View Article

  • Recent Progress of Antidiabetic Agents for The Treatment of Type 2 Diabetes; A Mini-Review

  • 1,5Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Health Science Campus, Kochi, 682041, India.
    2,4Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India.
    3,5School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, 302017, India.

Abstract

Diabetes mellitus is a group of metabolic disorders that involve hyperglycemia caused by defects in the production of insulin, its action, or both. Diabetes mellitus is primarily divided into type 1 diabetes and type 2 diabetes. Type 1 diabetes is typically treated with insulin therapy and is also known as Insulin-dependent Diabetes mellitus (IDDM), but type 2 diabetes is commonly treated with oral antidiabetic agents. Sulphonylureas, biguanides, insulin sensitizers, glucosidase inhibitors, dipeptidyl peptidase-4 inhibitors (DPP-4), and sodium-glucose co-transporter-2 (SGLT2) inhibitors are the main drugs used to treat type 2 diabetes. The overall work of these drugs is to reduce blood glucose levels by increasing levels of incretin hormones and pancreatic beta cell function. Patients who are unable to accomplish treatment goals with first-line oral antidiabetic agents as monotherapy frequently receive prescriptions for dual medication therapy. In the proposed review article, we have made an effort to study the pathophysiology of diabetes, treatment, herbal drugs, mode of action of drugs, adverse effects as well as current work on antidiabetic agents.

Keywords

Diabetes mellitus, targets, FDA-approved drugs, Herbal drugs, adverse effects.

Introduction

Diabetes, also known as a deadly syndrome, remains all over the life of patients. Diabetes mellitus is referred to as a combination of metabolic syndrome where high blood glucose levels persist before and after meal intake, for a long time [1].  Diabetes mellitus occurs due to damage in β-cells in islets of Langerhans of the pancreas [2]. The food eaten by us has carbohydrates which are transformed into glucose by an enzymatic process in the body. Glucose is essential for the maintenance of energy levels and extra glucose is stored in muscles with the help of insulin. Because of the damage in β-cells, decreases insulin secretion, or the absence of insulin, glucose remains in the blood and is out of reach to muscles for storage causing diabetes Figure 1) [3]. Type-1 and type-2 diabetes have been defined as chronic metabolic disorders that are caused by inadequate production of insulin by the pancreas or due to inefficient insulin consumption in the body [4]. When the body fails to produce enough insulin or develops resistance to it, type-2 diabetes develops. Whereas type-1 diabetes also known as juvenile or insulin-dependent diabetes, is a chronic condition in which the pancreas produces little or no insulin by itself [5,6]. There is a broadly agreed target to halt the rise in type-2 diabetes and obesity by 2025. About 529 million humans worldwide have diabetes, mainly belonging to low-and middle-income countries, and 1.5 million deaths are sprightly attributed to diabetes each year. The number of diabetes cases has been gradually increasing over the past few years. Over the last 10 years, diabetes prevalence has increased steadily in developing countries. In Southeast Asian countries such as India, Bangladesh, Maldives, Mauritius, Srilanka, Nepal, and Countries of Western Pacific regions a maximum of people livelihood with diabetes and they granted almost half the diabetes cases in the world [7,8]. Type-1 diabetes mellitus is characterized by a loss of insulin-producing in the pancreas, leading to insulin deficiency. The condition is normally diagnosed in children and young people, so it is called juvenile diabetes. Genetic factors, environmental factors, immune-mediated attack by T-cells, viral infection, oxidative stress on cells, etc. cause the damage of β-cells. In the deficiency of insulin, glucose remains in the blood and cannot be stored in muscle cells, in this condition body utilizes fat for energy by lipolysis, which releases glycerol (converts to glucose) and free fatty acids (converts to ketone bodies; diabetic ketoacidosis), this situation is responsible for several complications including death. [9-10]. Type-2 diabetes is also called adult-onset, obesity onset, and non-insulin-dependent diabetes mellitus. Type-2 diabetes mellitus occurs due to reduced secretion of insulin and insulin resistance by certain factors. Due to impaired glucose secretion and insulin resistance, glucose remains in the blood and leads to hyperglycemia. The high level of glucose in the blood provokes pancreatic β-cells to release insulin in excess, Consequently, patients with type 2 diabetes frequently produce an excess of insulin. [11-14]. Whereas gestational diabetes occurs due to insufficient insulin secretion and low sensitivity of insulin during pregnancy and disappears after delivery. GDM causes hyperbilirubinemia, which destructs RBC, and in some cases during delivery, perinatal death occurs due to vascular impairment in placental profusion. [15,16]

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-11.png" target="_blank">
            <img alt="Pathophysiology of diabetes mellitus.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-11.png" width="150">
        </a>
Figure 1. Pathophysiology of diabetes mellitus

Symptoms of diabetes may occur rapidly in type 1 diabetes within weeks or months, whereas they occur more slowly or may be absent in the case of type-2 diabetes. The major symptoms of untreated diabetes are loss of weight, polyuria (frequent urination), polyphagia (increased hunger), and polydipsia (increased thirst).  Diabetes mellitus symptoms can be either acute or chronic. The acute symptoms include loss of energy, fatigue, ketoacidosis, and harm to the neurological system Figure 2. The chronic symptoms consist of eye problems, stroke, heart disease due to the thickening of blood components, and chronic kidney failure. [17] The significant reasons or causes for diabetes mellitus include the damage to β-cells of the pancreas by oxidative stress, family history, low levels of physical activity, genetic fingerprints, lifestyle, environmental factors and health-related issues. There is no common cause of diabetes that matches every type of diabetes. The causes associated with diabetes differ based on the person and type of diabetes, so that's why there is no one specific cause. [18-19].

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-10.png" target="_blank">
            <img alt="Symptoms of diabetes mellitus.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-10.png" width="150">
        </a>
Figure 2. Symptoms of diabetes mellitus.

Several targets are there for type-2 diabetes and with the help of these targets, we can regulate glucose metabolism via potent antidiabetic drugs. All these targets are given below in Figure 3. Aldose reductase is an oxidoreductase enzyme that catalyzes the reduction of carbonyls and aldehydes, including monosaccharides. It is well-known that it promotes the conversion of sorbitol from glucose [20-22].  The enzyme human Glucokinase controls the metabolism of carbohydrates by phosphorylation of glucose to glucose-6-phosphate. Uncommon types of diabetes or hypoglycemia may occur via mutations in the human Glucokinase gene. When G-6-P is consumed, a series of reactions are started by an increase in ATP levels, which in turn causes the release of insulin. [23]. Glycogen synthase kinase (GSK) is involved in the metabolism of glucose by stimulating phosphatidyl inositol 3-kinase, which phosphorylates GSK-3 on the nonsupervisory Ser-9 and inactivates GSK-3. This facilitates the insulin-stimulated conflation of glycogen [24-26]. Peroxisome proliferator-activated receptor-γ (PPAR γ), is also known as the glitazone receptor. Two isoforms of PPAR γ are PPAR-γ1 and PPAR-γ 2. The PPAR-γ agonist thiazolidinediones is designed to increase glucose tolerance to improve insulin sensitivity and β-cells function. [27,28]. Adenosine monophosphate-activated protein kinase (AMP Kinase), helps in the activation of glucose transport and fatty acid oxidation caused by these stimuli; It is believed to as a type of master switch controlling glucose and lipid metabolism. The two most popular diabetes medications, metformin and rosiglitazone, partially exhibit their metabolic effects through AMPK activation. This results in improved fatty acid oxidation and decreased synthesis of glucose, cholesterol, and triglycerides in the liver. [29]. Cannabinoid Receptor is now becoming obvious that insulin activity in insulin-sensitive tissues is influenced by cannabinoid 1 receptor (CB1R) agonism/antagonism. [30]. Adapting the number of insulin-secreting β-cells in the islets of Langerhans is necessary for its significant role in glucose homeostasis. Insulin also positively controls β-cell proliferation in an autocrine manner via the insulin receptor signalling pathway. Glycogen Phosphorylase is known as the phosphorylase enzyme. It releases glucose-1-phosphate from the terminal α-1, a 4-glycosidic bond, which catalyzes the rate-limiting step in glycogenolysis in mammals [31]. Protein Tyrosine Phosphatases (PTPs), an enzyme present in tissues sensitive to insulin, are one of the possible possibilities for controlling the insulin signalling pathway [32,33]. Free fatty acid receptor-1 has been identified as a better antidiabetic target because it plays a vital role in regulating insulin secretion in β-cells of the pancreas this receptor works by interaction with the G protein alpha subunit, Gg family. Further, it activates Phospholipase-C and ultimately manages glucose concentration in the blood. [34,35]. G protein-coupled receptors (GPCR) like GPR119, GPR142, and GPR120 are identified as important targets in diabetes mellitus. The gastrointestinal tract's enteroendocrine cells and pancreatic β-cells are one of the main sites of GPR119 expression. Through Gα coupling, activation of the GPR119 receptor raises intracellular cyclic AMP levels and regulates adenylate cyclase. Consequently, this stimulates the pancreatic β-cells to release insulin. The enteroendocrine cells' expression of the GPR119 receptor stimulates the release of incretins including glucagon-like peptide-1 (GLP-1) and Glucose-dependent insulinotropic polypeptide (GIP) [36]. The glucagon receptor (GCGr) is a peptide chain of 29 amino acids, it plays a major function in converting glycogen to glucose in the liver by blocking hepatic glycogen synthesis, promoting glycogenolysis, and boosting gluconeogenesis. This receptor found in the pancreatic islets (α-cells) opposes insulin secretion from β-cells [37]. DPP-4 enzyme, also known as adenosine deaminase complexing protein, is encoded by the DPP-4 gene in humans. It is essential for the metabolism of glucose. It is responsible for degradation of incretins like GIP and GLP-1. The inhibition of DPP-4 increases the action of incretins, GIP and GLP which ultimately increases insulin production. Further, it promotes the management of type 2 diabetes [38-39].

