View Article

  • Design, Synthesis and Biological Evaluation of Alpha Amylase Inhibitor Against Diabetes Thiazolidine Ring

  • S. N. D. College of Pharmacy, Yeola Dist-Nashik

Abstract

Diabetes mellitus, a chronic metabolic disorder affecting millions worldwide, is characterized by hyperglycemia resulting from defects in insulin secretion, insulin action, or both. Postprandial hyperglycemia, a significant contributor to diabetic complications, can be effectively managed by inhibiting carbohydrate-breaking enzymes like alpha-amylase. Alpha-amylase inhibitors delay the digestion and absorption of carbohydrates, thereby mitigating the sharp rise in blood glucose levels after meals. The thiazolidine ring, a privileged scaffold in medicinal chemistry, has garnered considerable attention due to its diverse pharmacological activities, particularly its well-established role in insulin sensitization (e.g., glitazones). This review focuses on the rational design, various synthetic approaches, and comprehensive biological evaluation of novel alpha-amylase inhibitors incorporating the thiazolidine ring system. We will discuss the structure-activity relationships (SAR) for this class of compounds, highlighting key structural features that contribute to their inhibitory potency and selectivity. Furthermore, we will explore the potential of thiazolidine-based alpha-amylase inhibitors as promising therapeutic agents for the management of type 2 diabetes, considering their dual mechanism of action (alpha-amylase inhibition and potential insulin sensitization). Challenges in drug development, including selectivity, pharmacokinetic properties, and potential side effects, will also be addressed.

Keywords

Alpha-amylase inhibitor, diabetes mellitus, thiazolidine, hyperglycemia, drug design, synthesis, structure-activity relationship, glitazones.

Introduction

Diabetes mellitus represents a global health crisis, with its prevalence steadily increasing. The chronic elevation of blood glucose levels leads to severe macrovascular and microvascular complications, including cardiovascular disease, nephropathy, retinopathy, and neuropathy, ultimately impacting quality of life and lifespan. Current therapeutic strategies for type 2 diabetes include lifestyle modifications, oral hypoglycemic agents, and insulin therapy. Among the oral hypoglycemic agents, those that target postprandial hyperglycemia play a crucial role in preventing long-term complications.

Alpha-amylase (α-1,4-glucan-4-glucanohydrolase; EC 3.2.1.1) is a key enzyme involved in the initial digestion of complex carbohydrates in the small intestine. It catalyzes the hydrolysis of α-1,4-glucosidic linkages in starch and glycogen into smaller oligosaccharides and disaccharides (maltose, maltotriose, and dextrins). Inhibiting alpha-amylase activity can significantly slow down carbohydrate digestion, thereby reducing the rate of glucose absorption into the bloodstream and consequently lowering postprandial blood glucose excursions. Acarbose, voglibose, and miglitol are examples of currently available alpha-amylase inhibitors. However, these drugs often present with gastrointestinal side effects such as flatulence, abdominal pain, and diarrhea, limiting patient compliance. Therefore, there is an urgent need for the discovery and development of novel, more effective, and safer alpha-amylase inhibitors.

The thiazolidine ring system, a five-membered heterocyclic ring containing sulfur and nitrogen atoms, is a well-established pharmacophore in medicinal chemistry. Its derivatives, particularly the thiazolidinediones (TZDs), have been successfully employed as insulin sensitizers (e.g., rosiglitazone, pioglitazone) by activating peroxisome proliferator-activated receptor gamma (PPAR$\gamma$). Given the multifaceted nature of diabetes and the promising biological activities associated with the thiazolidine scaffold, exploring its potential as a core structure for developing novel alpha-amylase inhibitors presents an intriguing avenue for research. This review aims to provide a comprehensive overview of the design, synthesis, and biological evaluation of thiazolidine-based compounds as alpha-amylase inhibitors, highlighting their therapeutic potential in diabetes management.

Alpha-amylase and alpha-glucosidase enzymes play crucial roles in carbohydrate digestion by breaking down complex starches into simple sugars, directly contributing to postprandial glucose elevation. The inhibition of these enzymes represents an effective therapeutic approach for controlling hyperglycemia associated with type 2 diabetes. While currently approved inhibitors like acarbose, miglitol, and voglibose are available, their clinical utility is limited by gastrointestinal side effects and moderate efficacy.

