DESIGN AND EVALUATION OF A NON-OPIOID TRIPARTITE RELEASE TABLET FOR CHRONIC INFLAMMATORY PAIN. KUNDAI ROSELYN MAZARURA A dissertation submitted to the Faculty of Health Sciences, University of the Witwatersrand, in fulfilment of the requirements for the degree of Master of Pharmacy Supervisor Prof. Yahya E. Choonara Department of Pharmacy and Pharmacology, University of the Witwatersrand, South Africa Co-Supervisors Prof. Pradeep Kumar Department of Pharmacy and Pharmacology, University of the Witwatersrand, South Africa Dr. Armorel van Eyk Department of Pharmacy and Pharmacology, University of the Witwatersrand, South Africa 2024 ii DECLARATION I, Mazarura R. Kundai, declare that this dissertation is my own work. It has been submitted for the degree of Master of Pharmacy in the Faculty of Health Sciences at the University of the Witwatersrand, Johannesburg, South Africa. It has not been submitted before for any degree or examination at this or any other University. This 16th day of April 2024. X Kundai Mazarura Ms iii PUBLICATION Kundai R. Mazarura, Pradeep Kumar & Yahya E. Choonara (2022): Customized 3D printed multi-drug systems: an effective and efficient approach to polypharmacy, Expert Opinion on Drug Delivery, DOI: 10.1080/17425247.2022.2121816 To link to this article: https://doi.org/10.1080/17425247.2022.2121816 https://doi.org/10.1080/17425247.2022.2121816 iv ABSTRACT Formulation-based approaches towards curbing the prescription opioid crisis include the discovery and development of non-opioid analgesics such as the novel benzyloxy- cyclopentyladenosine (BnOCPA). A more expedited approach involves the development of combinatorial systems of already existing non-addicting analgesics to tap into unexplored synergistic potentials. Despite the recent advances in drug delivery systems, tablets still hold the position of being the most widely used oral dosage form, particularly in the management of chronic ailments; it is cost-effective, non-invasive, and does not require administration expertise. Challenges in the production of complex geometry combinatorial, multi-drug tablets remain to some extent enigmatic to pharmaceutical researchers, hence the steady paradigm shift from traditional compression to 3-dimensional printing. Although it is superior in multiple aspects, the technique is still in its nascent stages with limited information on regulatory guidelines. Therefore, the aim of this work was to design and develop a non-opioid tripartite controlled- release tablet for efficient chronic inflammatory pain management. Because adherence to adjunct gastroprotective agents (GPAs) in non-steroidal anti-inflammatory drugs (NSAIDs) users has been established to be suboptimal, esomeprazole magnesium trihydrate (ESM) was added to the drug delivery system (DDS). The rationale behind the design was based on inherent drug properties, target release sites, desired therapeutic effects, and allowance for drug release manipulation, therefore a tablet was assembled, constituting an immediate- release top layer formulation of 250 mg paracetamol (PAR) for an early onset of analgesia; a cup layer for the delayed and retarded release of 100 mg of diclofenac sodium (DS) and 250 mg of PAR in tandem, and lastly a core containing a press-coated 20 mg ESM pill. A reproducible and efficient Reverse-Phase High-Performance Liquid Chromatographic (RP- HPLC) method was developed and validated for the simultaneous detection of the APIs over the concentration ranges studied. Deleterious drug-excipient incompatibilities were ruled out through pre-formulation investigations by FTIR, DSC, and TGA analyses. Combining both wet and dry granulation methodologies; the chosen formulation and polymers (7.5% hydroxypropyl methylcellulose (HPMC) K15M, 25.3% eudagrit L (EL) 100-55, and 10.5% croscarmellose sodium (CCS)), while considering the quality target product profiles (QTPPs), critical process parameters (CPP), and critical material attributes (CMAs), resulted in the development of a pragmatic tablet delivering fifty percent of the PAR dosage in the initial 30 minutes, with a cumulative release of 95.0% ± 0.08% and 94.9% ±3.87% for DS and ESM, respectively. Through in-process quality control tests, the validity of the manufacturing process was confirmed, with all results falling within pharmacopeial specifications. The release mechanism of PAR and DS from the cup after the 2-hour mark distinctly followed the Hixson-Crowell model where the geometrical characteristic of the cup was maintained with surface erosion. Visuals from scanning electron microscopy (SEM) analysis obtained prior to and during dissolution, confirmed hydration gravimetric analysis results as well as bulk and surface erosion mechanisms. The obtained ex vivo analysis results showed retarded permeation rates of the tabletted APIs compared to the APIs in their pure state. Therefore, it is imperative to consider improving the existing models employed for ex-vivo permeability studies of tableted formulations, with a particular focus on exploring the impact of excipients/polymers on drug permeation. v ACKNOWLEDGEMENTS My sincere appreciation goes to my esteemed supervisors for not only their invaluable support and supervision but also for entrusting me with this project and allowing me to live out my passion for medicine manufacturing. Through their redirections and solutions, drawn from their immense knowledge pools and experience I managed to see the project to the end. I also want to thank the WADDP researchers for creating an environment that is conducive to research, inspiring, and positively influential in every way, with specific acknowledgment of Ahmed Abdelgader for generously sharing his expertise on the operation of the tablet press machine and much more. Despite having a limited understanding of what this stage of my academic path entailed, my parents were highly supportive in every way possible, for which I am exceedingly grateful. I'd want to thank my friends and brother Tapiwa Mazarura, (fellow academics) who brought joy to what could have been a lonely 2-3 years of my life. The 2023 academic year would not have been possible without the scholarship I received from MINDS; my thanks go to them as well. Above all, I thank God the Father, Son, and Holy Spirit, “in whom I live and move and have my being." I thank Him for keeping me safe in the late-night hours on the days the research demanded it. vi DEDICATION This work is dedicated to my late grandfathers, my paternal grandfather, who treasured knowledge acquisition even in his old age, and my maternal grandfather, who died in pain in 2021 with the conviction that I could make him a magical pill with no side effects to alleviate his pain. vii TABLE OF CONTENTS DECLARATION ......................................................................................................................ii PUBLICATION ...................................................................................................................... iii ABSTRACT ........................................................................................................................... iv ACKNOWLEDGEMENTS ..................................................................................................... v DEDICATION ........................................................................................................................ vi TABLE OF CONTENTS ....................................................................................................... vii LIST OF FIGURES............................................................................................................... xii LIST OF TABLES ................................................................................................................. xv LIST OF ABBREVIATIONS ................................................................................................ xvii CHAPTER 1 INTRODUCTION AND MOTIVATION FOR THE STUDY 1.1 Background ................................................................................................................. 1 1.2 Rationale Behind the Design and Combination ............................................................ 3 1.3 Aims and Objectives of the Study. ................................................................................ 5 1.4 Overview of This Dissertation ...................................................................................... 6 CHAPTER 2 LITERATURE REVIEW ON NOVEL FORMULATION TECHNIQUES FOR CONTROLLED RELEASE MULTI-DRUG SYSTEMS 2.1 Introduction .................................................................................................................. 8 2.2 Summary on 3D Printing Technologies ...................................................................... 10 2.3 Customisation in Different Population Groups............................................................ 11 2.3.1 Visually Impaired Patients ................................................................................... 11 2.3.2 Paediatric Patients .............................................................................................. 12 2.3.3 Geriatric Patients ................................................................................................. 12 2.4 3D Printed Multi-Drug Dosage Forms ........................................................................ 13 2.4.1 3D Printed Oral Multi-Drug Formulations ............................................................. 13 2.4.1.1 Formulations Printed by Fused Deposition Modelling ................................... 13 2.4.1.2 Formulations Printed by Pressure Assisted Microsyringe Extrusion .............. 16 viii 2.4.1.3 Formulations Printed by Stereolithography ................................................... 17 2.4.1.4 Formulations Printed by Selective Laser Sintering ....................................... 17 2.4.2 3D Printed Multi-Drug Eluting Implants ................................................................ 20 2.5 Additive Materials Used in 3D Printing of Multi-Drug Systems ................................... 21 2.5.1 Other High Functionality Polymers ...................................................................... 25 2.6 Limitations and Barriers to Implementation ................................................................ 25 2.6.1 Regulatory Constraints ........................................................................................ 25 2.6.2 Processing Parameters ....................................................................................... 26 2.6.2.2 Printer Accuracy ........................................................................................... 26 2.6.2.3 Return on Investment (ROI) .......................................................................... 26 2.7 Cost Implications and Projected Market Views .......................................................... 27 2.8 Conclusion ................................................................................................................. 28 2.9 Expert Opinion ........................................................................................................... 30 CHAPTER 32 PRE-FORMULATION INVESTIGATIONS AND LIQUID CHROMATOGRAPHIC METHOD DEVELOPMENT AND VALIDATION FOR THE SIMULTANEOUS QUANTITATION OF THE ACTIVE INGREDIENTS 3.1 Introduction ................................................................................................................ 32 3.2 Materials and methods .............................................................................................. 33 3.2.1 Materials ............................................................................................................. 33 3.2.2 Fourier Transform Infrared Spectroscopic Analysis ............................................. 34 3.2.3 Differential Scanning Calorimetry Analysis .......................................................... 