are other salts or polymorphs available? Is the API crystalline or amorphous? •

Has polymorph transition been observed during processing or on stability

Caco-2 Permeability • Apical to Basolateral, Basolateral to Apical? Identify known transporter interactions, (P-gp, BCRP, MRP family, PepT1, OATP, etc.) What is the primary clearance mechanism(s)? (renal/Biliary/Metabolic) If metabolic, what enzymes predominate? Bioavailability of current oral formulation in development?

Please describe any PO formulation attempted thus far 7

Contents



Introduction



Mechanism



Safety • Excipient Safety Profile

• Peptides

• LLC Toxicology Studies

• Small molecules

• LLC Regulatory and Clinical

• BCS Class III



Patents

• BCS Class II



Business Development



Manufacturing

8

Mechanism of Drug Delivery

9

Enteric Coat Prevents Tablet from Opening in Stomach at Low pH



Acid-stable enteric coating prevents tablet release in stomach • Less susceptible to food effects or dilution with liquids • API protected from degradation by acid and pepsin • Peptides

• Acid-labile small molecules

10

Enteric Coat Dissolves at Neutral pH in the Small Intestine



Water-soluble sub-coat acts as a partition layer between the enteric

coat and the acidic tablet core • Simultaneous release of API and excipients

11

pH Modifier, Permeability Enhancers and API Released



Organic acid sequestered in coated beads • Increases stability of tablet formulation • Compatible with peptides and small molecules

• Acts as protease inhibitor for peptides • Calcium chelator and membrane permeation enhancer • pH-lowering agent that increases absorptive flux • Membrane wetting/charge dispersal agent

12

API Absorbed Across Intestinal Wall via Paracellular Transport •

Lauroyl-L-carnitine (12-carbon fatty acid) • Modulates tight junctions in the intestinal enterocytes and enhances paracellular transport • Acts as a solubilizing agent due to surfactant properties • Inhibits P-gp efflux transporters

13

Components of Enteris’ Solid Dosage Formulation

14

Peptide Experience

15

Dog Model Predicts Bioavailability in Humans 4500 4000

Linear Regression for Cmax: Y=A+B*X 2 R =0.9801

1000

Human Cmax (pg/ml)

Plasma Cmax (pg/ml)

5000

3500 3000 2500

2000 1500 1000 500 0 0

1

2

3

Dose (mg)

4

5

y=.995x - 0.7548 R=0.958 R2=0.918

100

10

10

100

1000

10000

Dog Cmax (pg/ml)

Enteris' dog model for oral delivery shows high degree of linearity with respect to dose offering a wide range of dosing strategies. Comparability of PK results in dog and human shows that Enteris’ dog model is an appropriate success predictor for human studies

16

Peptide Bioavailability Formulated vs. Unformulated

Effect of Formulation on the Bioavailability of Various Peptides in Pre-Clinical Animal Models Program Type

Sponsored

No. of Amino Acids

Study Type

Ranges of Unformulated Bioavailability (%) for 12 Peptides

Ranges of Formulated Bioavailability* (%) for 12 Peptides

Rat

0.35 – 6.0

10.7 – 26.0

Dog

100

100 – 1000

Enteric Toxins

70 – 900

Viruses

600 – 1000

Bacteria

> 1000

Modeling of perturbed membrane in Caco-2 cells indicates an effective pore radius ca. 20Å

Modified from: Brayden, D. Permeation Enhancers and Oral Peptide Delivery. Presented at the Roche Colorado Peptide Symposium, Sept. 12, 2011. 55

Tight Junction Modifiers are Common • Drug Compounds: • Aspirin • NSAIDS • Phenothiazines

• Food and Drug Additives/Excipients: • • • •

EDTA C8-C18 fatty acids Various polymers Poly-L-lysine

• Natural/Food Products: • ZOT • ATP • Chitosan and chitosan derivatives • Wheat gluten • Oat saponins • Capsaicin • Alcohol

56

LLC Toxicology Studies

57

3-O-Lauroyl-L-Carnitine Preclinical Safety Studies •

Completed Safety Pharmacology Studies • • • • • •



Acute neurotoxicity in rat Acute respiratory in rat Acute cardiovascular in dog hERG CYP450 inhibition/ induction Metabolic profiling in hepatocytes (multiple species)

Completed Toxicology Studies • • • • • •

Oral MTD studies in rat and dog 4 day oral repeat dose finding in rat and dog 1 month oral toxicology with toxicokinetics in rat and dog Standard genotoxicity 6 month oral repeat dose in rat 9 month oral repeat dose in dog 58

All Safety Pharmacology Studies Generated Desirable Outcomes • Lauroyl-L-Carnitine did not: • Inhibit hERG tail current in vitro, at doses up to ca. 90 μg/mL • Affect ECG parameters in dogs dosed up to 100 mg/kg/day PO for 1 month • Produce cardiovascular effects in dogs at up to 100 mg/kg • Induce any respiratory effects in rats at up to 100 mg/kg by gavage • Produce any adverse effects on neurobehavioral function in rats at up to 100 mg/kg

59

LLC Regulatory and Clinical

60

FDA Feedback on LLC program

• FDA Advice Letter received August, 2009 ...the studies that have been completed to date in your development program and the proposed six- and ninemonth repeat dose studies in rats and dogs respectively, would serve to support the use of LLC as an excipient in drug products.