Thiazolidine derivatives have emerged as attractive scaffolds for developing novel antidiabetic agents due to their versatile chemical structure and multiple biological activities. The thiazolidine ring system, containing sulfur and nitrogen heteroatoms, provides opportunities for structural modifications that can enhance selectivity and potency against target enzymes.

2. Alpha-Amylase: Structure, Mechanism, and Inhibition

Human pancreatic alpha-amylase (HPA) is a monomeric enzyme composed of approximately 496 amino acid residues, typically organized into three distinct domains (A, B, and C). Domain A is a (β/α)8? barrel and contains the catalytic active site. Domain B is a loop region between β3? and α3? of domain A, while domain C is a β-sheet structure. Calcium ions are essential for the structural integrity and catalytic activity of alpha-amylase, while chloride ions act as allosteric activators.

The catalytic mechanism of alpha-amylase involves a double displacement reaction, proceeding through a covalent glycosyl-enzyme intermediate. Three key acidic residues are conserved in the active site: two glutamate residues acting as nucleophile and acid/base catalyst, and an aspartate residue involved in substrate binding and stabilization.

Alpha-amylase inhibitors primarily act by binding to the enzyme's active site, competing with the natural substrate (starch), and preventing its hydrolysis. These inhibitors can be broadly classified into pseudo-saccharides (e.g., acarbose) and non-pseudo-saccharides. The focus of this review is on synthetic non-pseudo-saccharide inhibitors, specifically those incorporating the thiazolidine ring.

3. Thiazolidine Ring as a Privileged Scaffold

The thiazolidine ring, a saturated analogue of thiazole, has been extensively explored in drug discovery due to its ease of synthesis, metabolic stability, and diverse pharmacological activities. Beyond its prominent role in PPAR$\gamma$ agonism, thiazolidine derivatives have demonstrated a wide array of biological activities, including anti-inflammatory, antimicrobial, anticancer, and antioxidant properties. The presence of both a nitrogen and a sulfur atom in the ring allows for various substitutions, enabling significant structural diversity and fine-tuning of pharmacological profiles.

The ability of the thiazolidine ring to interact with various biological targets, combined with its favorable pharmacokinetic properties, makes it an attractive scaffold for developing new therapeutic agents. Its potential to interact with multiple targets involved in diabetes pathogenesis, such as alpha-amylase and PPAR$\gamma$, offers an exciting opportunity for developing multi-target directed ligands.

4. Design and Synthetic Strategies of Thiazolidine-Based Alpha-Amylase Inhibitors

The rational design of alpha-amylase inhibitors typically involves mimicking the transition state of the enzyme-substrate complex or identifying compounds that can effectively block the active site. For thiazolidine-based inhibitors, various design strategies have been employed:

  • Structure-Based Design: Utilizing the known crystal structures of alpha-amylase complexed with inhibitors or substrates to guide the placement of the thiazolidine ring and its substituents within the active site. This approach often involves molecular docking and dynamics simulations to predict binding affinities and interactions.
  • Ligand-Based Design: Employing pharmacophore modeling or quantitative structure-activity relationship (QSAR) studies based on known alpha-amylase inhibitors to identify common structural features that can be incorporated into the thiazolidine scaffold.
  • Hybrid Approach: Combining elements of both structure-based and ligand-based design, for instance, by linking a known alpha-amylase inhibitor moiety to a thiazolidine ring, or by integrating features known to enhance binding to the enzyme.

4.1. General Synthetic Routes to Thiazolidine Derivatives:

Several well-established synthetic methodologies are available for the construction of the thiazolidine ring. The most common approaches include:

  • Condensation of a 1,2-amino thiol with an aldehyde or ketone: This is a widely used method, involving the reaction of a cysteine derivative or a similar 1,2-amino thiol with a carbonyl compound. This reaction forms a thiazolidine ring through the formation of an imine followed by intramolecular cyclization.
  • Cyclization of a β-keto ester or an α-halo ester with a thiourea or an amide: This approach offers flexibility in introducing various substituents on the thiazolidine ring.
  • Reaction of an imine with a thioglycolic acid derivative: This method provides a straightforward route to substituted thiazolidin-4-ones.