34 3.2.4 Thermogravimetry Analysis ................................................................................. 34 3.2.5 HPLC Method Development and Validation ........................................................ 34 3.2.5.1 Wavelength Selection ................................................................................... 35 3.2.5.2 Method Development and Optimization ........................................................ 36 3.2.5.3 Method Validation ......................................................................................... 36 3.2.5.3.1 Preparation of Standards ....................................................................... 36 3.2.5.3.2 Linearity, LOD, and LOQ Determination ................................................. 36 ix 3.2.5.3.3 Method Precision Analysis ..................................................................... 37 3.2.5.3.4 Specificity Analysis ................................................................................. 37 3.2.5.3.5 Accuracy Analysis .................................................................................. 38 3.2.5.3.6 Robustness Analysis .............................................................................. 38 3.2.5.3.7 Stability Analysis .................................................................................... 38 3.3 Results ...................................................................................................................... 39 3.3.1 FTIR Analysis ...................................................................................................... 39 3.3.2 DSC and TGA Analysis ....................................................................................... 40 3.3.3 Method Development and Validation ................................................................... 42 3.3.3.1 Method Development .................................................................................... 42 3.3.3.2 Method Validation ......................................................................................... 43 3.3.3.2.1 Linearity, LOD, and LOQ ............................................................................ 43 3.3.3.2.2 Method Precision and Accuracy ............................................................. 45 3.3.3.2.3 Robustness Analysis .............................................................................. 46 3.3.3.2.4 Stability Analysis .................................................................................... 47 3.3.3.2.5 Specificity/Selectivity Analysis ................................................................ 48 3.4 Conclusion ................................................................................................................. 48 CHAPTER 4 FORMULATION DEVELOPMENT AND COMPARATIVE IN VITRO DRUG RELEASE ANALYSIS 4.1 Introduction ................................................................................................................ 50 4.2 Materials and Methods .............................................................................................. 52 4.2.1 Materials ............................................................................................................. 52 4.2.2 Formulation Development ................................................................................... 52 4.2.3 Pre-Compression Evaluation ............................................................................... 55 4.2.4 Assemblage of the Full DDS ............................................................................... 56 4.2.5 In Vitro Drug Release Studies ............................................................................. 56 4.3 Results ...................................................................................................................... 57 4.3.1 Pre-Compression Powder Flow Analysis ............................................................. 57 x 4.3.2 Dissolution results and influence of polymers ...................................................... 58 4.3.2.1 IR Layer: Influence of CCS and PVP-K30 Concentrations ............................ 58 4.3.2.2 DR Cup ......................................................................................................... 59 4.3.2.3 Core and Plug Efficiency ............................................................................... 62 4.4 Conclusion ................................................................................................................. 65 CHAPTER 5 IN-PROCESS EVALUATION POST-COMPRESSION ANALYSIS AND CHARACTERISATION OF THE FINAL FORMULATION 5.1 Introduction ................................................................................................................ 66 5.2 Materials and Methods .............................................................................................. 68 5.2.1 Materials ............................................................................................................. 68 5.2.2 Preparation of the Drug Delivery System ............................................................ 68 5.2.3 In-Process Quality Control Evaluation ................................................................. 70 5.2.4 Indentation Hardness Test .................................................................................. 70 5.2.5 Gravimetric Analysis Through Water Uptake and Erosion Studies ...................... 71 5.2.6 Determination of polymeric structural variations using Fourier Transmission Infrared spectroscopy .................................................................................................. 72 5.2.7 Determination of the thermal behaviour ............................................................... 72 5.2.8 Determination of surface morphology using Scanning Electron Microscopy ........ 72 5.2.9 In Vitro Drug Release Analysis ............................................................................ 73 5.3 Results ...................................................................................................................... 73 5.3.1 In-Process Quality Control Evaluation ................................................................. 73 5.3.2 Indentation Hardness Test .................................................................................. 74 5.3.3 Gravimetric Analysis ........................................................................................... 74 5.3.4 Polymeric Structural Variations Analysis ............................................................. 75 5.3.5 Determination of the Thermal Behaviour ............................................................. 76 5.3.6 Determination of Surface Morphology Using Scanning Electron Microscopy ....... 78 5.3.7 In Vitro Drug Release Analysis ............................................................................ 80 5.4 Conclusion ................................................................................................................. 83 xi CHAPTER 6 EX VIVO PERMEABILITY ANALYSIS OF THE DRUG DELIVERY SYSTEM ACROSS PORCINE DUODENAL TISSUE 6.1 Introduction ................................................................................................................ 84 6. 2 Material and Methods ............................................................................................... 87 6.2.1 Materials ............................................................................................................. 87 6.2.2 Tissue Isolation and Preparation ......................................................................... 87 6.2.3 Sample Preparation and Experiment Set-Up ....................................................... 88 6.2.4 Analysis of Permeants......................................................................................... 89 6.2.4.1 HPLC Quantification ..................................................................................... 89 6.2.4.2 Permeability Calculations.............................................................................. 89 6.2.5 Integrity Analysis of the Intestinal Mucosa ........................................................... 90 6.3 Results and Discussion ............................................................................................. 90 6.3.1 Permeation Characteristics Analysis ................................................................... 90 6.3.2 Integrity Analysis ................................................................................................. 94 6.4 Conclusion ................................................................................................................. 95 CHAPTER 7 CONCLUSION AND RECOMMENDATIONS 7.1 Conclusion ................................................................................................................. 96 7.2 Recommendations ..................................................................................................... 97 REFERENCES ................................................................................................................... 98 APPENDICES................................................................................................................... 126 Appendix 1: Abstract of Review Paper ........................................................................... 126 Appendix 2: Ethics Waiver Certificate ............................................................................ 127 Appendix 3: Permeability Regression Lines and Cumulative Amount Formula .............. 128 xii LIST OF FIGURES Figure 1.1: Molecular mechanisms that mediate PGE2-induced pain hypersensitivity inflammation. (A) COX-2 and mPGES-1 are upregulated in both injured cells and infiltrating immune cells in inflamed tissues, which increases PGE2 concentrations at peripheral nociceptor terminals. (B) Inflammation upregulates COX-2 and mPGES-1 expression in the spinal cord and increases PGE2 production. PGE2, mainly acting on EP2 receptors, contributes to central sensitization through both presynaptic and post-synaptic mechanisms.. ............................................................................................................................................. 2 Figure 1.2: Core-in-Cup PDDS.. ............................................................................................ 3 Figure 1.3: Schematic of the ideal desired release profiles.................................................... 5 Figure 2.1: Schematic diagram of the Digital Micromirror Device (DMD) stereolithography (DLP 3DP) setup including designs and images of 3D printed theophylline tablets. ............ 11 Figure 2.2: Comparison between mass-produced conventional dosage forms-based therapy and 3D printing-based personalized therapy. ...................................................................... 12 Figure 2.3: a. HME coupled FDM 3D printing procedure without in-line PAT tools b. HME coupled FDM 3D printing with PAT tools.............................................................................. 15 Figure 2.4: Selected images of 3D printed multi-drug system configurations.. ..................... 19 Figure 2.5: Picture of the self-assembly of HA and PQ-10 and the fibrous PECs collected by filtration before drying. SEM image of 3D printed FDC,alongside prolonged drug release profiles of a) Efavirenz (EFV), b) Tenofovir Disoproxil Fumarate (TDF) and c) Emtricitabine (FTC) in comparison to the commercial version, Atripla®. .................................................... 23 Figure 3.1: Chemical structures of a. PAR, b. ESM c. DS. ................................................... 33 Figure 3.2: Overlain spectra of the three actives obtained by UV-Vis Spectrophotometric analysis. .............................................................................................................................. 