61

Registration Requirements

• Reproductive toxicology • Studies required for populations of premenopausal females, women of child bearing potential, or males

• Embryo-fetal studies – for registration • Recommended embryo-fetal studies in two species, with an assessment of teratogenicity as a minimum to complete drug approval applications in postmenopausal women

• Carcinogenicity – likely not required • The nonclinical findings to date could support the proposal that carcinogenicity studies are not necessary • If there are no preneoplastic lesions or serious adverse toxicologic effects in these studies, FDA would concur that carcinogenicity studies will not be necessary (None observed – reports submitted Sept, 2012)

62

LLC Type V DMF Submitted to FDA

“3-O-Lauroyl-L-Carnitine Hydrochloride (LLC) Preclinical and Clinical Data.” • Contains full study reports of all non-clinical and clinical studies • Non-clinical and clinical overview documents • Available to partners for cross-referencing

63

Summary



Extensive preclinical toxicology package

• Observed no preneoplastic lesions or serious adverse toxicologic effects • Completed toxicology study of 9 months duration •

Extensive Clinical Experience • < 8 single dose Phase 1 studies • 8 week Phase 2a study for oral sCT program • 24 week Phase 2 study for oral PTH program



Type V DMF containing safety data of LLC

64

Patents

65

Patents Summary

Oral Delivery Patents •

7 issued U.S. patents



2 allowed U.S. patent applications



3 pending U.S. provisional patent applications



29 issued Foreign patents



5 pending Foreign patent applications



Key issued patents extend through 2030

66

Business Development

67

Working with Enteris

Formulation Development

In-Vitro • Solubility • Permeability In-Vivo • Intra-duodenal: Rat Study In-Vivo • Capsule: Dog Study

Tablet Formulation Optimization

3 months

In-Vitro & In-Vivo • Stability; Design Space • Tablet: Dog Study

3 months

68

Manufacturing

69

Manufacturing

• Enteris cGMP Manufacturing • • • •

32,000 ft2 cGMP facility located in Boonton, NJ Separate tableting and nasal spray filling suites Full QA/QC and regulatory support Commercial product in US distribution 70

Recent Technical Achievements



Identified coated organic acid as compatible excipient with peptides and small molecules



Simple and scaleable manufacturing process



Optimized release characteristics and bioavailability



Demonstrated room temperature stability of peptide tablets for 24 months



Supplied CTM for Phase 1 and Phase 2 studies

71

Tableting and Capsuling Line

• • • • • • • •

Comil conical mill V-Blender Korsh XL-100 10 station tablet press - up to 10,000 tablets/hr Natoli NP-RD10a single station press for 1 to 300 tablets Vector LDCS coating pan for enteric coating Capsugel Profill capsule filler Phase 1 and 2 clinical supplies Clinical packaging •

Open label, double blind

72

Quality Control

• • • • • • •

Raw Material Release Testing Final Product Release Assay Development Assay Transfer, Optimization & Validation Special Projects and Investigations Stability Studies In process, intermediate and facility testing capabilities

73

Quality Assurance



Systems compliant with FDA, EMEA, MHRA and ICH



Customer focused, providing real time feedback on all quality related issues.



Full project participation: • process development → batch record design → GMP manufacture.



Manufacturing oversight through concurrent batch record review.



Vendor auditing and qualification program.

74

Inspection History

Regulatory Inspection History • Most recent FDA inspection – October, 2011 - No FDA-483 issued • Successful Pre-Approval Inspection – June, 2003



Most recent EMA inspection – October, 2006 - GMP Certificate issued • Successful Pre-Approval Inspection – February, 1998



GMP inspections by QPs from UK & Germany for Phase 3 - Clinical Trial – 2008 • GMP Certificates issued

75

E. coli Recombinant Peptide Manufacturing

DirectExpression ExpressionofofPeptides Peptides Direct • • •

Patented E. coli-based recombinant peptide expression technology Peptide secreted directly into culture medium Enzymatic amidation of recombinant precursor

Enteris’ Peptide Manufacturing Advantages • • • •

Robust yield and purity Scalable Efficient Cost-effective

76

Enteris’ Recombinant Manufacturing Process

77

High Yield and High Product Purity at Low Cost

PTH analog-gly

Glucose regulatory peptide analog-gly

Glucose regulatory peptide-gly

kDa

sCTgly

Recombinant Peptide Manufacturing

66.2 45.0 31.0 21.5 14.4 6.5

SDS PAGE and HPLC Trace of Crude Medium • Enriched Starting Material • Extracellular yields of 400 to 1300 mg/L • Reduced purification steps 78

Amidated Peptide Manufacturing Example rhPTH(1-34)NH2 Step 1. Recombinant production of rhPTH(1-34)Gly35OH precursor The peptide: human PTH(1 -34)-Gly

rhPTH(1-34)GLY Production and Fermentation Feed Rate

SVSEIQLMHNLGKHLNSMERVENLRKKLQDVHNFG MW = 4,116.8 Da P1 P2

KAN-R

ompA

800

0.300

PTH(1-34)gly 700

Term

0.250

P1

PTH(1-34)gly

pPTH(1-34)G-03

Term

7799 bp

P3

Ori

PRLA-4

0.200 PTH(1-34) mg/L

ompA

500 400

0.150

300

Feed Rate mL/min

600

P2

0.100 200 0.050 100

Term SecE P/O LAC-IQ

SEC-E

0

0.000 1

5

9

13

17

21

25

29

Hours Post Induction

Term

The host : E. coli B strain, BLM-6

PTH(1-34) mg/L

Feed Rate ml/min

79

Thank You

Suggest Documents