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-9.png" target="_blank">
            <img alt="Targets for diabetes mellitus.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-9.png" width="150">
        </a>
Figure 3. Targets for diabetes mellitus

α-glucosidase & α-amylase enzyme helps the small intestine to absorb complex carbohydrates, oligosaccharides, and disaccharides by breaking them down into monosaccharides.  Competitive and reversible inhibitors of intestinal α-glucosidases are known as α-glucosidase inhibitors [40]. The several enzymes present in the body regulate the level of both glucose and insulin, e.g. α-glucosidase, aldose reductase, glycogen phosphorylase, glucokinase, peroxisome proliferator receptor agonist, AMP Kinase, histone deacetylases, glycogen synthase kinase, PTP-1B and DPP-4 as we discussed above. The secretion of insulin is controlled and produced by incretins; GLP-1 and GIP while the DPP-4 enzyme deactivates GLP-1 and GIP, which offers an opportunity to develop new DPP-4 inhibitors with improved pharmacodynamics and pharmacokinetic properties. During the last 25 years, several orally active drugs have been discovered for the treatment of diabetic patients including, biguanides (metformin), sulfonylureas (tolbutamide), meglitinides, glucosidase inhibitors (miglitol), thiazolidinediones (pioglitazone), SGLT-2 inhibitors (canagliflozin), and recently DPP-4 inhibitors (sitagliptin). The currently marketed antidiabetic medications are potent and widely used for the treatment of diabetes-related illnesses but long-term treatment is difficult due to adverse reactions such as sulfonylureas causing hypoglycaemia and weight gain and biguanides results in lactic acidosis with impaired liver/ kidney functions. α-glucosidase inhibitors include diarrhoea, gas, and stomach pain. The most common adverse effect of thiazolidinediones is edema. Furthermore, a new investigation has linked the drug pioglitazone to an increased risk of bladder cancer. DPP-4 inhibitors, reported to cause upper respiratory tract infection, urinary tract infection, nasopharyngitis, blistering, hypersensitivity reactions, pancreatitis, headache, arthralgias, sore throat, fatigue, dizziness, edema, nausea, and diarrhoea. SGLT2 inhibitors are reported to genitourinary infections, volume depletion, ketoacidosis, amputations, and acute kidney injury. The side effects of GIP and GLP-1 are vomiting, diarrhoea, dizziness, headache, injection site reactions, and constipation. The common side effects of amylin analogues are nausea, vomiting, headache, and hypoglycaemia when taken along with insulin [41-42].

Treatment of Type 2 diabetes

Antidiabetic medications treat type 2 diabetes by decreasing glucose levels within the blood. [43-51] There are many types of anti-diabetic drugs, and the choice of one depends on the patient's age, condition, and type of diabetes, among other factors. These drugs act by different mode of action as shown in Figure 4. Over the past 20 years, several orally active drugs have been found to help type 2 diabetes patients. These are included in Table 2, and all FDA-approved antidiabetic drugs are in Table 1.

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-8.png" target="_blank">
            <img alt="Site of action for oral antidiabetic medications.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-8.png" width="150">
        </a>
Figure 4. Site of action for oral antidiabetic medications.

Table 1. List of FDA approved antidiabetic drugs

Entry

Drug and Generic name

Class of drug

IUPAC Name

Pharma Company

Year Approved

1

Rosiglitazone / Avandia

TZD

Insulin sensitizer

Woodward Pharma

5th May 1999

2

Pioglitazone / Actos

TZD

Insulin sensitizer

Takeda Pharmaceuticals USA

15th July 1999

3

Acarbose / Precose

α- glucosidase inhibitor

Prevent the digestion of carbohydrates, improve glycemic control

Bayer HealthCare Pharmaceuticals Inc.

9th June 1995

 

4

Miglitol / Glyset

α- glucosidase inhibitor

Prevent the digestion of carbohydrates, improve glycemic control

Pfizer

18th December 1996

5

Glyburide / Glynase

 

Micronase

Sulphonyl Urea

Stimulate insulin secretion

Pfizer

Pfizer

4th March 1992

1st May 1984

6

Glipizide / Glucotrol

Sulphonyl Urea

Stimulate insulin secretion

Pfizer

1984

7

Glimepiride / Amaryl

Sulphonyl Urea

Stimulate insulin secretion

SANOFI AVENTIS US

30th November 1995

8

Metformin / Glucophage

Biguanide

Inhibit gluconeogenesis, insulin sensitizer, pleotropic effects

EMD SERONO INC

3rd March 1995

9

Pramlintide / Symlin

Amylin analogue

Short acting

AstraZeneca AB

16th March 2005

10

Exenetide / Byetta

GLP1R agonist

Increase insulin secretion and inhibit glucagon secretion from pancreatic islet cells

AstraZeneca AB

28th April 2005

11

Liraglutide/ Victoza

GLP1R agonist

Increase insulin secretion and inhibit glucagon secretion from pancreatic islet cells

NOVO NORDISK INC

25th January 2010

12

Dulaglutide/ Trulicity

GLP1R agonist

Increase insulin secretion and inhibit glucagon secretion from pancreatic islet cells

ELI LILLY AND CO

18th September 2014

13

Albiglutide / Tanzeum

GLP1R agonist

Increase insulin secretion and inhibit glucagon secretion from pancreatic islet cells

GLAXOSMITHKLINE LLC

15th April 2014

14

Lixisenatide/ Adlyxin

GLP1R agonist

Increase insulin secretion and inhibit glucagon secretion from pancreatic islet cells

SANOFI-AVENTIS US

27th July 2016

15

Semaglutide / Ozempic

GLP1R agonist

Increase insulin secretion and inhibit glucagon secretion from pancreatic islet cells

NOVO

5th May 2017

16

Nateglinide / Starlix

Meglitinides

Stimulates insulin secretion

NOVARTIS

22nd December 2000

17

Repaglinide / Prandin

Meglitinides

Stimulates insulin secretion

GEMINI LABS LLC

22nd December 1997

18

Colesevelam / Welchol

Bile acid sequesterants

Clearance of LDL cholestrerol

Cosette

26th May 2000

19

Bromocryptine/ Cycloset

Dopamine receptor agonist

Stimulate hypothalamic dopamine D2 receptors, improve glycemic control.

VEROSCIENCE

5th May 2009

20

Canagliflozin / Invokana

SGLT2

Inhibitors

Block glucose absorption in the kidney

JANSSEN PHARMS

29th March 2013

21

Dapagliflozin / Farxiga

SGLT2

Inhibitors

Block glucose absorption in the kidney

AstraZeneca AB

8th January 2014

22

Empagliflozin / Jardiance

SGLT2

Inhibitors

Block glucose absorption in the kidney

BOEHRINGER INGELHEIM

1st August 2014

23

Ertugliflozin / Steglatro

SGLT2

Inhibitors

Block glucose absorption in the kidney

MSD SUB MERCK

19th December 2017

24

Sitagliptin / Januvia

DPP-4

Inhibitors

Inhibit glucagon release and increase insulin secretion

MERCK SHARP DOHME

16th October 2006

25

Saxagliptin / Onglyza

DPP-4

Inhibitors

Inhibit glucagon release and increase insulin secretion

AstraZeneca AB

31st July 2009

26

Linagliptin / Trajenta

DPP-4

Inhibitors

Inhibit glucagon release and increase insulin secretion

BOEHRINGER INGELHEIM

2nd May 2011

27

Alogliptin / Nesina

DPP-4

Inhibitors

Inhibit glucagon release and increase insulin secretion

TAKEDA PHARMS USA

25th January 2013

Sulfonylureas: These are the first class of oral hypoglycaemic drugs for the management of type-2 diabetes. They are additionally referred to as insulin secretagogues. sulfonylureas bind to the receptors found present in pancreatic β-cells. As a result, the β-cell membrane becomes depolarized and ATP-sensitive K+ channels close. Subsequently, it causes voltage-gated calcium channels to open, resulting in pancreatic β-cells to produce more insulin [52]. Examples of sulphonylureas are Chlorpropamide, Tolbutamide, Acetohexamide, glibenclamide, glipizide, glyburide, glimepiride

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-7.png" target="_blank">
            <img alt="Biguanides.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-7.png" width="150">
        </a>
Biguanides: They reduce blood glucose levels by lowering the amount of glucose that in liver produces and then releases into your bloodstream. Additionally, they help with lowering blood glucose levels by increasing the skeletal muscle tissue's tolerance to insulin, helping it to better absorb glucose for energy. Insulin resistance is decreased and insulin sensitivity is raised as a result. As they improve insulin resistance, they're also known as insulin sensitizers. Metformin, which is formed from 2-cyanoguanidine and dimethyl ammonium chloride, contains two methyl substituents in position 1. The phenethyl biguanide derivative of metformin known as Phenformin is described by the replacement of one terminal nitrogen atom with a 2-phenylethyl group. E.g. Phenformin, Metformin, Butformin [53].