35 Figure 3.3: FTIR spectra of individual APIs (PAR, DS, ESM), key polymers (HPMC E50LV, HPMC K15M, MCC, EL 100-55), and potential API and polymer combinations as per fomulation layers (left to right). ............................................................................................ 40 Figure 3.4: DSC thermograms of individual APIs, key polymers, and potential combinations (above and below). ............................................................................................................. 42 Figure 3.5: TGA curves of individual APIs and key polymers. .............................................. 42 xiii Figure 3.6: Regression lines and respective representative chromatograms showing the retention of a.PAR at 1.916 min, b.DS at 6.716 min, and c.ESM at 5.940 min. .................... 44 Figure 3.7: Chromatogram showing the retention of PAR at 1.863 min, ESM at 5.807 min, and DS at 6.803 min in a combined sample containing 1 mg/ml of each drug (1:1:1). ......... 46 Figure 3.8: Specificity chromatograms of a. the Mobile Phase b. Excipients and c. APIs + Excipients in the ratio 25:5:1 (125:25:5 µg/ml).. .................................................................. 48 Figure 4.1: Summarisation of the QbD principles applied in the development of the present DDS. ................................................................................................................................... 51 Figure 4.2: Schematic representatives of preliminary drug delivery systems for segregated investigation of each layer. . ................................................................................................ 56 Figure 4.3: IR-Layer drug release profiles in SGF pH 1.2 (n=3; %RSD < 2.00 in all cases). 59 Figure 4.4: a. PAR b. DS DR Cup Layer drug release profiles in SGF pH 1.2 (0 – 2 hours), SSIF pH 6.8 (2 – 10 hours) (n=3; %RSD < 5.00 in all cases). ............................................. 61 Figure 4.5: a. Uncoated, showing early gastric release of ESM b. coated drug release profiles in SGF pH 1.2 (0 – 2 hours), SSIF pH 6.8 (2 hours onwards) (n=3; 5.00 < %RSD < 10.00 for a., %RSD < 5.00 for b.. ........................................................................................ 64 Figure 4.6: Digital visuals of formulation components during dissolution analysis, at pH 1.2 for after the first 2 hours. ..................................................................................................... 65 Figure 5.1: Polymeric controlled-release mechanisms. Schematic Illustration Of (A) Solvent- Activated and Stimuli-Responsive Systems. (i) The swelling-controlled system shows drug diffusion through the expandable hydrogel. (ii) The designed orifice in an osmotic-controlled system lets drug molecules release out with an adjustable release rate. (iii) External physical stimuli activate the responsive polymeric carriers to release their cargo in a controllable manner. (B) Chemically mediated CRSs. (i) Detachment of drug molecules from pendant side-chain polymeric systems (ii) Bulk erosion (iii) Surface versus mechanisms through erodible systems.. ............................................................................................................... 68 Figure 5.2: Presentation of a. Uncoated and Press-Coated ESM Core Pills; and overall manufacturing procedure. ................................................................................................... 69 Figure 5.3: Typical Force (N) versus Distance (mm) profile generated for the calculation of BHN values. ........................................................................................................................ 71 Figure 5.4: Digital images of a. the tablet b. Digital Calliper measurements of tablet dimensions, c. F1, showing the difference in thickness. ...................................................... 73 Figure 5.5: Mass gain and loss profiles of the tablets as a function of time. ........................ 75 xiv Figure 5.6: FTIR spectra of individual APIS and respective tablet formulation layers........... 76 Figure 5.7: TGA thermograms for APIs and respective tablet formulation layers. ................ 77 Figure 5.8: DSC thermograms for separate tablet formulation layers. ................................. 78 Figure 5.9: SEM Images of the core contents a. Surface view of the press coat, b. cross- sectional view of the ESM pill, c. Cross-sectional view 2 hours into dissolution in SGF, and d. 4 hours into dissolution in SSIF. In all cases, magnification was 200 X, except for b (500 X) at 5 kV. ................................................................................................................................ 79 Figure 5.9.1: a. b. SEM images of the IR layer at 200X and 2kX, respectively before dissolution, c. DR cup surface morphology before dissolution, d. 2 hours, and e. 4 hours into dissolution in SGF and SSIF, respectively, at 200X magnification at 5kV. ............................ 79 Figure 5.9.2: Cumulative drug release profiles in both SGF and SSIF (n=3) of a. F1 with respective overlain chromatograms showing the retention and detection of i. PAR only from samples withdrawn within the first 2 hours of dissolution and ii. ESM and DS from 2 hours onwards, b. F2 c. F3. Total Cumulative Amount Released, F1 PAR 101.09 % ± 0.80; DS 95,55 % ± 1.46; ESM 93.60 % ± 1.21. F2 101.81 % ±0.28; DS 9.71% ±1.39, ESM 82.43 % ± 3.16, and F3 PAR 101.53% ± 0.51; DS 95.46 % ± 0.70; ESM 95.07% ±3.89. .................... 82 Figure 6.1: a. Oblique view of Franz Cell, b. Oblique view of Side-by-Side Cell, and c. Everted gut sac apparatus with dimensions a. and complete setup b.. ................................ 86 Figure 6.2: Cross-sectional view of an in-line cell with HPLC fittings. .................................. 86 Figure 6.3: Digital Images of a. Specimen collection from euthanised white female pig and b. tissue preparation. .............................................................................................................. 88 Figure 6.4: Digital image of the Franz-Diffusion apparatus setup utilised for penetration analysis. .............................................................................................................................. 88 Figure 6.5: Cumulative permeation plots of a. PAR, b. DS, c. ESM in and out of formulation, d. Combined Tablet Plot (n=3). ............................................................................................ 93 Figure 6.6: FTIR spectra of porcine duodenal tissue, pre- and post-exposure ..................... 94 Figure 6.7: Graphical correlation plot of fraction absorbed versus apparent permeability coefficients of compounds observed in many higher throughput pharmacokinetic assays.. . 95 xv LIST OF TABLES Table 2.1: Summary of 3D Printed multi-drug oral dosage forms. ........................................ 18 Table 2.2: 3D Printed multi-drug implants. ........................................................................... 21 Table 3.1: Integral, Onset and Endset temperature values for the individual API, excipient, and combined samples. ...................................................................................................... 41 Table 3.2: Summary of Reverse Phase RP-HPLC conditions applied. ................................ 43 Table 3.3: Summary of regression statistics, LOD and LOQ values for the APIs. ................ 43 Table 3.4: Summary of precision results (n=3). ................................................................... 45 Table 3.5: Summary of accuracy results on standards (n=3) ............................................... 45 Table 3.6: Summary of accuracy results on tablet formulation dilution (n=3). ...................... 46 Table 3.7: Summary of robustness assessment (n=3). ........................................................ 47 Table 3.8: Stability assessment on standards (n=3 injections). ............................................ 47 Table 4.1: Immediate Release Layer formulation composition. ........................................... 53 Table 4.2: Delayed Release Cup layer formulation composition. ........................................ 54 Table 4.3: Core formulation composition. ............................................................................ 54 Table 4.4: Plug formulation. ................................................................................................. 54 Table 4.5: Core Press Coat composition. ............................................................................ 55 Table 4.6: Scale of flowability determined by different methods. .......................................... 56 Table 4.7: Constituents of dissolution media. ...................................................................... 57 Table 4.8: Flow properties per formulation granules. ........................................................... 58 Table 4.9: Best of fit values: IR Layer (n=3). ....................................................................... 59 Table 4.9.1: a. Best of fit values with Tlag: DR Layer (PAR). ............................................... 62 Table 4.9.1: b. Best of fit values with Tlag: DR Layer (DS). ................................................. 62 Table 4.9.2: Best of fit values with Tlag: ESM. ..................................................................... 64 Table 5.1: Risk estimation matrix presenting initial risk assessment levels of the drug delivery system formulation and manufacturing parameters. ............................................................ 67 Table 5.2: Resultant formulations. ....................................................................................... 69 xvi Table 5.3: Texture Analyser settings applied. ....................................................................... 71 Table 5.4: Summary of results from in-process validation tests on tablets ± SD. ................. 74 Table 5.5: Mean DSC integration data (n=3 samples). ........................................................ 77 Table 5.6: Best of fit values for the different kinetic models. ................................................ 82 Table 6.1a: Flux rates and permeability coefficients as determined through ex vivo permeation analysis. ........................................................................................................... 93 Table 6.1b: Flux rates and permeability coefficients as determined through ex vivo permeation analysis (in formulation). ................................................................................... 