4.2. Synthetic Approaches for Thiazolidine-Based Alpha-Amylase Inhibitors:

Researchers have employed diverse synthetic strategies to synthesize thiazolidine derivatives with potential alpha-amylase inhibitory activity. These often involve:

  • Preparation of substituted thiazolidin-4-ones: Varying the substituents at the C-2, C-3, and C-5 positions of the thiazolidin-4-one core.
  • Incorporation of various heterocycles or aromatic rings: Attaching different heterocyclic or aromatic moieties to the thiazolidine scaffold to enhance interactions with the enzyme's active site.
  • Derivatization of the nitrogen atom: Introducing various groups on the nitrogen atom of the thiazolidine ring can significantly influence activity.
  • Exploration of different linkers: Using various linkers to connect the thiazolidine ring to other pharmacophores.

(This section will be expanded with specific reaction schemes and examples from literature, showing how various substitutions are introduced and how the thiazolidine core is formed. Specific examples of published synthetic routes for potent thiazolidine-based alpha-amylase inhibitors will be detailed here.)

5. Biological Evaluation of Thiazolidine-Based Alpha-Amylase Inhibitors

The biological evaluation of alpha-amylase inhibitors involves both in vitro and in vivo studies to assess their potency, selectivity, and efficacy in managing hyperglycemia.

5.1. In Vitro Alpha-Amylase Inhibition Assay:

  • Enzyme Source: Typically human pancreatic alpha-amylase (HPA) or porcine pancreatic alpha-amylase (PPA) is used.
  • Substrate: Starch (e.g., soluble starch, potato starch) or specific chromogenic substrates like p-nitrophenyl- α-D-maltopentaoside (PNPG5) are employed.
  • Assay Principle: The most common method involves measuring the reduction in the breakdown of starch into reducing sugars, often quantified using the dinitrosalicylic acid (DNS) method or by monitoring the change in absorbance of the chromogenic substrate.
  • Data Analysis: IC$_{50}$ values (concentration of inhibitor causing 50% inhibition) are determined to compare the inhibitory potency of different compounds.
  • Selectivity: It is crucial to assess the selectivity of alpha-amylase inhibitors against other carbohydrate-metabolizing enzymes, particularly alpha-glucosidase. This ensures that the compound primarily targets alpha-amylase and minimizes off-target effects.

5.2. In Vivo Studies:

  • Animal Models: Streptozotocin-induced diabetic rodents (mice or rats) are commonly used to mimic type 1 or type 2 diabetes, or high-fat diet/streptozotocin models for type 2 diabetes.
  • Oral Starch Tolerance Test (OSTT): This is a key in vivo assay to evaluate the ability of the inhibitor to attenuate postprandial glucose excursions after starch administration. Animals are pre-treated with the inhibitor, followed by a starch meal, and blood glucose levels are monitored over time.
  • Long-Term Efficacy Studies: For promising compounds, long-term studies are conducted to assess their effects on fasting blood glucose, HbA1c levels, body weight, and other metabolic parameters.
  • Toxicity Studies: Preliminary toxicity assessments are essential to evaluate the safety profile of the compounds.

(This section will include a detailed discussion of significant studies from the literature, presenting tables of IC$_{50}$ values, in vivo glucose lowering effects, and structure-activity relationships for various thiazolidine derivatives.)

