Dersleri yüzünden oldukça stresli bir ruh haline sikiş hikayeleri bürünüp özel matematik dersinden önce rahatlayabilmek için amatör pornolar kendisini yatak odasına kapatan genç adam telefonundan porno resimleri açtığı porno filmini keyifle seyir ederek yatağını mobil porno okşar ruh dinlendirici olduğunu iddia ettikleri özel sex resim bir masaj salonunda çalışan genç masör hem sağlık hem de huzur sikiş için gelip masaj yaptıracak olan kadını gördüğünde porn nutku tutulur tüm gün boyu seksi lezbiyenleri sikiş dikizleyerek onları en savunmasız anlarında fotoğraflayan azılı erkek lavaboya geçerek fotoğraflara bakıp koca yarağını keyifle okşamaya başlar

GET THE APP

Journal of Analytical & Bioanalytical Techniques - Solid State Characterization and Pharmaceutical Development
ISSN: 2155-9872

Journal of Analytical & Bioanalytical Techniques
Open Access

Like us on:

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Short Communication   
  • J Anal Bioanal Tech, Vol 12(6)
  • DOI: 10.4172/2155-9872.1000433

Solid State Characterization and Pharmaceutical Development

Kaushalendra Chaturvedi*
Research Department, J-Star Research Inc, United States
*Corresponding Author: Kaushalendra Chaturvedi, Research Department, J-Star Research Inc, United States, Email: kaushalendra.chaturvedi@my.liu.edu

DOI: 10.4172/2155-9872.1000433

Abstract

Pharmaceutical materials solid-state property plays a critical role from early discovery to finalizing the formulation type. Pharmaceutical material exists in a crystalline or amorphous state. The crystalline state generally shows a high melting point, less hygroscopic, low solubility, lower bioavailability, and higher Physico-chemical stability compared to the amorphous material. A major concern in the pharmaceutical industry is the selection of solids state forms for the final formulations. This is because it can significantly affect the drug product quality in terms of Physicochemical
stability, processibility, solubility, bioavailability, and having regulatory, legal, and commercial implications.

Keywords:  Pharmaceutical materials; Crystalline; Hygroscopic

Introduction

Pollution Intermolecular force differences can result in significant variation of the physicochemical properties between different solidstate forms of pharmaceuticals. Particle morphology, mechanical properties including powder flow ability, and compressibility are greatly influenced by the molecular packing of the crystal. The thermodynamically stable crystalline form shows a higher melting point form, lower solubility, and dissolution rate since it has higher intermolecular forces compared to metastable or amorphous form [1- 4].

Short Communication

Bahir Solid-state failure of ritonavir marketed formulation brought the attention of regulatory agencies and the scientific community about the potential need of organized decision-making tree to determine and established the best suitable solid form of the particular drug in a given condition [5-7]. As mentioned in many published literatures that solid form is important in pharmacology because an insoluble solid-state form may give undesired outcomes and lowering the efficacy, and potency of the drug. Secondly API manufacturing is also very important because regulatory agencies required information on which solid-form is being manufactured and supplied. To get the same solid form from batch-to-batch manufacturing it is important to establish properties such as solubility in crystallizing solvent, metastable limit, and other solid-state properties to make a robust manufacturing process. It is imperative to establish the solid-state properties of the drug used in the formulation in early developmental research to circumvent drug as well as drug product failure. Therefore, in addition to traditional analytical tests such as chromatography, UVspectroscopy, and dissolution testing it is important to include solid state characterization analytical techniques in decision making process [8-15]. These solid-state properties information will help the chemists and formulation scientists to assemble necessary information and develop a robust, most efficient process to design crystallization process and drug delivery system, respectively.

Significance of Solid-State Characterization

A solid form of the API plays a decisive role in the development of a commercial pharmaceutical product [16,17]. The formulator perspective's first choice is a selection of thermodynamically most stable form for the final formulation development to lower the potential risk associated with product stability. Most of the new drugs fall in the BCS class of II and IV because of poor water solubility, which ultimately causes a lower dissolution rate and that may not be adequate to achieve good bioavailability. Therefore, to achieve a certain level of the drug in the blood different enabling technologies Solid-lipid nanoparticles, self-micro emulsifying (SMEDD), self-nano emulsifying (SNEDD), liposomes, complexation, solid dispersion, and nanosuspensions being used to increase the apparent solubility of the drug [18-20]. However, most of the enabling formulations have their limitation and potential challenges leading to their inherently lower physical and chemical stability.

For example, the high hygroscopicity of amorphous material may lead to increased chemical reactivity and the propensity to crystallize, since the absorbed moisture acts as a plasticizer. The stability of amorphous material is improved by making solid dispersion where the drug is molecularly dispersed with the polymer and the polymer acts as crystallization inhibitor either because of steric hindrance or viscosity. In these approaches, physical stability is often a concern, however, since crystallization of the drug during storage would negatively impact the solubility and intended therapeutic efficacy of the product.