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-6.png" target="_blank">
            <img alt="Meglitinides.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-6.png" width="150">
        </a>

Meglitinides: It assists the pancreas in producing insulin. They bind at different locations, but they have the same effect on potassium channels as sulfonylureas. They bind directly to the pancreatic β-cells’ receptors. It depolarizes the β-cell membrane and causes ATP-sensitive K+ channels to close. After that, it causes voltage-gated Ca+2 channels to open, which increases insulin secretion from β-cells. We call these compounds insulin secretagogues. E.g. Repaglinide, Nateglinide [54].

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-5.png" target="_blank">
            <img alt="Thiazolidinediones.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-5.png" width="150">
        </a>
Thiazolidinediones: They are also known as PPAR-γ agonists because they bind to the receptor and activate peroxisome proliferator activated receptor-γ (PPAR-γ). To improve glucose uptake, they overexpress GLUT-1 and GLUT-4 receptors on their cell surface. They also decrease hepatic gluconeogenesis and insulin resistance. They reduce the HbA1c level as well as the postprandial glucose level [55]. E.g. Pioglitazone, rosiglitazone are two drugs approved by the FDA as monotherapy or combined with metformin or sulfonylureas for the treatment of type 2 diabetes.

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-4.png" target="_blank">
            <img alt="Glucosidase inhibitors.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-4.png" width="150">
        </a>
Glucosidase inhibitors: They inhibit the α-glucosidase enzyme present in the intestinal brush border epithelium, and also delaying the absorption and metabolism of carbohydrates. The enzyme α-glucosidase facilitates the breakdown of poly- and oligosaccharides into monosaccharides and helps in their digestion. Among this drug class's most common side effects are loose motion and flatulence. These medications have an anti-hyperglycaemic action; hypoglycemia is not produced by them [56, 57]. E.g. Acarbose, Miglitol, Voglibose.

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-3.png" target="_blank">
            <img alt="DPP-4 inhibitors.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-3.png" width="150">
        </a>
DPP-4 inhibitors

DPP-4 inhibitors are an effective treatment for type-2 diabetes because they inhibit the DPP-4 enzyme. The DPP-4 enzyme is inhibited, which increases and prolongs the activity of incretins, which are crucial for regulating blood glucose levels and secreting insulin. The small intestine releases the incretin hormones glucose-dependent gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) into the vasculature during a meal. In a glucose-dependent way, the hormones control insulin stashing. GLP-1 plays a variety of functions within the human body, including promoting the production of insulin, preventing the release of glucagon, delaying the emptying of the stomach, decreasing hunger, and promoting the regeneration of pancreatic β-cells. The DPP-4 enzyme is responsible for the metabolism of glucose as shown in Figure 5. Inhibition of the DPP-4 enzyme leads to potentiation of incretin hormone GIP and GLP- 1 and hence improves the treatment of type- 2 diabetes [58-61]. Examples are sitagliptin, vildagliptin, alogliptin, saxagliptin, linagliptin, denagliptin, trelagliptin.

<a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-2.png" target="_blank">
            <img alt="FIG-8.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-2.png" width="150">
        </a>
<a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-1.png" target="_blank">
            <img alt="DPP-4 inhibitors mode of action.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-1.png" width="150">
        </a>
Figure 5. DPP-4 inhibitors mode of action.

SGLT-2 inhibitors: SGLT-2 inhibitors acting on the SGLT-2 proteins found in the renal proximal convoluted tubules of the kidney. It functions by preventing the tubular lumen from reabsorbing filtered glucose. The proximal convoluted tubules of the kidneys express SGLT-2 proteins, which carry out their physiological role via reabsorbing filtered glucose at the tubular lumen. Each of the four SGLT-2 inhibitors lowers the renal threshold for glucose (RTG), increases urine glucose excretion, and decreases the reabsorption of filtered glucose [62, 63]. Examples of SGLT-2 inhibitors are canagliflozin, dapagliflozin, and empagliflozin and ertugliflozin has been approved by the FDA for the treatment of type 2 diabetes mellitus. Dapagliflozin is recently approved by FDA for the treatment of heart failure across the full spectrum of left-ventricular ejection fraction (LVEF).

        <a href="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-0.png" target="_blank">
            <img alt="GLP-1 receptor agonist.png" height="150" src="https://www.ijpsjournal.com/uploads/createUrl/createUrl-20250511132344-0.png" width="150">
        </a>
GLP-1 receptor agonist: The glucose-dependent and glucagon-like peptide incretins, or peptides originating from the gut, are also referred to as insulinotropic polypeptides. It is a group of endogenous metabolic hormones known as incretins, they reduce blood glucose levels. GLP-1 agonists are the new group of injectable for treatment of type 2 diabetes mellitus. Although GLP-1 has an alanine residue at the N terminus, DPP-4 is capable of breaking down it. Thus, other amino acids such as threonine, serine, and glycine were substituted for the alanine group to derive the novel GLP-1 analogues. In vitro, such derivatives exhibited greater stability than DPP-4. Not only, certain GLP-1 derivatives are twice as effective as GLP-1, but they were also stable. The first GLP-1 agonist medication to be released as an anti-diabetic was Exenatide. Due to the medication's fast clearance, it was given twice daily. Tirzepatide is the first dual GIP/GLP-1 receptor agonist approved for the treatment of type 2 diabetes [64-66].

Amylins: It is made up of a 37 amino acids single chain. Amylin is secreted by the pancreatic β cells with insulin. It preserves both fasting and the blood postprandial glucose level by delaying the stomach emptying and inhibiting the release of glucagon. By controlling the brain's appetite center, it regulates the amount of food consumed [67]. Amylins work by delaying the emptying of the stomach, preventing the release of glucagon following a meal, regulating the appetite center, and inhibiting food intake and weight gain. Amylins are offered in parenteral form used for the management of diabetes mellitus. The only drug in this class that is now available is Pramlintide acetate, (Symlin) and it is administered subcutaneously [68].

Table 2 Common targets for the management of type-2 diabetes mellitus.

Oral antidiabetic drugs

Mechanism

Agents

Advantages

Disadvantages

Biguanides

Decrease hepatic gluconeogenesis

Metformin

No hypoglycaemia, weight neutral

GI disturbance, lactic acidosis

Sulphonyl ureas

Stimulate insulin secretion

Glimepiride

Inexpensive

Hypoglycaemia, weight gain

Meglitinides

Stimulate insulin secretion

Repaglinide

The short onset of action, low postprandial glucose

Hypoglycaemia,

Weight gain

Glucosidase inhibitors

Decrease glucose absorption

Acarbose, Voglibose

Reduce postprandial glucose

flatulence

Thiazolidinediones

Improve insulin resistance

Pioglitazone

Lower insulin requirement

Edema, CHF, weight gain, fracture, macula edema

DPP-4 inhibitors

Prolong GLP-1 action

Sitagliptin

No hypoglycaemia

Weight neutral

Less clinical experience

Amylin

Improve glycaemic control, prevention of postprandial rise in glucagon levels

Pramlintide

suppresses release of glucagon

Nausea, loss of appetite, stomach cramps

GLP-1 agonist

Mimic the effect of incretin hormone

Exenatide, Liraglutide

No hypoglycaemia

 