6. Structure-Activity Relationships (SAR) of Thiazolidine-Based Alpha-Amylase Inhibitors

Understanding the SAR is critical for the rational design of more potent and selective inhibitors. For thiazolidine-based alpha-amylase inhibitors, the following general trends have been observed:

  • Substituents on the Thiazolidine Ring: The nature and position of substituents on the thiazolidine ring significantly influence inhibitory activity. For instance, bulky lipophilic groups might enhance binding to hydrophobic pockets within the enzyme, while polar groups could facilitate hydrogen bonding interactions.
  • Linker and Terminal Moieties: The nature of the linker connecting the thiazolidine core to other functional groups, as well as the terminal substituents, plays a crucial role in determining the overall potency and selectivity.
  • Chirality: The presence of chiral centers in some thiazolidine derivatives can lead to significant differences in activity between enantiomers.
  • Comparison with PPAR$\gamma$ Agonism: An interesting aspect of thiazolidine-based alpha-amylase inhibitors is their potential to also exhibit PPAR$\gamma$ agonistic activity. While this could offer a dual mechanism of action, it is important to analyze whether the same structural features are responsible for both activities, or if selective inhibition can be achieved.

7. Advantages and Challenges

7.1. Advantages:

  • Novel Scaffold: The thiazolidine ring offers a fresh perspective for developing alpha-amylase inhibitors, potentially leading to compounds with different binding mechanisms and improved profiles compared to existing drugs.
  • Dual Mechanism Potential: The inherent ability of some thiazolidine derivatives to act as PPAR$\gamma$ agonists alongside alpha-amylase inhibition could provide a powerful multi-pronged approach to diabetes management.
  • Synthetic Tractability: The well-established synthetic routes for thiazolidine derivatives allow for facile chemical modifications and the generation of diverse libraries for screening.

7.2. Challenges:

  • Selectivity: Ensuring high selectivity for alpha-amylase over alpha-glucosidase and other crucial enzymes in carbohydrate metabolism is paramount to minimize off-target effects and gastrointestinal side effects.
  • Pharmacokinetic Profile: Optimizing absorption, distribution, metabolism, and excretion (ADME) properties is crucial for achieving good in vivo efficacy and oral bioavailability.
  • Off-target Effects/Toxicity: Thorough toxicity assessments are required, especially given the history of some thiazolidinediones (e.g., troglitazone) being withdrawn due to liver toxicity.
  • Drug-Drug Interactions: Potential interactions with other co-administered medications need to be investigated.

8. Future Perspectives

The research into thiazolidine-based alpha-amylase inhibitors is a rapidly evolving field with significant promise. Future directions in this area could include:

  • Targeting Pancreatic vs. Salivary Alpha-Amylase: Designing inhibitors that selectively target pancreatic alpha-amylase could reduce side effects related to salivary amylase inhibition.
  • Co-crystallization Studies: Obtaining co-crystal structures of thiazolidine inhibitors with alpha-amylase would provide invaluable insights into their precise binding modes, facilitating more rational drug design.
  • Prodrug Strategies: Developing prodrugs to improve the pharmacokinetic properties and reduce potential side effects.
  • Combination Therapies: Investigating the synergistic effects of thiazolidine-based alpha-amylase inhibitors with other antidiabetic agents.
  • Computational Approaches: Leveraging advanced computational methods, such as artificial intelligence and machine learning, for high-throughput virtual screening and de novo drug design.

RESULT

Design and Synthesis Results:

  • Diverse Derivatization: Researchers frequently synthesize a series of thiazolidine derivatives by modifying various positions on the ring, particularly the C-5 and N-3 positions of the thiazolidine-2,4-dione (TZD) scaffold.
  • Molecular Hybridization: A common and successful strategy involves conjugating the thiazolidine ring with other known pharmacophores or heterocyclic systems (e.g., triazoles, pyrazoles, indoles, oxadiazoles, quinolines). This often leads to enhanced inhibitory activity due to improved binding interactions with the enzyme's active site.
  • Computational Tools (In Silico):
  • Molecular Docking: Consistently used to predict binding affinities and modes of interaction. Compounds showing good binding scores and interactions (e.g., hydrogen bonds with catalytic residues, hydrophobic interactions) are prioritized for synthesis and in vitro testing.
  • Molecular Dynamics (MD) Simulations: Increasingly employed to confirm the stability of the enzyme-inhibitor complex over time, providing more dynamic insights than static docking.
  • QSAR (Quantitative Structure-Activity Relationship) Studies: Help establish mathematical relationships between structural features and biological activity, guiding further optimization.
  • ADMET Prediction: In silico prediction of Absorption, Distribution, Metabolism, Excretion, and Toxicity profiles is routinely performed to filter out compounds with unfavorable drug-likeness properties early in the discovery process. Compounds often show good predicted oral bioavailability and low toxicity profiles.
  • Synthetic Routes: Well-established synthetic methodologies are utilized, including Knoevenagel condensation for C-5 functionalization, and "click chemistry" (e.g., CuAAC) for incorporating triazole moieties.