A second such example is nanoparticle formulation since the nanoparticles have higher surface energy which leads to its potential to increase the individual particle size by ripening or agglomeration over the period. This indicates that because of the high free energy environment drug in the formulation may not be stable during the intended downstream process or the storage time. Unlike small organic drug molecules, in the solid protein formulations, the excipient’s solid-state form plays a crucial role in product stability and performance. To stabilize the protein formulation interactions with stabilizing excipients are very important that can occur only when the excipient and protein molecules both are in the same amorphous phase. Therefore, in protein formulation protein aggregation and loss of therapeutic activity are mainly caused due to the crystallization of stabilizing excipient.

Drug or excipients solid-state forms are critical in defining product stability or efficacy; therefore, it is important to maintain consistent solid-state form through the drug product processing steps. However, as mentioned earlier having higher kinetic energy could lead to solid-state transformations, and it is not uncommon to see such incidences during pharmaceutical processing or storage. The solid-state formation could result in getting a pure phase of one form or a mixture of forms. Most commonly a mixture of forms is seen as a result of such transformation, one such example is an occurrence of hydrate-anhydrate mixtures caused because of routine manufacturing processes. Pharmaceutical manufacturing involves several unit operations including milling, granulation [21], drying, tablet compression [22], and coating; this involves high thermal and mechanical stresses, exposure to solvents, which may induce solid-state transformations. Unlike drugs, excipient manufacturing is not strictly controlled leading to different solid states between the manufacturers. To make good quality, robust, reproducible process product assessment of solid-state forms during manufacturing and storage are imperative. If pharmacologically accepted, then early detection and quantification of transformations will help to set the acceptable limit of such changes. Process analytical technology (PAT) tools are important to discover such solid-state transformations during the process and help to control and optimize the processes to make a cost effective and good quality product.

Solid-State Characterization Techniques

Solid-state characterization of the pharmaceuticals is essential in drug discovery as well as in preformulation studies. Literature suggests that moving solid-state characterization earlier and earlier in drug discovery ensures that a developable solid form is physically and chemically stable. In solid-state characterization, an important goal is identifying the differences between amorphous and crystalline materials.

Several solid-state characterization techniques are used to analyze pharmaceutical materials. Generally, data from two or more techniques are required to establish the relationship between the solidstate, and the choice of technique is depending on the level of understanding required. The most commonly used methods for solidstate characterization have been X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), dynamic vapor sorption (DVS), mid-infrared spectroscopy and microscopy and solid-state nuclear magnetic resonance (ss-NMR). Other routine techniques which are mainly used as process monitoring and control (PAT) tools are Raman spectroscopy, nearinfrared spectroscopy (NIR), and Blaze imaging techniques. The top six analytical techniques are briefly discussed later.

The long-range order of molecular packing can be easily observed with XRPD [23,24]. Crystalline material powder pattern will show sharp intense peaks in diffractogram unlike amorphous which only shows halo because of the lack of in-phase reflections from crystal planes. A different crystalline form of the same material will show a unique powder pattern which helps to determine the phase purity of the material. The second most important analytical tool could be differential scanning calorimetry (DSC) in which crystalline material will show sharp melting endothermic event, unlike amorphous material which will only show glass transition temperature (Tg) [25]. However, sometimes finding Tg of the amorphous material may need some modifications in DSC experiments involving slow ramp rate or changes in the modulation temperatures. Secondly, DSC can be very useful in determining the enantiotropic or monotropic relationship between two crystalline forms of the same material [26].

Conclusion

Most often DSC data is correlated with the TGA data to determine if the resultant crystalline form is solvated, hydrate or anhydrate. The water activity or DVS data helps to determine the stability of the solid form under different equilibrium relative humidity conditions. Vibrational spectroscopy mainly is routinely used in crystallization because of its ability to distinguish two crystalline forms dues to the differences in intermolecular bonding. A polarized microscope (PLM) is a quick test to find if the material is amorphous or crystalline, under PLM crystalline material will show birefringence which will be absent in amorphous material. However, PLM alone is not useful in distinguishing different crystalline forms of the same material unless they have specific crystal morphology.