Nausea, diarrhoea, vomiting

SGLT2 inhibitors

Helps eliminate glucose in the urine

Canagliflozin

Dapagliflozin

No hypoglycaemia

Decrease heart failure

Genital and urinary infections, frequent urination

Recent update on antidiabetic agents

Here we added recent work on all antidiabetic agents like sulfonylureas, DPP-4 inhibitors, SGLT2 inhibitors, thiazolidinedione’s, α-glucosidase inhibitors, GLP agonists, PTP1B inhibitory etc. after the review work, we found that there is no recent work on antidiabetic class like biguanides, meglitinides and sulphonylureas and recent work on only few classes like GLP agonists, α-glucosidase, thiazolidinedione, and DPP-4 inhibitors. Tiziano et. al., in their mini-review, explored various methods that have appeared in the literature to synthesize sulfonylureas placing significance on the reaction mechanisms of vital synthetic steps [69]. Sroor et. al., synthesized sulfonylurea derivatives having alkyl substituents such as isopropyl or n-butyl and also evaluated the anti-diabetic activities of compounds 4, 5, 7, and 10. Ghadeer et. al., in this study, evaluated the insulin signalling pathway via L6 skeletal muscle cell in vitro and in vivo study of glycosylated sulfonylureas [70]. Temel et al., synthesized 2-pyrazoline analogues and investigated DPP-4 inhibitory activity and cytotoxicity MTT assay on the L929 mouse fibroblast cell line [71]. Nidhar et. al., reported ourself, the design, synthesis, and evaluation of some pyrazole triazole persulfonimide and aryl triazole persulfonimide as potent antidiabetic agents [72]. Gupta et. al. (2023), synthesized new imeglimin-inspired 1,3,5-triazine derivatives, tested against DPP enzymes and revealed the selective and potent DPP-4 inhibitor activity of compound 8c by in-vivo antidiabetic activity test in Wistar rats. They also performed docking studies [73]. Guang-Jing Feng et. al., designed and synthesized novel thioglucoside gliflozins as potent gliflozin drugs and they were biologically Evaluated, where they studied two series of thioglucoside gliflozins, First series substitution of S-atoms at meta positions and another with substitution of S-atom at ortho position. These drugs showed greater stability and inhibitory activity against SGLT2 inhibitors [74]. Masahiko Seki et. al. synthesized dapagliflozin and canagliflozin as advanced SGLT2 inhibitors for the treatment of diabetes [75]. Yoshiaki Kitamura et. al., synthesized carbasugar analogues as SGLT2 inhibitors via click reaction Cu catalysed azide-alkyne cycloaddition, and compounds 7b and 7c were found as potent SGLT2 inhibitors [7]. CenJun et. al., worked on synthesized, physicochemical properties, crystal studies, and DFT calculation of empagliflozin. [77] Najmi, et. al., reported the design and synthesis of 5 novel benzylidene-thiazolidine-2,4-diones (5a–e) as PPAR-γ agonists and they were characterized using analytical methods. As revealed by the in vivo antidiabetic study, the compounds 5d and 5e have comparable activity with standard rosiglitazone. It is found to have no significant impact on body mass after 21 days of animal administration [78]. Amin et. al., synthesized novel pyrimidine thiazolidinedione derivatives, characterized them using spectroscopic analytical techniques and evaluated their anti-diabetic property. The best compounds 6e and 6m were evaluated for activity in STZ-induced rats and neither show an increase in body weight nor any toxicity [79]. Srinivasa et. al., synthesized and characterized a group of new 3-((5-phenyl-1,3,4-oxadiazol-2-yl) methyl) thiazolidine-2,5-dione derivatives (5a–5j) based on the prediction that thiazolidinediones and 1,3,4-oxadiazoles could improve the inhibition of α-amylase and α-glucosidase. They found that compounds 5a, 5b, and 5j could be a lead for the development of a new class of antidiabetic agents that might be potent against both enzymes [80].  Huneif et. al., designed and created five novel succinimide-thiazolidinedione hybrid derivatives (10a–e) in a multistep reaction. Following in vitro tests on multiple targets, the compound 10d was found to be the most active antidiabetic agent. Its activity is further investigated in experimental animals and a molecular docking study was also performed for compound 10d [81]. Shakour et. al., designed and synthesized new series of imidazolyl-methyl- l-2,4-thiazolidinediones 9a-9m, and carried out in silico studies. The studies revealed that compounds 9e and 9b are more active compared to pioglitazone and compound 9e was weight neutral or had no major side effects following its administration [82]. Singh et. al., synthesized and evaluated the α-amylase and α-glucosidase inhibition of nineteen thiazolidine-2,4-diones (TZDs)-hybrids. From the results, it was clear that the best dual inhibitors were the compounds 7i, 7k, and 7p and the derivatives were found to be good anti-oxidants. They are non-toxic and possess good GI absorption. The most effective molecule, according to the in vivo study, was 7p, whose activity was lower than that of the standard drug pioglitazone [83].  Mengyue et. al. synthesized a set of thiazolidine-2,4-dione derivatives (C1-C36) and all compounds demonstrated better α-glucosidase inhibition in comparison with standard drug acarbose. The reversible mixed-type inhibitor C23 was found to be the most active out of the compounds synthesized and did not possess cytotoxicity against LO2 and 293 cells. It showed binding interaction with α-glucosidase in the molecular docking study and reduced the glucose level in mice on oral administration [84]. Ghomi et. al., synthesized and spectroscopically analysed a group of twenty novel quinoline-linked benzothiazole hybrids (8a–t) as potential α-glucosidase inhibitors and the compounds 8b, 8h, 8n, and 8o demonstrated better α-glucosidase inhibitory activity compared with standard acarbose. A non-competitive inhibition was shown by the most active molecule (8h) on enzyme kinetic studies. The key interactions of the most active compound 8h are further identified by homology modelling, molecular docking, and molecular dynamics simulation studies, and the drug ability of the novel derivatives are predicted [85].  Ali et. al., designed, synthesized, and the structure confirmed by spectroscopic analysis, of a new class of 1,3,4-thiadiazole containing Schiff base analogues (1–12) and compounds 4, 8, and 9 exhibited excellent inhibition of α-glucosidase enzymes by using standard acarbose. Molecular docking, pharmacokinetics, cytotoxic evaluation, and density functional theory study were also conducted [86].   Yousefnejad et. al., synthesized and evaluated benzo[d]imidazole-amide containing 1,2,3-triazole-N-arylacetamide derivatives 8a–n, and the N-2-methylphenylacetamide derivative 8c was found to be the most promising inhibitor of α-glucosidase. As revealed by the kinetic study compound 8c is a competitive inhibitor and the ADMET study predicted the oral activity of compound 8c. Furthermore, the compounds 8c, 8e, and 8g did not show cytotoxic effects against the cancer and normal cell lines [87].  Emadi et. al., designed a new group of phthalimide-phenoxy-1,2,3-triazole-N-phenyl (or benzyl) acetamides (11a-11n) and synthesized them. They were screened for in-vitro inhibitory activity against the α-glucosidase enzyme. Compounds 11j and 11i were most active with positive control acarbose. Kinetic analysis, molecular docking, molecular dynamics, and in silico pharmacokinetic study were also performed [88]. Tariq et. al., through facile chemical reactions, synthesized, and characterized coumarin–hydrazone hybrids (7a-7i), and the in vitro α-glucosidase inhibitory activity of the synthesized compounds were carried out and compound 7c was found to be the most potent α-glucosidase inhibitor among the synthesized compounds [89] Dong et. al. (2024), designed and synthesized a novel better acting long-acting dual GLP-1/GIP RA that compared to standard tirzepatide which exhibited beneficial impact on blood sugar and body weight when evaluated in diet-induced and db/db obese (DIO) mice [90] Zhang et. al. (2023), designed, and synthesized by liquid-phase synthesis, Sixteen GLP-1 receptor agonists (13-28) with dual fatty acid side chains and they were biologically evaluated. Their structures were confirmed and evaluated for receptor affinity, activity, and plasma stability. It was discovered that conjugate 19 was superior to Semaglutide [91]. Ehsasatvatan and Kohnehrouz (2023) designed and computationally analysed chimeric long-lasting GLP-1 receptor agonists including stability, and solubility and they predicted the secondary and tertiary structures and validated them. The structures were stable throughout the molecular dynamic simulation studies [92]. Girdhar et. al. (2022), Designed, synthesized, and biologically evaluated a set of small molecule oral glucagon-like-peptide-1 receptor agonists. They found that the small molecule PK2 (6-((1-(4-nitrobenzyl)-1H-1,2,3-triazol-4-yl) methyl)-6H-indolo[2,3-b] quinoxaline) which showed solid binding with GLP1R ectodomain is orally active nonpeptidic able to maintain or replenish viable β-cell mass. [93]. Thareja et. al. (2023), designed and synthesized novel biphenyl thiazolidinedione conjugates (8a-n), and their in-vitro PTP1B inhibitory potency and in-vivo anti-hyperglycaemic efficacy were assessed with Suramin and Pioglitazone as standard respectively. Additionally, the effect on weight, pancreatic safety, and in-silico ADMET investigations of drug-likeness were also performed. The conjugate 8j showed the best result in which all the molecules 8a-n exhibited good activity [94]. Sroor et. al. (2022) designed, synthesized, characterized, and evaluated novel series of sulfonylurea analogues (3–11) and most of the analogues showed significant In-vivo anti-diabetic activity. The most active (compared with Diamicron) analogues 4, 5, and 10 were further evaluated for liver enzyme activities, antioxidant, and oxidative stress biomarkers, and a histological examination was also carried out. As it is clear from SAR, the anti-diabetic property was increased by the alkyl substituents such as isopropyl or n-butyl [95]. Shah et. al. (2023) based on their previous work, optimized, synthesized, and evaluated new potent multitarget antidiabetic compounds 47–49 and 55–57 with central 5-benzylidine thiazolidine-2,4-dione moiety as inhibitors of α-amylase, α-glucosidase, PTP-1B and DPP-4. It resulted in compounds with an in-vitro potency that is many times higher than the lead (Z)-5-(4-hydroxy-3-methoxybenzylidene)-3-(2-morpholinoacetyl) thiazolidine-2,4-dione (Z-HMMTD) in which compound 56 showed remarkable results as the glucose-uptake promoter [96-98].