2. Biological Evaluation Results:

  • Potent In Vitro α-Amylase Inhibition: Many synthesized thiazolidine derivatives demonstrate significant in vitro α-amylase inhibitory activity, often with IC$_{50}$ values comparable to or even surpassing the standard drug Acarbose.
    • For instance, recent studies have reported thioxothiazolidine derivatives with IC$_{50}$ values as low as 0.71 ± 0.01 $\mu$M, significantly outperforming acarbose.
    • Thiazolidine-2,4-dione tethered 1,2,3-triazole derivatives have also shown comparable inhibition to acarbose, with potent compounds exhibiting IC$_{50}$ values around 0.040 $\mu$mol mL$^{-1}$.
  • Mechanism of Inhibition: Kinetic studies (e.g., Lineweaver-Burk plots) often reveal a mixed-type or competitive inhibition mechanism, indicating that the compounds bind within or near the active site of α-amylase, interfering with substrate binding or catalysis.
  • Selectivity: While some compounds exhibit dual α-amylase and α-glucosidase inhibition, some recent designs specifically aim for selective α-amylase inhibition. This selectivity can be advantageous in potentially reducing the gastrointestinal side effects often associated with strong α-glucosidase inhibition (e.g., flatulence, diarrhea).
  • In Vivo Antidiabetic Efficacy: The most promising compounds from in vitro studies are advanced to in vivo evaluation in diabetic animal models (e.g., streptozotocin-induced or alloxan-induced diabetic rats).
    • Reduced Blood Glucose: A consistent and crucial finding is the significant reduction in fasting and postprandial blood glucose levels in treated diabetic animals, confirming the antidiabetic potential.
    • Improved Biochemical Markers: Treatment often leads to favorable changes in various biochemical parameters, including improved urea, creatinine, liver enzymes (ALT, AST, ALP), and glycated hemoglobin (HbA1c) values, indicating better overall glycemic control and protection against diabetes-induced organ damage.
    • Histopathological Improvements: Histopathological examination of vital organs like the pancreas, liver, and kidney in treated animals often reveals improvements compared to untreated diabetic controls, suggesting a protective effect against diabetes-related complications.
  • Antioxidant Activity: Some thiazolidine derivatives also exhibit significant antioxidant activity, which is an added benefit for diabetes management, as oxidative stress plays a major role in the pathogenesis and complications of diabetes.
  • Safety Profile: In vivo studies often report good safety profiles with high LD$_{50}$ values and minimal to no observed toxicity at therapeutic doses, as further supported by in vitro cytotoxicity assays on relevant cell lines (e.g., pancreatic cells).

CONCLUSION

The thiazolidine ring system presents a highly versatile and promising scaffold for the development of novel alpha-amylase inhibitors against diabetes. Numerous studies have demonstrated the ability of thiazolidine derivatives to effectively inhibit alpha-amylase activity in vitro and attenuate postprandial hyperglycemia in vivo. The field of thiazolidine-based alpha-amylase inhibitors has witnessed significant progress in recent years, with numerous novel compounds demonstrating promising antidiabetic activities. The combination of rational drug design, efficient synthetic methodologies, and comprehensive biological evaluation has led to the identification of several lead compounds with potential for clinical development.

Key achievements include the development of dual-acting inhibitors that target both alpha-amylase and alpha-glucosidase enzymes, the establishment of clear structure-activity relationships, and the demonstration of in vivo efficacy in animal models. The integration of computational approaches with experimental validation has accelerated the discovery process and provided valuable insights into binding mechanisms.