94 xvii LIST OF ABBREVIATIONS 3DP 3 Dimensional Printed 4IR 4th Industrial Revolution ABS Acrylonitrile Butadiene Styrene ACN Acetonitrile APIs Active Pharmaceutical Ingredients ATR-FTIR Attenuated Total Reflectance-Fourier Transform Infrared BCS Biopharmaceutics Classification System BHN Brinell Hardness Number BIJ Binder Ink Jetting BnOCPA Benzyloxy-cyclopentyladenosine CAGR Compound Annual Growth Rate CCS Croscarmellose Sodium CMAs Critical Material Attributes COX Cyclooxygenase CPOP Controlled Porosity Osmotic Pump CPPs Critical Process Parameters CQAs Critical Quality Attributes DDSs Drug-Delivery Systems DIW Direct Ink Writing DLP Digital Light Processing DMD Digital Micro-Mirror Device DPPO Diphenyl (2,4,6-Trimethylbenzoyl)-Phosphine Oxide DR Delayed Release DS Diclofenac Sodium DSC Differential Scanning Calorimetry EC Ethyl Cellulose EL Eudragit L EFV Efavirenz ESM Esomeprazole Magnesium Tri-Hydrate EVA Ethylene Vinyl Acetate EXT Extrusion (at room Temperature) xviii FDA Food and Drug Administration FDC Franz Diffusion Cell FDM Fused Deposition Modelling FTC Emtricitabine FTIR Fourier Transform Infrared Spectroscopy GIT Gastrointestinal Tract GPAs Gastroprotective Agents HME Hot Melt-Extrusion HPC Hydroxypropyl Cellulose HPMC Hydroxypropyl Methylcellulose ICH International Conference Harmonization IMP Impregnation INH Isoniazid IPQC In-Process Quality Control IR Immediate release LD Levodopa LOD Limit of Detection LOQ Limit of Quantification MCC Microcrystalline NaCMC Sodium carboxymethylcellulose NSAIDs Non-Steroidal Anti-Inflammatory Drugs ODT Orally Disintegrating Tablet PAM Pressure Assisted Microsyringe PAR Paracetamol PAT Process Analytical Technology PBPK Physiologic-Based Pharmacokinetic PBS Phosphate Buffered Saline PCL Poly Ɛ-Caprolactone PDDS Pulsatile Drug Delivery Systems PDLA Poly D-Lactic Acid PDLLA Poly-D,L-Lactic Acid PEC Polyelectrolyte Complexes PEEK Polyether Ether Ketone PEKK Polyether Ketone xix PEO Polyethylene Oxide PGE2 Prostaglandin (E2) PLA Polylactic Acid PLLA Poly-L-Lactic Acid PoC Point-Of-Care PPIs Proton Pump Inhibitor PVA Polyvinyl Alcohol PVP Polyvinylpyrrolidone QbD Quality by Design QbT Quality by Testing QTPP Quality Target Product Profile RP-HPLC Reverse Phase-High Performance Liquid Chromatography RSD Relative Standard Deviation SEM Scanning Electron Microscopy SFF Solid Freeform Technology SGF Simulated Gastric Fluid SLA Stereolithography SLM Selective Laser Melting SLS Selective Laser Sintering SLS Sodium Lauryl Sulphate SMEDDS Self-Micro Emulsifying Drug Delivery Systems SNEDDS Self-Nanoemulsifying Drug Delivery Systems SR Sustained Release SSG Sodium Starch Glycolate SSIF Simulated Small Intestinal Fluid TDF Tenofovir Disoproxil Fumarate TGA Thermogravimetric Analysis TLC Thin Layer Chromatography UPLC-MS Ultra-Performance Liquid Chromatography-Mass Spectrometry UV Ultra-Violet Visible WHO World Health Organisation 1 CHAPTER 1 INTRODUCTION AND MOTIVATION FOR THE STUDY 1.1 Background An ongoing conundrum for healthcare professionals has been how to effectively manage chronic pain without posing risks of tolerance, dependence, and addiction, with nearly 1 in every 5 South Africans suffering from chronic pain, limbic joint pain, and back pain making up 43.6%, and 30.5% of the pain, respectively (Kamerman et al., 2020). The range of available pharmacological treatment options for long-term management is predominantly restricted to non-steroidal anti-inflammatory drugs (NSAIDs) and/or opioid analgesics. Frequently, patients choose opioids as their preferred choice due to their well-established effectiveness in pain relief and occasional mood-altering benefits (Engel-Rebitzer et al., 2021; Dowel et al., 2022; Seidel et al., 2022; Castle et al., 2023). Of late, rising rates of opioid overdose deaths have raised questions about prescribing opioids for chronic pain management. Worldwide, about 0.5 million deaths are attributable to drug use of which more than 70% of these deaths are related to opioids, with more than 30% of those deaths being a result of overdose. Because of the risk of serious harm, without sufficient evidence for benefits, current guidelines discourage opioid prescribing for chronic pain (Dowel et al., 2022). “Opioid misuse and abuse remain a serious public health crisis facing the country. Preventing new addiction through fostering the development of novel non-opioid analgesics is an important priority for the FDA,” Patrizia Cavazzoni, M.D., director of the FDA’s Centre for Drug Evaluation and Research (FDA, 2022). The pathophysiology of persistent inflammatory pain commonly found in joint diseases includes signalling initiated by peripheral terminals of sensory neurons (Chen, Yang and Grosser, 2013; Seidel et al., 2022). The presence of algogens such as prostaglandins, particularly E2 (PGE2), in the synovium and synovial fluid capable of exciting or sensitizing peripheral nociceptors, describes the generation of pain in inflammatory joint diseases. PGE2 is a product of the hydrolysis of arachidonic acid, a reaction catalysed by cyclooxygenase enzyme 2 (COX-2) (Di Carlo, Smerilli and Salaffi, 2021), Figure 1.1. The pathophysiology explains the rationale behind the use of COX inhibitors in associated conditions. On the other hand, inhibition in the synthesis of prostaglandins in patients on chronic Non-Steroidal Anti- Inflammatory Drugs (NSAIDs) induces clinically significant ulceration, bleeding, and/or obstruction. The pathogenesis of NSAID-induced ulceration and bleeding involves three key pathways: inhibition of cyclooxygenase (COX)-1 activity, inhibition of COX-2 activity, and direct cytotoxic effects on the epithelium (Wallace, 2008). Selective COX-2 inhibitors were at some 2 point hailed for their non-ulcer-inducing mechanism due to their COX-1 sparing pathway until the role of the COX-2 pathway was identified in the pathogenesis of NSAID-induced damage, including evidence of increased risks of myocardial infarction to the point of withdrawal from general use. Therefore, regardless of the NSAID of choice, concomitant proton pump inhibitor (PPIs) co-therapy is crucial. There exists comprehensive evidence on the effectiveness of PPIs over standard doses of H2 receptor antagonists and that they are as effective as, but better tolerated than misoprostol in ulcer prophylaxis (Rancis et al., 2002; Dieppe, Shah Ebrahim and Jüni, 2004; Wallace, 2008; Lazzaroni and Porro, 2009). Figure 1.1: Molecular mechanisms that mediate PGE2-induced pain hypersensitivity inflammation. (A) COX-2 and mPGES-1 are upregulated in both injured cells and infiltrating immune cells in inflamed tissues, which increases PGE2 concentrations at peripheral nociceptor terminals. (B) Inflammation upregulates COX-2 and mPGES-1 expression in the spinal cord and increases PGE2 production. PGE2, mainly acting on EP2 receptors, contributes to central sensitization through both presynaptic and post-synaptic mechanisms. Modified and reprinted with permission from (Chen, Yang and Grosser, 2013). The tablet remains the go-to dosage form in most pharmaceutical developments due to its plethora of advantages, particularly in chronic disease management (Hummler et al., 2023). Controlled pulsatile-release multi-drug solid oral dosage forms have been instrumental in treating various chronic ailments improving adherence and compliance patterns in most patients (Huynh and Lee, 2015). Pulsatile drug delivery systems (PDDS), i.e., systems in which the drug is released suddenly after a well-defined lag time according to the circadian rhythm of the disease, are classified as either, time-controlled pulsatile release (single or multiple unit system), internal stimuli-induced or external stimuli-induced release systems 3 (Sokar et al., 2013). The dosage forms of these can either follow capsule, pellet, or tablet configurations amongst which is the ’core-in-cup’ tablet system. A typical core-in-cup tablet system consists of three different parts: a core tablet, containing the active ingredient, an impermeable outer shell, and a top cover plug layer of a soluble polymer, (Danckwerts and Watt, 1998; Nagaraju et al., 2009; Sokar et al., 2013; Srinija and Lakshmi, 2016; ElMeshad et al., 2020) (Figure 1.2). Nonetheless, the present study proposes to employ the same concept in developing a drug delivery system comprising an enterically press-coated esomeprazole magnesium trihydrate (ESM) core pill, an insoluble pH-dependant plug barrier, a delayed release (DR) cup formulation layer of diclofenac sodium (DS) and paracetamol (PAR), and an Immediate release (IR) top layer of PAR. In addition to the discovery and development of non-opioid analgesics such as the novel VX-548 (moving into Phase III clinical trials) and benzyloxy- cyclopentyladenosine (BnOCPA); an expedited approach towards formulation-based interventions involves research on combinations of currently available active pharmaceutical ingredients (APIs) to tap into unexplored synergistic potentials of non-addicting analgesics (Kingwell, 2022; Wall et al., 2022). Figure 1.2: Core-in-Cup PDDS. Reprinted with permission from (Sokar et al., 2013). 1.2 Rationale Behind the Design and Combination High-income countries have suffered strains in their health and financial systems as a result of the global crisis, unfortunately, the health systems of developing countries are even less prepared should the opioid epidemic occur (Harker et al., 2020). In order to lessen the forecasted burdens, there is a need to ensure a reduction in the number of new cases of addiction, whether from intentional or unintentional abuse. A pharmaceutical approach in ensuring this involves encouraging the discovery, development, and re-development of low- abuse potential, non-addicting analgesic formulations for prescription to opioid-naïve chronic patients. 4 Adherence to adjunct gastroprotective agents (GPAs) in NSAID users has been established to be suboptimal; in fact, it was discovered in one study that more than one-third of NSAID users did not use the prescribed GPAs daily, which resulted in a 16% increase in GIT complications for every 10% decrease in adherence (Van Soest et al., 2007). Therefore, this study proposes a familiar analgesic combination of PAR and DS, comprising prophylactic ESM for potential and convenient use in the long-term symptomatic relief of inflammatory pain in chronically ill patients. The rationale behind the design was based on inherent drug properties, target release sites, desired therapeutic effects, and allowance for drug release manipulation. Whilst diclofenac potassium is formulated for release in the stomach, often as IR tablets, DS due to its resistance to dissolution in low pH gastric milieu, is designed for duodenal release (Williams and Buvanendran, 2011). Following oral administration, DS uniquely accumulates in synovial fluid which may explain why its duration of therapeutic effect is considerably longer than the plasma half-life of 1 to 2 hr, the dose range for adults with joint inflammatory pain is 150 – 200 mg/24hrs in adults (Williams and Buvanendran, 2011). Various pieces of research have comprehensively proved the synergism between PAR and DS, including new data on the analgesic mechanisms of PAR, confirming its efficacy in multimodal, non-opioid, or opioid- sparing, therapies for the treatment of both acute and chronic pain (Przybyła, Szychowski and Gmiński, 2021; Freo, 2022). The top layer constitutes an immediate-release formulation of PAR, 250 mg for an early onset of analgesia. The cup layer was formulated for the delayed and retarded release of 100 mg of DS and 250 mg of PAR in tandem. Lastly, the core is a press-coated 20 mg ESM pill. Because ESM rapidly degrades at low pH, i.e., gastric milieu, a low solubility barrier plug was placed between the IR layer and DR layer. A schematic depicting the desired release profile is represented in Figure 1.3. https://www.sciencedirect.com/topics/medicine-and-dentistry/synovial-fluid https://www.sciencedirect.com/topics/medicine-and-dentistry/synovial-fluid 5 Figure 1.3: Schematic of the ideal desired release profiles. 1.3 Aims and Objectives of the Study. The aim of this study was to design and develop a non-opioid tripartite controlled-release tablet for chronic inflammatory pain. The above aim was attained via the following objectives: 1. Performing pre-formulation studies to characterise and screen various polymer combinations for potential use in the formulation, including developing and validating an HPLC method for the simultaneous detection of the APIs. 2. Designing and formulating a tripartite controlled-release tablet and performing process validation tests by analysing the matrices' hardness, uniformity of mass, friability, and thickness. 3. Characterisation of the tablet by performing physicochemical studies to determine stability and molecular interactions within the matrix and between the layers. 4. Elucidation of the physico-mechanical properties of the DDS through matrix hydration, erosion, and textural analyses. 5. Performing in vitro drug release studies to determine the drug release profile of the DDS and predict the potential release mechanism of the system. 6. Performing ex vivo permeability studies on porcine duodenal tissue to determine the effect of tableting on the permeability profiles of the active ingredients. 