Recent update on Herbal antidiabetic agents

Hibiscus sabdariffa: Hibiscus sabdariffa also known as roselle or red sorrel, is a flowering shrub. The study investigates the possible use of Roselle Extracts to manage diabetes mellitus. Previous studies have demonstrated the extracts' effects on blood glucose levels and insulin sensitivity in diabetic mice. The study also discovers chemicals in Roselle Calyces that could be used to treat type 2 diabetes by inhibiting Phosphoenolpyruvate carboxykinase (PEPCK). The extracts block pancreatic enzymes and modulate gene expression related to glucose regulation, hence improving insulin sensitivity. [99]

Catharanthus roseus: A systematic evaluation of research papers on the antihyperglycemic activities of Catharanthus roseus Linn., a plant used in Ayurveda to treat diabetes, discovered that the herb appeared to reduce blood sugar levels in test subjects. This possible antidiabetic advantage could be attributed to the plant's ability to stimulate insulin secretion from damaged beta cells in response to glucose and boost the activity of genes involved in sugar uptake (GLUT-2 and GLUT-4).  [100]

Ficus racemose: Ficus racemosa fruit methanolic extract, a medicinal plant used to treat various diseases. The presence of secondary metabolites such as bioflavonoids, glycosides, alkaloids, tannins in the crude methanolic extract may contribute to its antidiabetic and antimicrobial properties. Ayurvedic medicine extensively uses the herb F. racemosa Linn. (Moraceae) to treat various ailments, including kidney stones, biliary disorders, jaundice, diarrhoea, inflammatory conditions, liver disorders, haemorrhoids, respiratory and urinary diseases. [101]

Coriandrum sativum: Coriander contains a variety of phytochemicals, including polyphenols, reducing sugars, terpenoids, carotenoids, glycosides, sterols, isocoumarins, flavonoids, tannins, alkaloids, fatty acids, and coumarins. Consuming coriander has been shown to be an effective strategy of keeping blood glucose levels stable in pre-diabetic patients. A study of 10 individuals with type 2 diabetes found that coriander seed consumption reduced fasting blood sugar, serum insulin, and Homeostasis Model Assessment of Insulin Resistance. Coriander oil contains antioxidant properties and helps treat renal and pancreatic pathological problems caused by diabetes. Phenols and flavonoids have antioxidant properties that reduce oxidative damage and boost glucose metabolism. It improves α amylase enzyme inhibition, which contributes to their anti-diabetic properties. Linalool and petroselinic acid have been linked to anti-inflammatory qualities that aid in blood sugar regulation. [102]

Cinnamomum verum: The anti-diabetic properties of an ethanolic extract of C. verum bark utilising in vitro and in silico models. C. verum inhibits carbohydrate digestion enzymes α-glucosidase and α-amylase, and affects glucose uptake in 3T3 adipocytes. HPLC-quantified molecules were docked with the GLUT 4 glucose transporter and PPAR-γ. It inhibits α-glucosidase and α-amylase enzymes and promotes adipocyte glucose uptake. [103]

Azadirachta indica: Azadirachta indica, a medicinal plant used in Ayurveda to treat diabetes, was investigated for its effects on insulin signal transduction and glucose homeostasis in high-fat, fructose-induced type 2 diabetic male rats. According to the study, the extract restored normal levels of blood glucose, serum insulin, lipid profile, insulin signalling molecules, glycogen concentration, and glucose oxidation in skeletal muscle. This shows that Azadirachta indica might help manage type 2 diabetes by enhancing insulin signalling molecules and glucose utilization in the skeletal muscle. [104]

Trigonella foenum-graecum: Fenugreek, a popular Asian dietary herb, has been traditionally used for diabetes treatment due to its hyperglycaemic-regulating components. Fenugreek's antidiabetic impact was linked to increased glucose transporter type-4 activity, lowered glucose-6-phosphatase and fructose-1,6 bisphosphatase activities, inhibited α-amylase and maltase activities, protected β cells, and raised insulin release. Further clinical studies revealed better blood glucose levels and lipid profiles. [105]

Caesalpinia bonducella: The study found that phytochemicals from Caesalpinia bonducella seeds have antioxidant and anti-diabetic activities. The seed extract was found to inhibit the enzyme responsible for glucose metabolism, with maximum inhibition at 0.25 mg/mL, and the chemical was discovered to be highly reactive to free radicals. In the DPPH assay, the plant seed extract had the highest inhibitory activity of 92.04% at high concentrations. Additionally, the plant seed extract inhibited H2O2 scavenging by 88.3% at a dosage of 0.75 g/mL. These findings suggest therapeutic benefits for diabetes and oxidative stress disorders. [106]

Gynura procumbens: The GLUT-4 expression levels in Alloxan-induced diabetic rats were compared with untreated healthy rats and rats treated with different doses of G. procumbens extract using enzyme-linked immunosorbent assay (ELISA) specified for rat GLUT-4. The researchers found that all test groups, including the metformin control group, showed elevated GLUT-4 expression in liver tissue compared to the diabetic control group. Result shows that the ethanolic extract of G. procumbens facilitated serum-glucose modulatory antidiabetic activity by upregulating GLUT-4 expression and increasing glucose uptake. [107] G. procumbens was found to have a higher hypoglycaemic impact when combined with Azadirachta indica or Andrographis paniculata. [108-109] Add herbal drug table

CONCLUSION

In this article, we have included an overview of diabetes, and the management of type-2 diabetes. After a careful survey of target enzymes, antidiabetic drugs, herbal drugs and their mode of action, we can conclude that there are many antidiabetic drugs available in the market to manage diabetic conditions but the following are showing efficacy Metformin, Glipizide, Glimepiride, Invokana, Jardiance, Pioglitazone, Januvia, toward lowering blood sugar level but long-term treatment is limited due to their side effects. The ultimate role of these drugs is to reduce the blood sugar level by increasing the level of insulin and decreasing the level of glucagon in pancreatic beta cells. After the review, we found that available drugs have good potency but need to be focused on to reduce adverse effects, and we can also use these drugs for the long term. Currently, it is imperative to develop more effective antidiabetic medications to counteract the adverse effects associated with antidiabetic treatments.

ACKNOWLEDGEMENTS

Manisha Nidhar, Ashish Kumar Tewari and authors would like to express their gratitude to Banaras Hindu University's and Amrita Vishwa Vidyapeetham, Amrita School of Pharmacy, Kochi, Kerala for Financial support.