References

  1. Gruss M (2019) Regulatory Aspects for Formulation Design–with Focus on the Solid State, Innovative Dosage Forms: Design and Development at Early Stage. Innovative Dosage Forms 155-207.
  2. Zane P, Gieschen H, Kersten E, Mathias N, Ollier C, et al. (2019) In vivo models and decision trees for formulation development in early drug development: A review of current practices and recommendations for biopharmaceutical development. European J Pharm Biopharm. 142: 222-231
  3. Chow K, Tong HH, Lum S, Chow AH (2008) Engineering of pharmaceutical materials: An industrial perspective. J Pharm Sci. 97: 2855-2877.
  4. Shah HS, Chaturvedi K, Dave RH, Morris KR (2021) Molecular insights into warfarin sodium 2-propanol solvate solid form changes and disproportionation using a low volume two-stage dissolution approach. Mol Pharm. 18: 1779-1791.
  5. Shah HS, Chaturvedi K, Hamad M, Bates S, Hussain A, Morris K (2019) New insights on solid-state changes in the levothyroxine sodium pentahydrate during dehydration and its relationship to chemical instability, AAPS Pharm Sci Tech, 20: 1-10.
  6. Serajuddin AT, Thakur AB, Ghoshal RN, Fakes MG, Ranadive SA, et al. (1999) Selection of solid dosage form composition through drug excipient compatibility testing, J pharm sci. 88: 696-704.
  7. Chemburkar SR, Bauer J, Deming K, Spiwek H, Patel K, et al. (2000) Dealing with the impact of ritonavir polymorphs on the late stages of bulk drug process development. Org Process Res Dev.  4: 413-417.
  8. Chaturvedi K, Nanda R (2010) Hyphenated gas chromatography. Int J Pharm Sci Rev Res. 5: 18-27.
  9. Chitlange SS, Chaturvedi KK, Wankhede SB (2011) Development and validation of spectrophotometric and HPLC method for the simultaneous estimation of salbutamol sulphate and prednisolone in tablet dosage form, J Anal Bioanal Tech. 2(2).
  10. Chitlange SS, Shinde PS, Chaturvedi KK, Pandkar SV, Wankhede SB (2011) High performance thin layer chromatographic estimation of thiocolchicoside and aceclofenac in bulk and in pharmaceutical dosage forms, Inventi Rapid: Pharm Ana & Qual Assur.
  11. Chitlange SS, Chaturvedi KK, Tawargeri SR, Wankhede SB (2011) UV spectroscopic and stability-indicating tlc-densitometric method for simultaneous estimation of salbutamol sulphate and prednisolone in pharmaceutical dosage form. J Res Chem. 4(5): 786-790.
  12. Chitlange SS, Tawargeri SR, Chaturvedi KK (2011) Simultaneous determination of amoxicillin trihydrate and ambroxol hydrochloride in solid dosage form by spectrophotometric and stability indicating rp-hplc method. J Res Chem. 4(6).
  13. Chitlange SS, Tawargeri SR, Chaturvedi KK, Wankhede SB (2011) UV spectrophotometric estimation of amoxicillin trihydrate and ambroxol hydrochloride in bulk and combined pharmaceutical dosage form, inventi impact: Pharm Ana & Qual Assur.
  14. Chaturvedi K, Shah HS, Sardhara R, Nahar K, Dave RH, et al. (2021) Protocol development, validation, and troubleshooting of in situ fiber optic bathless dissolution system (FODS) for a pharmaceutical drug testing, J Pharma Biomed Anal. 195: 113833.
  15. Sardhara R, Chaturvedi K, Shah HS, Vinjamuri BP, Al-Achi A, et al. (2021) Predictive performance comparison of computed linear and quadratic multivariate models for in-situ uv fiber optics tablet dissolution testing. Eur J Pharm Sci, 161: 105806
  16. Variankaval N, Cote AS, Doherty MF (2008) From to function: Crystallization of active pharmaceutical ingredients, AIChE j, 54(11): 1682-1688
  17. Nasr MM, Krumme M, Matsuda, Trout BL, Badman C, et al. (2017) Regulatory perspectives on continuous pharmaceutical manufacturing: Moving from theory to practice: September 26-27, 2016, international symposium on the continuous manufacturing of pharmaceuticals. J pharm sci. 106 (11): 3199-3206.
  18. Dangre P, Chaturvedi K, Pofali P (2019) Lipidic nanoparticles: A platform for advancement in drug delivery systems, molecular chemistry and biomolecular engineering: Integrating Theory and Research with Practice. 191.
  19. Chalikwar SS, Surana SJ, Goyal SN, Chaturvedi KK, Dangre PV (2021) Solid self-microemulsifying nutraceutical delivery system for hesperidin using quality by design: assessment of biopharmaceutical attributes and shelf-life. J Microencapsul. 38(1): 61-79.
  20. Dangre PV, Dusad PP, Singh AD, Surana SJ, Chaturvedi KK, et al. (2021) Fabrication of hesperidin self-micro-emulsifying nutraceutical delivery system embedded in sodium alginate beads to elicit gastric stability. Polymer Bulletin. 1-22.
  21. Chaturvedi K, Gajera BY, Xu T, Shah H, Dave RH (2018) Influence of processing methods on physico-mechanical properties of Ibuprofen/HPC-SSL formulation. Pharm Dev Technol. 23(10): 1108-1116.
  22. Saddik J, Chaturvedi K, Dave R (2011) Investigating the impact of manufacturing and formulation factors on the compactability of acetaminophen tablets using heckel and multivariate analysis. Int J Pharm. 413(1-2): 29-35.
  23. Chaturvedi K, Shah HS, Nahar K, Dave R, Morris KR (2020) Contribution of crystal lattice energy on the dissolution behavior of eutectic solid dispersions. ACS omega. 5(17): 9690-9701.
  24. Shah HS, Chaturvedi K, Zeller M, Bates S, Morris K (2019) A threefold superstructure of the anti-epileptic drug phenytoin sodium as a mixed methanol solvate hydrate, Acta Crystallogr C Struct Chem. 75(pt 9): 1213-1219
  25. Chaturvedi KK (2019) Modeling of adhesion in tablet compression at molecular level using thermal analysis and molecular simulations, long island university, The Brooklyn Center.
Top