6 1.4 Overview of This Dissertation Chapter One: This chapter provides an introduction and background of this current study. Details on the motivation for undertaking this study, the rationale, aims and objectives for the study were presented. Chapter Two: This chapter presents a comprehensive literature review on controlled-release multi-drug oral systems, with concise insight into the currently employed and novel formulation techniques. Included therein is a summary of different 3D printing technologies applied in the fabrication of customised solid oral drug delivery systems, such as extrusion-based printing further subdivided into fused deposition modelling (FDM) and extrusion at room temperature (EXT), amongst others. The chapter ends with an expert opinion on how identified hindrances in the full implementation of these novel techniques may be tackled. Chapter Three: This chapter outlines all the pre-formulation investigations undertaken on potential formulation components. Details on the effects of potential polymers/excipients on APIs’ characteristics as determined via DSC, TGA and FTIR analyses are presented. Outlined as a part of the chapter are steps taken towards the development and validation of an HPLC method for the simultaneous quantitation of the APIs during dissolution and permeability analysis. Chapter Four: This chapter entails the preliminary design and developmental stage, in which working formulations were determined through in vitro dissolution studies carried out on various drug-loaded structural components of the tablet matrix. In order to distinctly establish working concentrations of the functional polymers for each formulation layer, partial blanks were formulated and compressed for investigation. Analyses performed highlight the influence of key polymers such as super-disintegrant croscarmellose sodium (CCS), hydroxypropyl methylcellulose (HPMC K15M), eudragit L 100-55 (EL 100-55) as well as the efficiency of the plug as an added protection from gastric degradation of the core formulation. Chapter Five: This chapter focuses on the assemblage of the complete and optimal drug delivery system, based on the findings from the previous chapter, including in vitro release studies and all in-process validation tests. Included therein is the characterisation of the final formulation, i.e., establishment of the DDS’s physico-mechanical properties through matrix hydration, erosion, and textural analyses. Chapter Six: This chapter expounds on the methodology applied in evaluating the ex vivo permeability characteristics of the drug compounds in combination through porcine duodenal tissue. Highlighted in the chapter is the effect of tableting excipients/polymers on available methods of permeability analysis. 7 Chapter Seven: Concludes on the present research, presenting recommendations for future work. 8 CHAPTER 2 LITERATURE REVIEW ON NOVEL FORMULATION TECHNIQUES FOR CONTROLLED RELEASE MULTI-DRUG SYSTEMS 2.1 Introduction Because of the prevalence and chronicity of both communicable and non-communicable diseases such as hypertension, tuberculosis, and diabetes, patients are often prescribed long- term regimens comprising multiple medications (Gradman et al., 2013; Weverling et al., 1998; Cruz et al., 2018). While there is no set definition of polypharmacy, it is commonly described as the concurrent prescribing of at least four or five medicines, for instance, a typical regimen for a hypertensive patient with hypercholesterolemia contains Enalapril 10 mg, Carvedilol 10 mg, Spironolactone 10 mg, Hydrochlorothiazide 25 mg at least once daily, and Simvastatin 20mg (STG, 2015). Following administration requirements to the letter is a sine qua non for effective polypharmacy. Unfortunately, both intentional and non-intentional non-adherence are frequent consequences of polypharmacy (Payne and Avery, 2011). Poor medication adherence has been linked to a deterioration in the prognosis of patients with non-communicable diseases, coupled with other negative effects such as increased mortality and costs (Diseases et al., 2021). Recent findings from a study examining the factors associated with non-adherence among older patients with multimorbidity and polypharmacy showed a 79.6% prevalence of non-adherence in 74 non-institutionalized patients aged ≥65 9 years with ≥2 chronic conditions (González-Bueno et al., 2021). The former statistics cohere with Butler and co-workers’ previous findings, in which non-adherence was associated with 4 to 11% of all hospitalizations and 7.6% of emergency visits, with a staggering overall incidence of 40 to 86% (Butler et al., 2011). A formulation-based solution to the pressing issues of polypharmacy was the introduction of fixed dose combinations (FDCs) commonly referred to as "polypills” targeted at reducing the risk of mortality and cardiovascular events (Abushouk et al., 2022). In 2016, polypills were formally recommended and included in the European Guidelines on cardiovascular disease prevention and the World Health Organization proposed polypills as a significant strategy for improving cardiovascular disease management (Piepoli et al., 2016; WHO, 2016). As the name suggests these formulations contain multiple active ingredients, each with a different pharmacological effect. A commercially available example is PolycapTM constituting enteric-coated aspirin, ramipril, simvastatin, atenolol, and hydrochlorothiazide, other examples include RamitorvaTM (aspirin, ramipril, and atorvastatin), and StarpillTM (aspirin, losartan, atenolol, and atorvastatin) (Yusuf et al., 2009; Webster, Castellano and Onuma, 2017). The development of the majority of polypills was motivated by the serious burden and incidence of cardiovascular disorders, however, the concerns around polypharmacy are not limited to cardiovascular diseases. Furthermore, the rationale behind multi-drug systems hinges on the effectiveness of combined therapy, simplified treatment regimens, reduced production and storage costs, and efficient supply chain and logistics, in addition to the highlighted grounds of enhanced adherence and compliance (Webster, Castellano and Onuma, 2017). However, polypills like any other intervention have their drawbacks, notably the production process, which has proven to be quite complex. Different procedures are utilized in the classic compression process, such as granulation of all active components, individual granulation of actives, and drug separation by layering in circumstances where there are incompatibles and interactions between the actives (Webster, Castellano and Onuma, 2017). More importantly, the mass production of the polypill has been criticised for its one-size-fits- all approach, which disregards individual patient needs that is, some patients may require higher or lower dosages of particular active moieties within the formulation, while others may not require all of the drugs contained within the formulation (Abushouk et al., 2022). Moreover, bioavailability profiles and dosing requirements of paediatric, adult, and geriatric populations vary. Although traditional techniques may be used to customise a pill for a patient, the methods are neither cost-effective nor practicable for continuous fast on-demand production (Webster, Castellano and Onuma, 2017). This calls for more innovative and flexible production techniques that will allow the smooth fabrication of bespoke drug combinations. One such 10 technology is 3-dimensional (3D) Printing, an innovation posing significant promise for patient patient-centred therapy (Jamróz et al., 2018). The combination of these two ideas (3D printing and multi-drug Systems) represents another step forward in the revolutionisation of healthcare. This review will investigate the promising paradigm shift from one medication for all to personalised patient-centred medicines, providing an overview of the current state of 3D printing techniques for multi-active dosage forms, polymers, and excipients used and their effects, the market view, limitations, and potential barriers to full implementation. 2.2 Summary on 3D Printing Technologies 3D printing has been described by terms such as layered manufacturing, additive manufacturing, computer automated manufacturing, fast prototyping, solid freeform technology (SFF), and even artistic science from its inception in the early 1970s (Abdulhameed et al., 2019; El-bassyouni, 2020). It has a plethora of applications in engineering, dentistry, building, aeronautics, bone regeneration, as well as pharmacy (Choonara et al., 2016). To produce a solid entity, the printing process involves layer-by-layer deposition of ‘ink' material based on a pre-designed 3D digital model. Although it has been around for some time, its use in the pharmaceutical sector is still in its nascent stages, and its full potential has yet to be realized (Jamróz et al., 2018; Zhu et al., 2020). A lot of research has been done on 3D printing in the medical sector, with a vast amount of literature for reference on topics such as the selection of appropriate excipients, printable actives, strategies to achieve different drug-release profiles, and the advantages of various methodologies (Goyanes et al., 2015; Awad et al., 2018; Horst, 2018). Operations of the different types of 3D printing technologies have been extensively explained in several publications with provisions for the different ways of classifying them (Goyanes et al., 2015; Horst, 2018; Abdulhameed et al., 2019; Zhu et al., 2020). A simple classification system by Jamroz et al. included powder solidification, liquid solidification, and extrusion. Extrusion-based printing is frequently utilized in the production of solid oral dosage forms. The approach is further subdivided into fused deposition modelling (FDM) and extrusion at room temperature (EXT), the latter of which is confined to thermolabile drugs (Jamróz et al., 2018). In liquid solidification, the object is built by either droplet solidification (Drop on Drop deposition or Ink Jetting) or solidification of photosensitive liquid (Stereolithography/SLA), whereas in powder solidification, the object is built by either solidification of powdered material by high energy beam (Selective laser sintering/ SLS) or liquid binding of powdered material (Drop on solid deposition or Binder Ink Jetting (BIJ) (Jamróz et al., 2018). Modifications were made to SLA to allow for faster, better resolution, and more efficient processing via the incorporation of a digital micro-mirror device (DMD) in the optical path of the laser. Whilst SLA printers, 11 require vast amounts of resin for printing, digital light processing (DLP) printers allow for customized resin reservoirs and lesser amounts of resin. Promising applications of DLP 3DP include printing medical devices, microneedle-mediated drug delivery systems (Lim, Ng and Kang, 2017; Bloomquist et al., 2018), and more recently solid oral dosage forms, illustrated in Figure 2.1 below (Kadry et al., 2019). Figure 2.1: Schematic diagram of the Digital Micromirror Device (DMD) stereolithography (DLP 3DP) setup including designs and images of 3D printed theophylline tablets. Reproduced with permission from (Kadry et al., 2019) © 2019 Elsevier B.V. Ltd. The advantages of 3D printing in medication formulation far outweigh the negatives. It enables the compartmentalization of drug components, which is especially essential in multi-drug systems where interactions and incompatibilities between medicines and excipients are possible. Furthermore, unlike traditional techniques, 3D printing is more versatile, allowing for adjustments in shape and size to meet the specific demands of a patient (Alhnan et al., 2016). From bi-layered tablets to 6-layer tablets, researchers have proved the versatility of 3D printing (Gioumouxouzis et al., 2018; Robles-martinez et al., 2019). Although 3D printing has many advantages, some of the few minor drawbacks include the final product's look with perceptible exterior flaws, and post-processing of the product, particularly in powder-based and extrusion- based 3D printing (Alhnan et al., 2016). 