REFERENCES

  1. American diabetes association. Diagnosis and classification of diabetes mellitus. Diabetes care, 2009;32:62-67.
  2. Goyal R, Jialal I. Diabetes Mellitus Type 2. Book StatPearls Treasure Island, 2021.
  3. Westman Eric C. Type 2 Diabetes Mellitus: A Pathophysiologic Perspective, Frontiers in Nutrition, 2021, 8.
  4. https://www.who.int/health-topics/diabetes#tab=tab_1
  5. Reusch, J.E. Manson. Management of type 2 diabetes in 2017: Getting to goal, Jama. 2017; 317:1015-1016.
  6. Sebokova, Elena Christ, Andreas Boehringer, Markus Mizrahi, Jacques. Dipeptidyl peptidase IV inhibitors: the next generation of new promising therapies for the management of type 2 diabetes, Curr. Topics in Medi. Chem. 2007, 7, 547-555.
  7. Sapra A, Bhandari P. Diabetes Mellitus. StatPearls, Treasure Island (FL) 2022.
  8. Chaudhury Arun, Duvoor Chitharanjan, Reddy Dendi Vijaya Sena, Kraleti Shashank, Chada Aditya, Ravilla Rahul, Marco Asween, Shekhawat Nawal Singh, Clinical Review of Antidiabetic Drugs: Implications for Type 2 Diabetes Mellitus Management, Frontiers in Endocrinology, 2017, 8, 6.
  9. Yan Zheng, Sylvia H. Ley & Frank B. Hu Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nature Reviews Endocrinology, 2018;14:88–98
  10. Banday MZ, Sameer AS, Nissar S. Pathophysiology of diabetes: An overview. Avicenna J Med. 2020 13;10(4):174-188.
  11. Jahangir Moini, Chapter 3 - Pathophysiology of Diabetes, Editor(s): Jahangir Moini, Epidemiology of Diabetes, Elsevier, 2019; 25-43,
  12. Lucier J, Weinstock RS. Diabetes Mellitus Type 1. In: StatPearls. Treasure Island 2021.
  13.  Global Burden of Disease Collaborative Network. Global Burden of Disease Study 2019. Results. Institute for Health Metrics and Evaluation. 2020.
  14. Wetsman EC type 2 diabetes mellitus; Pathophysiologic perspective, front. Nuti. 2021; 8:707371.
  15. Alfadhli EM. Gestational diabetes mellitus. Saudi Med J. 2015;36(4):399-406
  16. https://www.niddk.nih.gov/health-information/diabetes/overview/symptoms-causes
  17. Ramachandran A. Know the signs and symptoms of diabetes. Indian J Med Res. 2014;140(5):579-81. 
  18. Atkinson MA, Eisenbarth GS. Type 1 diabetes new perspective on diabetes pathogenesis and treatment, lancet. 2001; 358:221-229.
  19. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2011, 34;62-9.
  20. Gabbay KH, Merola LO, Field RA "Sorbitol pathway: presence in nerve and cord with substrate accumulation in diabetes" Science 1966;151, 3707:209–10.
  21. Lindstad RI, McKinley-McKee JS "Methylglyoxal and the polyol pathway. Three-carbon compounds are substrates for sheep liver sorbitol dehydrogenase". FEBS Lett. 1993; 330:31–5.
  22. Cheng HM, González RG "The effect of high glucose and oxidative stress on lens metabolism, aldose reductase, and senile cataractogenesis". Metab. Clin. Exp., 1986; 10–4.
  23. Gersing, S., Cagiada, M., Gebbia, M. et al. A comprehensive map of human glucokinase variant activity. Genome Biol, 2023; 24: 97.
  24. Greene ME, Blumberg B, McBride OW, Yi HF, Kronquist K, Kwan K, Hsieh L, Greene G, Nimer SD. "Isolation of the human peroxisome proliferator activated receptor γ cDNA: expression in hematopoietic cells and chromosomal mapping". Gene Expr. 1995; 4: 281–99.
  25. Elbrecht A, Chen Y, Cullinan CA, Hayes N, Leibowitz M, Moller DE, Berger J "Molecular cloning, expression and characterization of human peroxisome proliferator activated receptors γ1 and γ2". Biochem. Biophys. Res. Commun.,1996; 224.
  26. Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CN, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. “International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors” Pharmacol. Rev. 2006;58: 726–41.
  27. Embi N, Rylatt DB, Cohen P, “Glycogen synthase kinase-3 from rabbit skeletal muscle: separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase” Eur J Biochem., 1980;107:519 –527.
  28. Kanzariya NR, Patel RK, Patel NJ. Antidiabetic and vasoprotective activity of lithium: Role of glycogen synthase kinase-3. Indian J Pharmacol. 2011;43(4):433-6.
  29. Gruzman A, Babai G, Sasson S. Adenosine Monophosphate-Activated Protein Kinase (AMPK) as a New Target for Antidiabetic Drugs: A Review on Metabolic, Pharmacological and Chemical Considerations. Rev Diabet Stud. 2009;6(1):13-36.
  30. Nagappan A, Shin J, Jung MH. Role of Cannabinoid Receptor Type 1 in Insulin Resistance and Its Biological Implications. Int J Mol Sci. 2019,29;20(9):2109.
  31. Livanova NB, Chebotareva NA, Eronina TB, Kurganov BI "Pyridoxal 5′ Phosphate as a Catalytic and Conformational Cofactor of Muscle Glycogen Phosphorylase b". Biochemistry, 2002;10:1089–1998.
  32. Annie Bourdeau, Cytoplasmic protein tyrosine phosphatases, regulation and function: the roles of PTP1B and TC-PTP, Current Opinion in Cell Biology, 2005; 17:203–209.
  33. Alan Cheng, Michel L. Tremblay, CHAPTER 119 - Insulin Receptor PTP: PTP1B, Editor(s): Ralph A. Bradshaw, Edward A. Dennis, Handbook of Cell Signaling, Academic Press, 2003, 729-732,
  34. Kerru N, Singh-Pillay A, Awolade P, Singh P. Current anti-diabetic agents and their molecular targets: A review. Eur J Med Chem. 2018, 25; 152:436-488.
  35. Xiaodi Zhao, Dohee Ahn, Gibeom Nam, Jihee Kwon, Songyi Song, Min Ji Kang, Hyejin Ahn, Sang J. Chung, Identification of Crocetin as a Dual Agonist of GPR40 and GPR120 Responsible for the Antidiabetic Effect of Saffron, Nutrients, 2023; 15, 22, 4774.
  36. Dallatana A, Cremonesi L, Trombetta M, Fracasso G, Nocini R, Giacomello L, Innamorati G. G Protein-Coupled Receptors and the Rise of Type 2 Diabetes in Children. Biomedicines. 2023; 11(6):1576.
  37. Sue Chan, Glucagon Receptor, Editor(s): S.J. Enna, David B. Bylund, Pharm: The Comprehensive Pharmacology Reference, Elsevier, 2007;9: 1-7,
  38. Kasina SVSK, Baradhi KM. Dipeptidyl Peptidase IV (DPP IV) Inhibitors, 2023. In: StatPearls, Treasure Island (FL): StatPearls Publishing; 2024.
  39. Deacon CF, Physiology and pharmacology of DPP-4 in glucose homeostasis and the treatment of type 2 diabetes, front. Endocrinol., 2019; 10:80.
  40.  Faizan A. Beerwala, Shruti V. Kolambkar, Vishal S. Patil, Harish R. Darasaguppe, Nayeem A. Khatib, Vishwambhar V. Bhandare, Harsha V. Hegde, Subarna Roy, Decoding the alpha-amylase inhibitory activity of Garcinia indica Choisy by computational and experimental studies, South African Journal of Botany, 2024;165:14-29.
  41. Razavi-Nematollahi, L., Ismail-Beigi, F. Adverse Effects of Glycemia-Lowering Medications in Type 2 Diabetes. Curr Diab Rep, 2019; 19:132.
  42. https://londondiabetes.com/news-and-events/common-side-effects-of-diabetes medication.
  43. Ganesan K, Rana MBM, Sultan S. Oral Hypoglycemic Medications. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024.
  44. Blaslov, K., Naran?a, F. S., Kruljac, I., & Renar, I. P. Treatment approach to type 2 diabetes: Past, present, and future. World journal of diabetes, 2018; 9(12):209–219.
  45. Dowarah J, Singh VP. Anti-diabetic drugs recent approaches and advancements. Bioorg Med Chem. 2020; 28:115263.
  46. Thaer Idrees, Iris A Castro-Revoredo, Alexandra L Migdal, Emmelin Marie Moreno & Guillermo E Umpierrez., Update on the management of diabetes in long-term care facilities. BMJ Open Diabetes Research & Care, 2022; 10(4): 002705.
  47. Borse S, P, Chhipa A, S, Sharma V, Singh D, P, Nivsarkar M: Management of Type 2 Diabetes: Current Strategies, Unfocussed Aspects, Challenges, and Alternatives, Med Princ Pract. 2021;30(2):109-121.
  48. Gaba Monika, Singh Sarbjot, Gaba Pun, Dipeptidyl Peptidase-4 Inhibitors: a new approach in diabetes treatment, Int J. Drug Dev. & Res. 2009;1(1):146-156.
  49. Goyal R, Jialal I. Diabetes Mellitus Type 2. Book StatPearls Treasure Island, 2021.
  50. Nagaraju Kerru, Ashona Singh-Pillay, Paul Awolade, Parvesh Singh, Current antidiabetic agents, and their molecular targets: A review. Eur J Med Chem., 2018;152(25):436-488.
  51. Sapra A, Bhandari P. Diabetes Mellitus. StatPearls, Treasure Island (FL) 2022.
  52. Lv, Wei; Wang, Xianqing; Xu, Qian; Lu, Wencong Mechanisms and Characteristics of Sulfonylureas and Glinides, Current Topics in Medicinal Chemistry, 2020;20:37-56.
  53. Di Magno L, Di Pastena F, Bordone R, Coni S, Canettieri G. The Mechanism of Action of Biguanides: New Answers to a Complex Question. Cancers (Basel), 2022 30;14(13):3220
  54. Black C, Donnelly P, McIntyre L, Royle PL, Shepherd JP, Thomas S. Meglitinide analogues for type 2 diabetes mellitus. Cochrane Database Syst Rev. 2007 Apr 18.
  55. Kathryn M. Hurren & Marissa Waldman Dunham, thiazolidinediones a preferred drug treatment for type 2 diabetes, Expert Opinion on Pharmacotherapy, 2021, 22:2, 131-133.
  56. Akmal M, Wadhwa R. Alpha Glucosidase Inhibitors. [Updated 2022 Aug 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan.
  57. Liu S-K, Hao H, Bian Y, Ge Y-X, Lu S, Xie H-X, Wang K-M, Tao H, Yuan C, Zhang J, Zhang J, Jiang C-S and Zhu K., Discovery of new alfa-glucosidase inhibitors structure-based virtual screening and biological evaluation, front. Chem. 2021; 9:639279.
  58. Shubham Kumar, Anu Mittal, Amit Mittal, a review upon medicinal perspective and designing rationale of DPP-4 inhibitors, Bioorganic & Medicinal Chemistry, 2021;46:116354.
  59. Kasina SVSK, Baradhi KM. Dipeptidyl Peptidase IV (DPP IV) Inhibitors. StatPearls Treasure Island (FL): StatPearls Publishing; 2024.
  60. Deacon, C.F. Dipeptidyl peptidase 4 inhibitors in the treatment of type 2 diabetes mellitus. Nat Rev Endocrinol, 2020;16, 642–653.
  61. Gallwitz B Clinical Use of DPP-4 inhibitors, front. Endocrinol., 2019; 10:389.
  62. Padda IS, Mahtani AU, Parmar M. Sodium-Glucose Transport Protein 2 (SGLT2) Inhibitors, In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022.
  63. Available from: https://www.ncbi.nlm.nih.gov/books/NBK576405/
  64. Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Curr Opin Endocrinol Diabetes Obes. 2017;24(1):73-79.
  65. Hinnen D. Glucagon-Like Peptide 1 Receptor Agonists for Type 2 Diabetes. Diabetes Spectr., 2017;30(3):202-210.
  66. Collins L, Costello RA. Glucagon-Like Peptide-1 Receptor Agonists, In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024.
  67. Adeghate E, Kalász H. Amylin analogues in the treatment of diabetes mellitus: medicinal chemistry and structural basis of its function. Open Med Chem J. 2011;5(2):78-81.
  68. Edelman S, Maier H, Wilhelm K, Pramlintide in the treatment of diabetes mellitus, bio drugs, 2008;22(6): 375-86.
  69. Tiziano De Ventura, Vinicio Zanirato, Recent Advances in the Synthesis of Sulfonylureas, 2021;8: 1201-1214.
  70. Sroor, F.M., Basyouni, W.M., Aly, H.F. et al. Design, synthesis and SAR of novel sulfonylurea derivatives for the treatment of Diabetes mellitus in rats. Med Chem Res, 2022;31:195–206.