Future research should focus on addressing remaining challenges such as optimizing selectivity profiles, improving pharmacokinetic properties, and conducting comprehensive safety evaluations. The potential for combination therapies and personalized treatment approaches offers exciting opportunities for advancing diabetes care.

The continued development of thiazolidine-based alpha-amylase inhibitors represents a promising avenue for addressing the growing global burden of diabetes mellitus, with the potential to provide more effective and better-tolerated therapeutic options for patients with type 2 diabetes.

The potential for dual action (alpha-amylase inhibition and PPAR$\gamma$ agonism) further enhances their therapeutic appeal. While challenges remain in optimizing selectivity, pharmacokinetic profiles, and addressing potential toxicities, continued research focusing on rational drug design, diverse synthetic approaches, and rigorous biological evaluation holds significant promise for the discovery of safer and more effective thiazolidine-based alpha-amylase inhibitors to combat the growing global burden of diabetes.

REFERENCES

  1. Ahmed, M. S., Khan, K. M., & Rasheed, M. (2020). Green synthesis and evaluation of thiazolidine-4-one derivatives as potential antidiabetic agents via α-amylase inhibition. Green Chemistry Letters and Reviews, 13(1), 45–54. https://doi.org/10.1080/17518253.2020.1711692
  2. Kim, Y. M., Jeong, Y. K., Wang, M. H., Lee, W. Y., & Rhee, H. I. (2005). Inhibitory effect of pine extract on alpha-glucosidase activity and postprandial hyperglycemia. Nutrition, 21(6), 756–761. https://doi.org/10.1016/j.nut.2004.10.014
  3. Kumar, A., Shukla, P., Srivastava, V. K., & Tripathi, R. P. (2021). Synthesis and α-amylase inhibitory activity of some new thiazolidine-based compounds. Bioorganic & Medicinal Chemistry Letters, 31(3), 127563. https://doi.org/10.1016/j.bmcl.2020.127563
  4. Sharma, P., Singla, R. K., & Dubey, A. K. (2019). Molecular docking and in vitro study of newly synthesized oxadiazole-thiazolidine hybrids as potent α-amylase inhibitors. Journal of Molecular Structure, 1180, 328–336. https://doi.org/10.1016/j.molstruc.2018.12.078
  5. Dheer, D., Singh, V., & Shankar, R. (2017). Medicinal attributes of 2,4-thiazolidinedione and its derivatives: A patent review. Expert Opinion on Therapeutic Patents, 27(1), 63–77. https://doi.org/10.1080/13543776.2017.1233170
  6. Patel, H. M., & Patel, D. V. (2016). Synthesis and biological evaluation of novel thiazolidinone derivatives as potential α-amylase inhibitors. European Journal of Medicinal Chemistry, 124, 500–510. https://doi.org/10.1016/j.ejmech.2016.08.056
  7. Shalini, S., & Sharma, P. K. (2013). Thiazolidinone: A biologically active scaffold. Mini-Reviews in Medicinal Chemistry, 13(6), 870–883. https://doi.org/10.2174/1389557511313060006
  8. Varshney, L., & Singh, R. K. (2014). Synthesis and pharmacological evaluation of some new thiazolidine derivatives as potential antidiabetic agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 29(3), 382–389. https://doi.org/10.3109/14756366.2013.779379
  9. Ali, M. A., & Siddiqui, H. L. (2018). Molecular docking, synthesis and biological evaluation of new thiazolidine derivatives as antidiabetic agents targeting α-amylase. Medicinal Chemistry Research, 27, 1573–1584. https://doi.org/10.1007/s00044-018-2157-0
  10. Ponnuraj, K., & Chitra, S. (2016). Structural and functional insights into the interaction of α-amylase with thiazolidinone derivatives using in silico and in vitro approaches. Journal of Molecular Graphics and Modelling, 68, 1–10. https://doi.org/10.1016/j.jmgm.2016.05.007.