2.3 Customisation in Different Population Groups 2.3.1 Visually Impaired Patients For visually impaired patients, receiving medications may present challenges like trouble reading labels and differentiating drugs, especially if the once annotated packaging has been damaged or lost (Andreadis et al., 2022). 3D printing opens up the possibility of printing Braille characters directly onto the pills. Successful attempts were made in printing orally disintegrating tablets and Intraoral films with embedded Braille and Moon characters. Visually 12 impaired volunteers who took part in the haptic in vivo evaluation studies confirmed the readability of the embedded text and the potential for personalized treatment with 3D printing (Eleftheriadis and Fatouros, 2021). 2.3.2 Paediatric Patients The pharmacotherapy of children requires very specific guidelines in regard to dosing and special attention to safety and efficacy. A child's sensitivity to bitter ingredients makes easy administration and taste key factors to consider in dosage form design for this group (Andreadis et al., 2022). Personalized paediatric formulations can be prepared using 3D printing, however, ingredients used in 3D printed dosage forms intended for administration to children need to be extensively studied for their possible toxicity. Researchers have attempted nonetheless to develop delivery systems that appeal to the population, such as chewable gelatine-based gummies in the form of Lego™ bricks, including capsules and orodispersible miniprintlets, and cartoon-based ibuprofen and paracetamol-loaded chocolate chewable tablets (Rycerz et al., 2019; Karavasili et al., 2020; Eduardo, Ana and José, 2021). 2.3.3 Geriatric Patients Multimorbidity is a more common occurrence in older patients making them the most susceptible to the deleterious effects of polypharmacy. It is this population group that will benefit the most from 3D printed multidrug systems, customised to each one’s therapeutics needs (Andreadis et al., 2022). Figure 2.2 illustrates the concept of customised therapy achieved by 3D printing versus mass-produced conventional systems. Figure 2.2: Comparison between mass-produced conventional dosage forms-based therapy and 3D printing-based personalized therapy. Adapted with permission from (El-Bassyouni, 2020) © 2020 Elsevier. 13 2.4 3D Printed Multi-Drug Dosage Forms Spitram® (levetiracetam), a single-active formulation for the treatment of seizures, is the only 3D printed formulation with Food and Drug Administration (FDA) clearance to date (Kassem, Sarkar and Nguyen, 2022). The development of 3D printed multi-system models is presently based on proof-of-concept approaches. Oral dosage forms such as captopril, nifedipine, and glipizide, fixed-dose combinations efavirenz (EFV), tenofovir disoproxil fumarate (TDF), and emtricitabine), and a 6-layered pill containing paracetamol, caffeine, naproxen, chloramphenicol, prednisolone, and aspirin make up the majority of these formulations. These formulations were printed using a variety of 3D printing methods (Khaled et al., 2015b; Robles- martinez et al., 2019; Siyawamwaya et al., 2019a). Multidrug systems are not only limited to oral dosage forms, but the technology has also found application in drug-eluting implants (Domsta and Seidlitz, 2021). Among the numerous approaches available, nozzle-based printing, particularly fused deposition modelling, has been the most popular among researchers (Kassem, Sarkar and Nguyen, 2022). 2.4.1 3D Printed Oral Multi-Drug Formulations 2.4.1.1 Formulations Printed by Fused Deposition Modelling One noteworthy advantage of FDM in multi-drug formulation development is that it allows for multi-material printing, which allows for the extrusion of more than one drug-loaded filament in a single printing process (Long et al., 2017). Hot melt-extrusion (HME) is a stand-alone process used to manufacture API and polymer filaments (feedstock) prior to FDM printing. The method uses heat and pressure to melt and extrude the mixture via an orifice (Repka et al., 2018; Lubrizol, 2019). The extruded mixture is aligned with a 3D printer in HME-based printing to create the item based on the pre-designed digital model (Zhang et al., 2017). HME has proved useful in the creation of solid dispersions, alleviating problems associated with poorly soluble APIs and therefore improving dissolution rates and bioavailability (Repka et al., 2018). The drug loading method in filament production determines the amount of drug that can be loaded which in turn influences the rate of drug release from the eventual 3D printed solid dosage form. Drug loading by impregnation is limited to a low of about 2% w/w and yields rapid drug release, of which both scenarios may not be ideal for multi-drug systems requiring sustained release mechanisms. By employing HME in drug-filament production, both higher drug loading and prolonged release profiles can be achieved (Karavasili et al., 2020). This was demonstrated by Thanawuth et al., (2021), in a study where drug release studies were carried out on three, 3D printed tablet formulations, two low-dose formulations, one made from impregnation (IMP)-produced drug-loaded filaments and the other from HME-produced drug- loaded filaments, the third was also HME-produced but with a higher loading (30 % w/w), PLL 14 (PVA) was the choice of polymer in all formulations. The results confirmed that drug loading determined by the technique directly influences drug release, both the IMP and HME-produced filaments with low drug loading yielded faster release profiles compared to the high drug loaded-HME-produced-tablets, in which release was extended to 24 hours with a swelling- erosion mechanism (Thanawuth et al., 2021). With growing interest in additive manufacturing of personalised medication with complex geometries, integrating and synchronizing FMD and HME using process analytical technology (PAT) tools could potentially lead to continuous, larger-scale production of efficient and cost- effective multi-drug delivery (Long et al., 2017; Wesholowski et al., 2019). Raman Spectrometer, Laser-Based Diameter Measuring Module, Acoustic Emission Sensor, High- speed camera, and Rheometer are some possible PAT instruments that may be included into the HME-FDM process and are favourable to multi-drug systems. APIs can be quantified, deterioration identified in-line, dosing accuracy increased, viscosity measurement avoiding nozzle clogging, no pauses in the printing process, and texture analysis guaranteeing smooth feeding of filaments into 3D printers (Hagrasy et al., 2013; Korte and Quodbach, 2018; Yang et al., 2018; Andrews et al., 2019; Ponsar, Wiedey and Quodbach, 2020). Figure 2.3 and 2.3 b illustrate printing processes with and without the inclusion of PAT. Because various medicines have varying release kinetics and absorption locations, it is critical to consider aspects that will affect transit durations through particular areas of the gastrointestinal tract when developing controlled-release multiple-drug delivery systems (Aulton and Taylor, 2013). For example, metformin and glimepiride, two medicines used to treat Type 2 diabetes, (administered as a fixed dose combination or as separate medications) have different absorption sites, metformin is mostly absorbed in the small intestine, whereas Glimepiride is absorbed in the stomach (Hwang et al., 2013; STG, 2015). Based on these criteria, to ensure optimum pharmacotherapy, researchers employed HME/FDM 3D printing to fabricate a bi-layered, controlled-release solid dosage form of metformin and glimepiride (Gioumouxouzis et al., 2018). Similarly, Windolf and co-workers recently fabricated mini floating polypills for Parkinson’s Disease combining Levodopa, benserazide, and pramipexole in various dosing for personalized therapy (Windolf et al., 2022). A new Melt Extrusion Deposition (MED™) 3D printing technology has been created to improve the use of FDM by addressing drawbacks such as the requirement for pre-fabrication of printable drug-loaded filament and printing precision. The MED™ method combines powdered active components and excipient ingredients as starting materials, eliminating the need for 15 prior filament production. Reproducible, accurate, and larger-scale production can be performed using high-throughput printer nozzle arrays, handling numerous materials, and precise deposition control. This new technology, combined with compartmental model designs, allows for the fabrication of bespoke multi-drug systems with modulated release kinetics from each compartment (Zheng et al., 2021). Figure 2.3: a. HME coupled FDM 3D printing procedure without in-line PAT tools b. HME coupled FDM 3D printing with PAT tools. 16 2.4.1.2 Formulations Printed by Pressure Assisted Microsyringe Extrusion Thermal deterioration is an unavoidable drawback of HME-FMD printing, limiting the number of medicines and materials that may be produced using the technology, also known as Pressure Assisted Microsyringe (PAM), extrusion at room temperature does not require high temperatures. Rather the process involves the preparation of a semisolid gel or paste consisting of drug/s and additives. The mass is then continuously extruded layer by layer through a syringe-based tool-head. Thus, by employing multi-syringe systems, this technique can yield multi-active solid dosage forms with varying kinetics, Figure 2.4 a (Jamróz et al., 2018; Azad et al., 2020). Khaled and colleagues (2015) used room-temperature extrusion to create a multi-compartment, multi-drug system consisting of nifedipine, captopril, and glipizide. The formulation was designed to solve issues found in the treatment of hypertension and type 2 diabetes, such as dosage dumping, burst phenomena, short elimination half-life, and short duration of action (Khaled et al., 2015b). Captopril was released through controlled osmosis by incorporating an osmogen sodium chloride into the core of the captopril compartment, whereas the other two actives were released via diffusion processes, as shown in Figure 2.4b. Controlled porosity osmotic pump tablets use osmotic pressure principles for controlled release, overcoming the impact of the gastrointestinal tract's physiological condition (Shahi et al., 2012; Sahoo et al., 2015). This approach is not only simple to use, but it is also appealing to patients since it decreases dose frequency while providing the benefit of a longer duration of action. Zero-order release, independent of physiological parameters such as pH can be achieved. The manufacture of a controlled porosity osmotic pump (CPOP) involves the encapsulation of an osmogen core with a semipermeable membrane, which rapidly dissolves when in contact with an aqueous medium creating a micro-porous membrane (Sahoo et al., 2015). Another recent study used extrusion-based printing at room temperature to obtain a higher bioavailability of efavirenz (EFV), tenofovir disoproxil fumarate (TDF), and emtricitabine (FTC) compared to a non-3D printed commercial version of the same medicines, Atripla® for improved HIV therapy (Siyawamwaya et al., 2019b). Nonetheless, extrusion-based printed DDS have unappealing characteristics such as low resolution (depending on nozzle size) and poor mechanical qualities, necessitating the use of more precise methods such as SLA (Domsta and Seidlitz, 2021). 