  71. Temel, Halide Edip, Alt?ntop, Mehlika Dilek, Sever, Belgin, Özdemir, Ahmet and Akal?n Çiftçi, Gül?en. "In vitro evaluation of 2-pyrazoline derivatives as DPP-4 inhibitors" Turkish Journal of Biochemistry, 2023; 48 (1): 104-109.
  72. Manisha Nidhar, Shaziya Khanam, Priyanka Sonker, Priya Gupta, Archisman Mahapatra, Swaraj Patil, Brijesh Kumar Yadav, Rahul Kumar Singh, Ashish Kumar Tewari, click inspired novel pyrazole-triazole-persulfonimide & pyrazole-triazole-aryl derivatives; Design, synthesis, DPP-4 inhibitor with potential anti-diabetic agents, Bioorganic Chemistry, 2022; 120, 105586.
  73. Gupta A, Bhat HR, Singh UP. Discovery of novel 1,3,5-triazine derivatives as an antidiabetic agent in Wistar rats via inhibition of DPP-4. Future Med Chem. 2023;15(10):829-852.
  74. Guang-Jing Feng, Yang-Fan Guo, Yuming Tang, Min Li, Yufei Jia, Zhimeng Li, Shuangshuang Wang, Hongmei Liu, Yuzhou Wu, and Hai Dong, Design, Synthesis, and Biological Evaluation of Thioglucoside Analogues of Gliflozin as Potent New Gliflozin Drugs, Journal of Medicinal Chemistry, 2023;66 (17):12536-12543.
  75. Masahiko Seki, Sandeep Ramesharao Tapkir, Maheshwara Reddy Nadiveedhi, Subarna Jyoti Kalita, Shaheen Kasim Mulani, and Kazushi Mashima, Synthesis of SGLT2 Inhibitors by Means of Fukuyama Coupling Reaction, The Journal of Organic Chemistry 2023; 88 (21):15367-15373.
  76. Yoshiaki Kitamura, Mahmoud Kandeel, Erina Oba, Chiori Iwai, Keitaro Iritani, Nanako Nagaya, Reo Namura, Hiroshi Katagiri, Hiroshi Ueda, Yukio Kitade, A Diversifiable Synthetic Platform for the Discovery of New Carbasugar SGLT2 Inhibitors Using Azide–Alkyne Click Chemistry, Chemical and Pharmaceutical Bulletin, 2023;71(3): 240-249.
  77. CenJun Li, Chao fan, Chunchao Wang, Jianchao Chen, Chengjun Wu, TieMin Sun, Synthesis, crystal structure, DFT calculation and confirmation of absolute configuration of empagliflozin, Journal of Molecular Structure, 2024;1301:137331.
  78. Asim Najmi, Md Shamsher Alam, Neelaveni Thangavel, Manal M. E. Taha, Abdulkarim M. Meraya, Mohammed Albratty, Hassan A. Alhazmi, Waquar Ahsan, Anzarul Haque & Faizul Azam, Synthesis, molecular docking, and in vivo antidiabetic evaluation of new benzylidene-2,4-thiazolidinediones as partial PPAR-γ agonists, Scientific Reports, 2023; 13(1) 2045-2322.
  79. Amin S, Sheikh KA, Iqubal A, Ahmed Khan M, Shaquiquzzaman M, Tasneem S, Khanna S, Najmi AK, Akhter M, Haque A, Anwer T, Mumtaz Alam M. Synthesis, in-Silico studies and biological evaluation of pyrimidine based thiazolidinedione derivatives as potential anti-diabetic agent. Bioorg Chem. 2023; 134:106449.
  80. Novel hybrids of thiazolidinedione-1,3,4-oxadiazole derivatives: synthesis, molecular docking, MD simulations, ADMET study, in vitro, and in vivo anti-diabetic assessment. Mahendra Gowdru Srinivasa, Jagdish Gopal Paithankar, Sumit Rao Saheb Birangal, Aravinda Pai, Vasudev Pai, Shridhar N. Deshpandea and B. C. Revanasiddappa. RSC Adv., 2023;13:1567-1579.
  81. Huneif, M.A.; Mahnashi, M.H.; Jan, M.S.; Shah, M.; Almedhesh, S.A.; Alqahtani, S.M.; Alzahrani, M.J.; Ayaz, M.; Ullah, F.; Rashid, U.; et al. New Succinimide–Thiazolidinedione Hybrids as Multitarget Antidiabetic Agents: Design, Synthesis, Bioevaluation, and Molecular Modelling Studies. Molecules 2023;28: 1207.
  82. Neda Shakour, Amirhossein Sahebkar, Gholamreza Karimi, Maryam Paseban, Aida Tasbandi, Fatemeh Mosaffa, Zahra Tayarani-Najaran, Razieh Ghodsi, Farzin Hadizadeh. Design, synthesis and biological evaluation of novel 5-(imidazolyl-methyl) thiazolidinediones as antidiabetic agents, Bioorganic Chemistry, 2021; 15:105162.
  83. Singh G, Singh R, Monga V, Mehan S. 3,5-Disubstituted-thiazolidine-2,4-dione hybrids as antidiabetic agents: Design, synthesis, in-vitro and in vivo evaluation. Eur J Med Chem. 2024, 15; 266:116139.
  84. Mengyue Li, Jinping Sun, Bingwen Liang, Xiaofeng Min, Jinhui Hu, Rihui Wu, Xuetao Xu, Thiazolidine-2,4-dione derivatives as potential α-glucosidase inhibitors: Synthesis, inhibitory activity, binding interaction and hypoglycemic activity, Bioorganic Chemistry, 2024;144:107177.
  85. Minoo Khalili Ghomi, Navid Dastyafteh, Mohammad Nazari Montazer, Milad Noori, Somayeh Mojtabavi, Mohammad Ali Faramarzi, Seyedeh Mahdieh Hashemi & Mohammad Mahdavi: Synthesis, in vitro potency of inhibition, enzyme kinetics and in silico studies of quinoline-based α-glucosidase inhibitors, Scientific Reports, 2024; 14(501). 
  86. Zahid Ali, Wajid Rehman*, Liaqat Rasheed, Abdullah Y. Alzahrani, Nawab Ali, Rafaqat Hussain, Abdul-Hamid Emwas, Mariusz Jaremko, and Magda H. Abdellattif: New 1,3,4-Thiadiazole Derivatives as α-Glucosidase Inhibitors: Design, Synthesis, DFT, ADME, and in Vitro Enzymatic Studies, American Chemical Society, 2024, 9(7):7480–7490.
  87. Yousefnejad, F., Mohammadi-Moghadam-Goozali, M., Sayahi, M. H., Halimi, M., Moazzam, A., Mohammadi-Khanaposhtani, M., Mojtabavi, S., Asadi, M., Faramarzi, M. A., Larijani, B., Amanlou, M., & Mahdavi, M.  Design, synthesis, in vitro, and in silico evaluations of benzo[d]imidazole-amide-1,2,3-triazole-N-arylacetamide hybrids as new antidiabetic agents targeting α-glucosidase. Scientific Reports, 2023;13(1): 12397.
  88. Emadi, M., Halimi, M., Moazzam, A., Hosseini, S., Mojtabavi, S., Faramarzi, M. A., Ghadimi, R., Moghadamnia, A. A., Nasli-Esfahani, E., Mohammadi-Khanaposhtani, M., & Mahdavi, M., Design, synthesis, in vitro anti-α-glucosidase evaluations, and computational studies of new phthalimide-phenoxy-1,2,3-triazole-N-phenyl (or benzyl) acetamides as potential anti-diabetic agents, Scientific Reports, 2023;13(1): 10030. 
  89. Tariq, H. Z., Saeed, A., Ullah, S., Fatima, N., Halim, S. A., Khan, A., El-Seedi, H. R., Ashraf, M. Z., Latif, M., & Al-Harrasi, A., Synthesis of novel coumarin–hydrazone hybrids as α-glucosidase inhibitors and their molecular docking studies. RSC Advances, 2023;13(37):26229–26238.
  90. Yuanzhen Dong, Jinhua Zhang, Hongjiang Xu, Hengqiao Shen, Qin Lu, Jun Feng, Zhengyan Cai, Design of a Novel Long-Acting Dual GLP-1/GIP Receptor Agonist, Bioorganic & Medicinal Chemistry, 2024;100: 117630.
  91. Jinhua Zhang, Hongjiang Xu, Jianguang Lu, Yuanzhen Dong, Jun Feng. Design, synthesis, and biological evaluation of a potential long-acting glucagon-like peptide-1 (GLP-1) analog. Bioorganic & Medicinal Chemistry, 2023; 85:117291.
  92. Maryam Ehsasatvatan & Bahram Baghban Kohnehrouz. Designing and computational analyzing of chimeric long-lasting GLP-1 receptor agonists for type 2 diabetes. Scientific Reports, 2023; 13:17778.
  93. Khyati Girdhar, Shilpa Thakur, Pankaj Gaur, Abhinav Choubey, Surbhi Dogra, Budheswar Dehury, Sunil Kumar, Bidisha Biswas, Durgesh Kumar Dwivedi, Subrata Ghosh, Prosenjit Mondal, Design, synthesis, and biological evaluation of a small molecule oral agonist of the glucagon-like-peptide-1 receptor, Journal of Biological Chemistry, 2022;298(5):101889.
  94. Suresh Thareja, Sant Kumar Verma, Akhlesh Kumar Jain, Manoj Kumar, Tilak Raj Bhardwaj, Rational design and synthesis of novel biphenyl thiazolidinedione conjugates as inhibitors of protein tyrosine phosphatase 1B for the management of type 2 diabetes, Journal of Molecular Structure, 2023;1274(1):134546.
  95. Farid M. Sroor, Wahid M. Basyouni, Hanan F. Aly, Sanaa A. Ali & Azza F. Arafa. Design, synthesis, and SAR of novel sulfonylurea derivatives for the treatment of Diabetes mellitus in rats. Med Chem Res., 2022; 31:195–206.
  96. Shah M, Jan MS, Sadiq A, Khan S, Rashid U. SAR and lead optimization of (Z)-5-(4-hydroxy-3-methoxybenzylidene)-3-(2-morpholinoacetyl) thiazolidine-2,4-dione as a potential multi-target antidiabetic agent, Eur J Med Chem. 2023;5(258)115591.
  97. Dr. Raja Biswas, Paul, K., Mukherjee, A. K., Bhattacherjee, I., and Mondal, S., “A comparative assessment of efficacy and safety of oral antidiabetic drug combinations, metformin plus pioglitazone versus metformin plus glimepiride in type-2 diabetes mellitus” in Indian journal of pharmacology, 2008.
  98. Ravi Kumar, Ponnusamy, Ravindran, Zinjarde, and Bhargava, “Evaluation of traditional Indian antidiabetic medicinal plants for human pancreatic amylase inhibitory effect in vitro”, Evidence-based Complementary and Alternative Medicine, vol. 2011; 2011.
  99. Roohallah Yousefi1,2. The Potential Application of Roselle Extracts (Hibiscus sabdariffa L.) in Managing Diabetes Mellitus. 1-11 http://aprh.journals.ekb.eg/
  100. Keyur Dudhat*, Anita Wanjari, Elijah Skarlus Doelakeh A Systematic Review: Antihyperglycemic Properties of Catharanthus roseus Linn. 1-8 https://www.jyoungpharm.org
  101. Harshvardhan  Pant a, Sandeep Negi a, Kumud Saklani b,*, Subhash Chandra c,*Lucia Raquel de Lima d, Henrique Douglas Melo Coutinho. Chemical composition and pharmacological activities of Ficus racemosa fruit methanolic extract.1-6 https://doi.org/10.1016/j.prenap.2025.100163
  102. Gabriella Alisha Sudarisman1, Sajuni Widjaja2, Puri Safitri Hanum3. Effect of Giving Papaya Leaf Extract (Carica Papaya L.) on Changes in Fasting Blood Glucose in Male Wistar Rats (Rattus Norvegicus) Induced by Streptozotocin. 1-6 https://doi.org/10.47191/ijmscrs/v5-i02-03
  103. Jinan M. Abugharsa Systematic Review: The Efficacy of Coriander,Cinnamon, Fenugreek, and Flaxseed, in Regulating Blood Sugar. 1-21 http://www.doi.org/10.62341/jima1954
  104. Unathi L. Mnge a, Corinne R. Ngnameko a,b, Veronica F. Salau a,Kolawole A. Olofinsan a, Abhay P. Mishra a, Motlalepula G. Matsabisa. Cinnamomum verum (Syn. C. zeylanicum) bark ethanolic extract inhibits carbohydrate digestive enzymes and enhances glucose uptake in 3T3-adipocytes: Insights from in vitro and computational perspectives. 1-12 https://doi.org/10.1016/j.sciaf.2025.e02539
  105. K. Satyanarayana1, K. Sravanthi2, I. Anand Shaker1, R. Ponnulakshmi3 Molecular approach to identify antidiabetic potential of Azadirachta indica.165-10 https://DOI:10.4103/0975-9476.157950
  106. Dipto Kumer Sarker1 | Pallobi Ray1 | Ashit Kumar Dutta2 | Razina Rouf3 |Shaikh Jamal Uddin. Antidiabetic potential of fenugreek (Trigonella foenum-graecum):A magic herb for diabetes mellitu. 1-26 https://DOI:10.1002/fsn3.4440
  107. shanmugapriya A.1, Jannathul Firdous2*, Karpagam T.1, Suganya V.1, Varalakshmi B. Anti-Diabetic and Free Radical Scavenging Activity of Phytochemicals from Caesalpinia bonducella. 1-10 https://doi.org/10.31632/ijalsr.2024.v07i03.015
  108. Juhaer Anjum1+, Maniza Muni1+, Nusrat Jahan Shawon2 , Fahima Aktar1 , Md. Mehadi Hasan3 , Mohammad Sofi3 , Md. Rafat Tahsin4 , Md Reaz Uddin5 , Abu Asad Chowdhury1 , Jakir Ahmed Chowdhury5 , Shaila Kabir1 , Md. Al Amin Sikder1 and Md. Shah Amran1 *. Exploration of the GLUT-4 Modulation Potential of Gynura procumbens as a Mechanism Behind its Antidiabetic Activity. 1-10 https://dx.doi.org/10.13005/bpj/3066
  109. Hui-Li Tan1, Kok-GanChan2, Priyia Pusparajah 1, Learn-HanLee1* and Bey HingGoh1* Gynura procumbens: An Overview of the Biological Activities. 1-10 https://doi.org/10.3389/fphar.2016.00052.