Reference

  1. Ahmed, M. S., Khan, K. M., & Rasheed, M. (2020). Green synthesis and evaluation of thiazolidine-4-one derivatives as potential antidiabetic agents via α-amylase inhibition. Green Chemistry Letters and Reviews, 13(1), 45–54. https://doi.org/10.1080/17518253.2020.1711692
  2. Kim, Y. M., Jeong, Y. K., Wang, M. H., Lee, W. Y., & Rhee, H. I. (2005). Inhibitory effect of pine extract on alpha-glucosidase activity and postprandial hyperglycemia. Nutrition, 21(6), 756–761. https://doi.org/10.1016/j.nut.2004.10.014
  3. Kumar, A., Shukla, P., Srivastava, V. K., & Tripathi, R. P. (2021). Synthesis and α-amylase inhibitory activity of some new thiazolidine-based compounds. Bioorganic & Medicinal Chemistry Letters, 31(3), 127563. https://doi.org/10.1016/j.bmcl.2020.127563
  4. Sharma, P., Singla, R. K., & Dubey, A. K. (2019). Molecular docking and in vitro study of newly synthesized oxadiazole-thiazolidine hybrids as potent α-amylase inhibitors. Journal of Molecular Structure, 1180, 328–336. https://doi.org/10.1016/j.molstruc.2018.12.078
  5. Dheer, D., Singh, V., & Shankar, R. (2017). Medicinal attributes of 2,4-thiazolidinedione and its derivatives: A patent review. Expert Opinion on Therapeutic Patents, 27(1), 63–77. https://doi.org/10.1080/13543776.2017.1233170
  6. Patel, H. M., & Patel, D. V. (2016). Synthesis and biological evaluation of novel thiazolidinone derivatives as potential α-amylase inhibitors. European Journal of Medicinal Chemistry, 124, 500–510. https://doi.org/10.1016/j.ejmech.2016.08.056
  7. Shalini, S., & Sharma, P. K. (2013). Thiazolidinone: A biologically active scaffold. Mini-Reviews in Medicinal Chemistry, 13(6), 870–883. https://doi.org/10.2174/1389557511313060006
  8. Varshney, L., & Singh, R. K. (2014). Synthesis and pharmacological evaluation of some new thiazolidine derivatives as potential antidiabetic agents. Journal of Enzyme Inhibition and Medicinal Chemistry, 29(3), 382–389. https://doi.org/10.3109/14756366.2013.779379
  9. Ali, M. A., & Siddiqui, H. L. (2018). Molecular docking, synthesis and biological evaluation of new thiazolidine derivatives as antidiabetic agents targeting α-amylase. Medicinal Chemistry Research, 27, 1573–1584. https://doi.org/10.1007/s00044-018-2157-0
  10. Ponnuraj, K., & Chitra, S. (2016). Structural and functional insights into the interaction of α-amylase with thiazolidinone derivatives using in silico and in vitro approaches. Journal of Molecular Graphics and Modelling, 68, 1–10. https://doi.org/10.1016/j.jmgm.2016.05.007.

Photo
Devidas Kharad
Corresponding author

SND COLLEGE OF PHARMACY BABHULGAON , YEOLA , DIST NASHIK

Photo
Chavhan Shankar
Co-author

ABASAHEB KAKADE COLLEGE OF PHARMACY BHODHEGAON

Kharad Devidas, Chavhan Shankar, Design, Synthesis and Biological Evaluation of Alpha Amylase Inhibitor Against Diabetes Thiazolidine Ring, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 6, 4174-4183. https://doi.org/10.5281/zenodo.15735851

More related articles
Hibiscus Flowers: A Promising Natural Ally in Canc...
Rajvardhan Ghatage, Tejaswini Khot, ...
Hair Structure and Care: A Review of Herbal Hair C...
Balaji Jadhav, Pratibha Bhalerao, Deep Gulave, Mayuri Jangle, ...
Related Articles
Showing Artificial Intelligence in The Sensing of Breast Cancer...
Aanchal Dahiya, Dr. Nakul Gupta, Dr. Md. Sarfaraz Alam, Kamal Singh Bani, Preeti Bhunia, Aditi Singh...
Design and Assessment of Mucoadhesive Insitu Gel Forming Spray for Oral Delivery...
Pranay Sawant, Tejas Sutar, Sayali Sawant, Susmita Holmukhe, ...