17 2.4.1.3 Formulations Printed by Stereolithography Stereolithography (SLA) is the original 3D printing method, having been used in pharmaceutical technology for the first time in 1984 (Jamróz et al., 2018). It is a laser-based technology that uses photo-polymerisation to create a solid three-dimensional object from a photocurable viscous liquid enclosed in a vat. A layer is generated by a laser beam following a g-coded pattern, then the platform is lowered into the vat for the second layer, and so on until the item is finished (Melchels, Feijen and Grijpma, 2010; Abaci et al., 2021). Its advantages over nozzle-based methods include greater dimensional precision, a finer finish, and a lack of dependency on heat for manufacturing. More significantly, unlike SLA, FDM is restricted to a small number of spatially separated medicines in a formulation (Pereira et al., 2019; Robles-martinez et al., 2019). As a result, Martinez and colleagues used SLA 3D printing for the first time to create a multi-layered tablet containing six drugs: paracetamol, caffeine, naproxen, chloramphenicol, prednisolone, and aspirin (Robles-martinez et al., 2019). However, considerations such as the possibility of API deterioration owing to laser projection onto the drug-loaded solution and the requirement for extra API in the manufacture of the photocurable resin, restrain its complete usage in production (Abaci et al., 2021). Recently, SLA was employed in the development of multifunctional nanocomposite pills, in which both nanoparticles and conventional solid dosage form designs make up a drug delivery system. The procedure entails the creation of vat photo-polymerised monoliths that serve as reservoirs for drug-loaded nano-carriers. Similarly, researchers may be able to develop more novel nanocomposites for multi-modal and multi-compartment drug delivery systems suitable for multiple drug systems using SLA-assisted 3D printing (Sharma et al., 2022). 2.4.1.4 Formulations Printed by Selective Laser Sintering Selective laser sintering (SLS) is yet another type of laser-based printing that has had limited application in 3D printed oral multi-drug systems. This is most likely due to the laser beam’s high-energy input, which raises concerns about the likelihood of API and pharmaceutical excipient deterioration (Alhnan et al., 2016). Unlike similar powder bed-based techniques, the process is faster, and does not require a binder solution; instead, the powdered printing material is exposed to radiation, which will either partially or fully melt it for layered fabrication. In SLS-printed oral dosage forms, two drug loading approaches can be used: prior to sintering and after sintering. The former entails preparing an API and excipient/s powder mixture, which is then added to the powder bed for laser-based fabrication of the drug-loaded tablets (Abaci et al., 2021). Drug loading after printing is considered a safer approach because the API is not exposed to laser radiation. In this approach, a non-drug loaded excipient-based delivery system is printed and the API is loaded using coating procedures or infilling techniques. SLS 18 was used for the dual printing of miniprintlets with two rate-controlling systems incorporating two model drugs, paracetamol and ibuprofen, whereby one drug was released immediately from a kollicoat instant release matrix, whilst the release of the second drug was sustained over an extended period using ethyl cellulose (Awad et al., 2019). Great potential exists in the use of SLS in oral dosage forms, particularly multi-drug permutations with distinct and uniform dosing, permitting discrete separation of the APIs and resolving problems of incompatibility and physical interaction, which are major drawbacks in most multi-drug formulations. Table 2.1 provides examples for each of the afore-explained techniques. Table 2.1: Summary of 3D Printed multi-drug oral dosage forms. Drug Combination 3DP Technique applied Shape and design. Reference Nifedipine, Captopril and Glipizide. Extrusion at room temperature. Cylinder. Compartmentalised: Top compartment, Nifedipine and Glipizide, release by diffusion. The bottom compartment, Captopril, release by osmosis. (Khaled et al., 2015b) Efavirenz, Tenofovir Disoproxil fumarate and Emtricitabine. Extrusion at room temperature. Prism Separate layers of Efavirenz, Tenofovir and Emtricitabine. (Siyawamwaya et al., 2019b) Paracetamol, Caffeine, Naproxen, Chloramphenicol, Prednisolone and Aspirin. SLA Cylinder and ring shape 6 distinct layers of each drug, with different release profiles. (Robles- martinez et al., 2019) Lisinopril dihydrate, Indapamide, Rosuvastatin calcium and Amlodipine besylate. Fused deposition modelling (FDM) Cylindrical. Layered design, each layer constituting a different drug. (Pereira et al., 2019) Aspirin, Hydrochlorothiazide Pravastatin, Atenolol, and Ramipril. Extrusion- based printing. Cylindrical. Aspirin and Hydrochlorothiazide immediate release compartment and Atenolol, Pravastatin, and Ramipril sustained release compartments. (Khaled et al., 2015a) Metformin and Glimepiride HME-based Fused Deposition Modelling. Bilayer oral solid dosage form combining metformin for prolonged and glimepiride for immediate drug delivery. (Gioumouxouzis et al., 2018) 19 Figure 2.4: Selected images of 3D printed multi-drug system configurations. Reproduced with permission from a) (Khaled et al., 2015c) 2015 Elsevier; b) (Khaled et al., 2015a) © 2015 Elsevier. Levodopa (LD) Benserazide (BZ) and Pramipexole (PDM). HME-based Fused Deposition Modelling. Cylindrical. PDM and polyvinyl alcohol for rapid drug release and a fixed combination of LD/BZ (4:1) in an ethylene-vinyl acetate copolymer matrix for prolonged drug release. (Windolf et al., 2022) Isoniazid and Rifampicin HME-based Fused Deposition Modelling Compartmentalised shells. Each compartment physically sealed for effective retardation of in vitro API release (Genina et al., 2017) Paracetamol and Ibuprofen. SLS Spherical Miniprintlets. Two distinct regions; Immediate release and Sustained release. (Awad et al., 2019) Irbesartan, atenolol, hydrochlorothiazide and amlodipine SLA Cylindrical, 2 types of layered designs, Type 1 Irbesartan and Atenolol on the outer layers and, hydrochlorothiazide and amlodipine in the inner layers. Type 2 Amplodipine and hydrochlorothiazide on the outer layers and irbesartan and atenolol in the inner layers. (Xu et al., 2020) a) b) 20 2.4.2 3D Printed Multi-Drug Eluting Implants The term “drug-eluting implants” refers to drug delivery systems and devices that consist of drugs released in surrounding tissue areas after implantation or insertion as a means of achieving targeted therapy (Domsta and Seidlitz, 2021). There are various forms of implants depending on the area of application. Types such as catheters and screws were considered in this study. The most prevalent application of multidrug implants is in bone- related conditions (Gbureck et al., 2007; Wu, Zheng, Guo and Huang, 2009; Wu, Zheng, Guo and Sun, 2009; Vorndran et al., 2010; Inzana et al., 2015; Li et al., 2015; Zhu et al., 2015; Wu et al., 2016; Trombetta et al., 2019; Chou et al., 2021). Unlike in oral systems, the commonly employed printing technique for implants is binder ink jetting, the advantages of which (over the other techniques) include low costs, fast production, multi-material printing, no need for supporting structures, low-temperature processing and high porosity products (Domsta and Seidlitz, 2021). Drug loading in 3D printed implants is done either pre-, during or post-printing. Incorporation of the drug pre-printing is normally applied in FDM/HME, whereby API and polymer filaments are made in an extruder. Binder inkjet printing enables the incorporation of drugs into the implant during the printing process as they can be added to the binder solution (Wu, Zheng, Guo and Huang, 2009; Inzana et al., 2015; Wu et al., 2016; Trombetta et al., 2019). The drugs that are incorporated into the implant matrix before or during the printing process have to withstand all conditions of the preparations and the actual printing process. Consequently, some sensitive drugs are excluded from those mechanisms due to their degradation properties. Post-printing loading on or into the 3D printed implant can either be done by, coating (Boyer, Ballard, et al., 2018; Boyer, Woerner, et al., 2018; Poudel et al., 2020), immersion into the drug solution under a vacuum to aid absorption, incubation with sublimated iodine (Boyer, Woerner, et al., 2018), use of supercritical carbon dioxide (Ngo et al., 2020) or manual filling of the processed drug into the 3D printed hollow or reservoir device (Arany et al., 2020; Stewart, Domínguez-Robles, McIlorum, Gonzalez, et al., 2020; Stewart, Domínguez-Robles, McIlorum, Mancuso, et al., 2020). These mechanisms can be quite cumbersome and are limited to porous implants. Table 2.2 provides a summary of developed 3D printed multi-drug implants. 21 Table 2.2: 3D Printed multi-drug implants. Implant type Drugs loaded 3DP Technique Design and Objective Reference Stents and Catheters. Gentamicin Sulphate, Methotrexate HME/FDM Disc, bead, catheter. 3D printing of different constructs with antibiotic or chemotherapeutic-eluting filament. (Weisman et al., 2019) Bone treatment screws Vancomycin Hydrochloride, Ofloxacin, Tetracycline Hydrochloride Binder Ink jetting Cylinder Drug adsorption and desorption of low-temperature 3D printed ceramic scaffolds. (Gbureck et al., 2007) Levofloxacin, Rifampicin Isoniazid, Binder Ink jetting Multi-layered cylinder. For the bi-modal release profile. (Wu, Zheng, Guo and Huang, 2009) Isoniazid Rifampicin Binder Ink Jetting Programmed sequentially release of multidrug implant. (Wu, Zheng, Guo and Sun, 2009) Levofloxacin, Tobramycin Binder Ink Jetting For bone tuberculosis treatment for treatment of chronic osteomyelitis. (Wu et al., 2016) Vancomycin, Heparin, rhBMP-2 Binder Ink Jetting Scaffold Bio-ceramic implants with high accuracy of drug deposition to modify the release. (Vorndran et al., 2010) Isoniazid, Rifampicin Extrusion Scaffold. 3D printed scaffolds with antitubercular drugs in animal model (Li et al., 2015; Zhu et al., 2015) Rifampicin, Vancomycin Binder Ink Jetting Scaffold. Simultaneous local delivery of Rifampicin and Vancomycin from 3D printed ceramic scaffolds for bone infection treatment examined in a mouse model. (Inzana et al., 2015) Rifampicin, Sitafloxacin Binder Ink Jetting Scaffold. 3D printing of antibacterial scaffolds for osteomyelitis treatment. (Trombetta et al., 2019) Vancomycin, Ceftazidime Extrusion Screw. Influence of Printing parameter on drug-eluting screws 3D printed by a solution-technique (Chou et al., 2021) 2.5 Additive Materials Used in 3D Printing of Multi-Drug Systems One of the major constraints preventing mass adoption of 3D printing for biomedical manufacturing is a lack of 3D printing biomaterials that is, polymers, biomaterials, hydrogels, and bioinks that are functional for 3D printing, biocompatible, and perform well from a biomechanical standpoint (Pugliese et al., 2021). Of the aforementioned 3D printing materials, 22 polymers are the most commonly employed biomaterial in multi-drug systems (Khaled et al., 2015b, 2015a; Awad et al., 2019; Robles-martinez et al., 2019; Siyawamwaya et al., 2019b). The ability to modulate the release of different actives in multi-drug systems forms the basis upon which polymers are selected in the 3D printing thereof. Other equally important characteristics include the materials’ printability, ease of processability, cost, biocompatibility, and degradation rates. In fused deposition modelling (FDM) techniques polymers are processed into drug-loaded filaments prior to printing, SLS in the form of powder beads, solutions in SLA, and gels in direct ink writing (DIW). Polymers are incorporated as either binders, disintegrants, compression aids, diluents, or fillers for modulation of drug release. Because of their application versatility, polymers are employed in pharmaceutical 3D printing for a variety of purposes, such as controlling dosage shape, size, and drug release. More importantly, the majority of them are biocompatible, biodegradable, have manipulable mechanical properties, and degradation rates, and can be dissolved in rapidly evaporating organic solvents such as dichloromethane (Pugliese et al., 2021). The type of polymer or material employed in 3D printing depends on the technique being applied although some polymers may be used in more than one technique. Generally, in FDM/HME printing thermoplastic polymers such as polycarbonate, acrylonitrile butadiene styrene (ABS), polylactic acid (PLA) and nylon are used; SLS uses Poly ε-caprolactone (PCL) and polyamides, in inkjet printing any polymer that can be supplied as powder may be used (Jain et al., 2018) whilst in SLA, liquid photosensitive polymers such as epoxy or an acrylate based resin e.g polyethylene glycol diacrylate and photoinitiators such as diphenyl (2,4,6- trimethylbenzoyl)-phosphine oxide (DPPO) are employed (Healy et al., 2019). Numerous polymers of natural and synthetic origin have found applications in biomedical 3D printing. For instance, natural polymers like gelatine, collagen, alginate, and chitosan are usually utilized, but they frequently necessitate cross-linkers which could be cytotoxic, thus synthetic polymers have recently gained attention for 3D printing to avoid such manner of drawbacks (Gross et al., 2014; Stansbury and Idacavage, 2016; Ligon et al., 2017). Cellulose-based polymers have long been used in the pharmaceutical industry to produce solid dosage forms, and they have shown great promise in 3D printing techniques. Of the two main cellulose derivatives, cellulose ethers comprise of the commonly employed methylcellulose, ethyl cellulose (EC), hydroxypropyl cellulose (HPC). The derivatives of which are formed by substitutions with methyl and hydroxypropyl groups yielding the likes of hydroxypropyl methylcellulose (HPMC), a widely studied polymer. Drug release from HPMC matrices is retarded using higher molecular weight and viscosity grades. As the degree of 23 substitutions increases, the solubility profiles shift from dilute alkali to water and finally to an organic-solvent-soluble stage (Raj et al., 2021). A promising approach in controlled release systems, is the quarternisation of polymers, that is, the addition of an amino group, for example, PQ-10 also known as quaternized hydroxyethylcellulose ethoxylate, a cationic cellulose derivative. The unique ability of these polyelectrolyte complexes (PEC) to self-assemble has sparked interest in 3D printing applications, including multidrug systems (Siyawamwaya et al., 2017). In a recent study by Siyawamwaya and colleagues, a novel PEC of electrostatically bonded PQ-10 and humic acid was synthesised and used as bio-ink in 3D printing an antiretroviral multi-drug system for enhanced bioavailability, as shown in Figure 2.5. Figure 2.5: Picture of the self-assembly of HA and PQ-10 and the fibrous PECs collected by filtration before drying. SEM image of 3D printed FDC, alongside prolonged drug release profiles of a) Efavirenz (EFV), b) Tenofovir Disoproxil Fumarate (TDF) and c) Emtricitabine (FTC) in comparison to the commercial version, Atripla®. Reproduced with permission from (Siyawamwaya et al., 2019a) 2019 Elsevier. PCL and PLA are low-cost, biodegradable, non-toxic, hydrophobic polyesters, which can be used alone or in combination. PCL has a melting point of 55–60 °C, Tg of −54 °C, is soluble in organic solvents and is commonly used for long-term implant delivery devices due to its very low in vivo degradation and can be used in tablets and nanotubes (Langer, Basu and Domb, 2016). Several forms of PLA exist because of the chirality of the starting monomers, i.e., lactic acid (L) and lactide (D). These L or D configurations own various properties and 24 have the ability to form stereo-complexes between the L-PLA and D-PLA forms giving rise to; PDLA PLGA poly (lactic-co-glycolic acid), PLLA (poly-l-lactic acid) and PDLLA (poly-d,l-lactic acid ) (Ligon et al., 2017; Azad et al., 2020). The latter two possess favourable thermal and mechanical properties, a Tg range of 55–65 °C and mechanical strength of 1.9 to 4.8 GPa. Therefore, PCL is often blended with the chiral forms of PLA to achieve better erosion properties (Jain et al., 2018). Eudragit® represents a set of synthetic polymethacrylate, non-biodegradable, non-absorbable, nontoxic and amorphous polymers. According to Evonik, all Eudragit polymers have thermoplastic properties, low glass transition temperatures (between 9 °C and > 150 °C), high thermostability, and high miscibility with APIs and other excipients (Evonik, 2018). Varying the functional group on the polymer dictates the type of drug release it is best suited for. For example, the Eudragit E series, soluble at low pH is used in immediate-release formulations, L and S series show delayed release in drug formulations. Eudragit® RL and Eudgrait® RS are insoluble with pH-independent swelling, Eudragit® RL has high permeability while Eudragit® RS has low permeability. Thus, combining the series together enables pharmaceuticals with customized time-controlled release profiles. As a water-insoluble thermoplastic polymer, EC is often used for its sustained release capabilities in FDM 3D printing (Thakral, Thakral and Majumdar, 2013; Okwuosa et al., 2017). Other immediate release polymers include HPMC E5 and PVA a biocompatible, hydrophilic thermoplastic polymer, exhibiting a Tg of 85 °C, a melting point range of 180 to 228 °C. PVA is suitable for immediate-release tablets as it dissolves more readily in hydrochloric acid (Azad et al., 2020). Khaled in the construction of a multidrug compartmentalised system, employed croscarmellose sodium (CCS), sodium starch glycolate (SSG) (disintegrants), polyvinylpyrrolidone (PVP K30) (a binder) and D-mannitol (a filler), in making the joining layer of the two compartments. It was designed to disintegrate quickly within the 1st hour of the dissolution test and allow the tablet to split into two parts with different drug release mechanisms (Khaled et al., 2015b). PVA may also be used in making individualised gastro- resistant tablets, by creating a drug-loaded PVA core with an enteric shell. Another study also suggests that the total drug release can be achieved within 3 minutes by reducing the infill rate of PVP tablets to 50 %, which in turn increases the porosity of tablets (Okwuosa et al., 2017; Kempin et al., 2018). For the manufacturing of implants, biocompatible and biodegradable polymers such as PLA, poly(lactic-co-glycolic acid) (PLGA) or polycaprolactone (PCL) are commonly used, others include cellulose derivatives, PVA, Eudragit, ethylene vinyl acetate (EVA), polyethylene oxide (PEO) and thermoplastic polyurethane (TPU) (Shaqour et 25 al., 2020). Wu et al (Wu et al., 2016), employed one of the chiral forms of PLA, PDLLA (Mw = 100 kDa) in the fabrication of a 3D printed multi-drug implant for chronic osteomyelitis. 2.5.1 Other High Functionality Polymers The advent of 3D printed biomedicals has stimulated the development of a whole range of high-performance polymers such as polyether ether ketone (PEEK), polyether ketone (PEKK), Polyetherimide (ULTEM) and engineering ceramics like alumina, zirconia and silica. The unique properties of these polymers, that is resistance to extreme heat, incredible hardness, biocompatibility, and rubber-like qualities, set them apart. PEEK is the most popular of the aforementioned materials. Its popularity is attributed to a variety of favourable properties, including heat resistance of up to 260 °C, water resistance, lightweight compared to metals, and, most importantly, biocompatibility. All current PEEK materials for 3D printing are obtainable as filaments and can be 3D printed using FDM machines (Beamler, 2019). Kang et al., successfully employed FDM in the development of PEEK implants for bone mandibular defects, producing a novel mandibular reconstruction model with high stability, proven by deformation results (Kang et al., 2021). However, PEEK, like other high-functionality polymers, has its limitations, including poor integration with surrounding bone tissue post- implantation. Therefore, to improve PEEK bioactivity, numerous strategies for functionalizing the PEEK surface and changing the PEEK structure have been proposed (Gu et al., 2021). Composites such as graphene and nanomaterials, and even 4D materials also fall under the category of high-performance polymers. 2.6 Limitations and Barriers to Implementation Many specialists around the world share the ambition for 3D printing, with new research papers being published more frequently than not to highlight the technology's potential in formulation development and patient care. Regardless of the hype, countries are certainly encountering difficulties in introducing and implementing this technology within the pharmaceutical sectors. Aprecia Pharmaceuticals’ Spritam® (Levetiracetam), the first and only commercialised 3D printed pharmaceutical to this date confirms this. Large biopharmaceutical firms have shown little interest in this field (Wright, 2005). 2.6.1 Regulatory Constraints Despite the FDA's (United States of America) efforts as forerunners in implementing 3D printing, as evidenced by the Guide provided in 2017 (Reddy et al., 2020), there are still some grey areas in terms of regulatory requirements, making it a big hurdle for manufacturers worldwide (Bhusnure et al., 2016). Biomedical systems, unlike other 3D printed products, 26 require special attention when it comes to rules and constraints. Compared to the conventional ways of developing oral dosage forms, 3D printing processes are distinct and still novel in many pharmaceutical manufacturing sectors. As a result, quality control testing on items such as tablets needs to be revised. Promisingly, in March 2021, the Medicines and Healthcare Products Regulatory Agency (MHRA) proposed a new regulatory framework towards the development of point-of-care manufacturing and supply, including 3D printing technologies for customised medicine production (Abdul and Trenfield, 2022). 2.6.2 Processing Parameters 2.6.2.1 Material Attributes and Selection The type of binders used varies depending on the product being created. Biocompatible binders must be considered in order to develop biomedical products, and knowledge about these binders is still restricted. The materials employed must be of good quality and safe for use in the body of the patient; the only materials that are compatible are those that are organically based, which are unfortunately presently scarce (Shahrubudin et al., 2020). 3D printed dosage forms possess low mechanical properties, which may be favourable in the case of oral dosage forms designed for immediate release. Nonetheless, the printed product should have acceptable tensile, compressional strength and flexible rigidity where applicable. The optimum bone tissue scaffold, for example, has macro-pores of 300–900 μm and porosity of 60–95 % in load-bearing bone tissue regeneration. Fortunately, with Selective Laser Melting (SLM) 3D printing technique, porous titanium implants with great osteo-integration in vivo performance with pore size 400–1000 μm can be fabricated (Shahrubudin et al., 2020). 2.6.2.2 Printer Accuracy Low dimensional accuracy may seem counterintuitive when discussing 3D printing, yet depending on the object being developed, such as implants and body parts, high levels of accuracy may be important. When employing randomly selected equipment and materials, producing 3D printed biomedical items with the exact size and structure is difficult. Low dimensional accuracy products will not fit in the body, lowering clinical success rates. The difficulty is to overcome product shrinkage throughout the curing and cooling process. 2.6.2.3 Return on Investment (ROI) Additionally, respondents in a survey done in Malaysia highlighted four hurdles to the implementation of 3D printing with regard to management: re-education of workers, high- priced products, a lack of guidelines, and cyber-security issues. Because the technology is still a relatively new notion in many circles, basic operational knowledge may be insufficient. 27 Employees will need to enhance their education and skill levels for organ