Reference

  1. American diabetes association. Diagnosis and classification of diabetes mellitus. Diabetes care, 2009;32:62-67.
  2. Goyal R, Jialal I. Diabetes Mellitus Type 2. Book StatPearls Treasure Island, 2021.
  3. Westman Eric C. Type 2 Diabetes Mellitus: A Pathophysiologic Perspective, Frontiers in Nutrition, 2021, 8.
  4. https://www.who.int/health-topics/diabetes#tab=tab_1
  5. Reusch, J.E. Manson. Management of type 2 diabetes in 2017: Getting to goal, Jama. 2017; 317:1015-1016.
  6. Sebokova, Elena Christ, Andreas Boehringer, Markus Mizrahi, Jacques. Dipeptidyl peptidase IV inhibitors: the next generation of new promising therapies for the management of type 2 diabetes, Curr. Topics in Medi. Chem. 2007, 7, 547-555.
  7. Sapra A, Bhandari P. Diabetes Mellitus. StatPearls, Treasure Island (FL) 2022.
  8. Chaudhury Arun, Duvoor Chitharanjan, Reddy Dendi Vijaya Sena, Kraleti Shashank, Chada Aditya, Ravilla Rahul, Marco Asween, Shekhawat Nawal Singh, Clinical Review of Antidiabetic Drugs: Implications for Type 2 Diabetes Mellitus Management, Frontiers in Endocrinology, 2017, 8, 6.
  9. Yan Zheng, Sylvia H. Ley & Frank B. Hu Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nature Reviews Endocrinology, 2018;14:88–98
  10. Banday MZ, Sameer AS, Nissar S. Pathophysiology of diabetes: An overview. Avicenna J Med. 2020 13;10(4):174-188.
  11. Jahangir Moini, Chapter 3 - Pathophysiology of Diabetes, Editor(s): Jahangir Moini, Epidemiology of Diabetes, Elsevier, 2019; 25-43,
  12. Lucier J, Weinstock RS. Diabetes Mellitus Type 1. In: StatPearls. Treasure Island 2021.
  13.  Global Burden of Disease Collaborative Network. Global Burden of Disease Study 2019. Results. Institute for Health Metrics and Evaluation. 2020.
  14. Wetsman EC type 2 diabetes mellitus; Pathophysiologic perspective, front. Nuti. 2021; 8:707371.
  15. Alfadhli EM. Gestational diabetes mellitus. Saudi Med J. 2015;36(4):399-406
  16. https://www.niddk.nih.gov/health-information/diabetes/overview/symptoms-causes
  17. Ramachandran A. Know the signs and symptoms of diabetes. Indian J Med Res. 2014;140(5):579-81. 
  18. Atkinson MA, Eisenbarth GS. Type 1 diabetes new perspective on diabetes pathogenesis and treatment, lancet. 2001; 358:221-229.
  19. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2011, 34;62-9.
  20. Gabbay KH, Merola LO, Field RA "Sorbitol pathway: presence in nerve and cord with substrate accumulation in diabetes" Science 1966;151, 3707:209–10.
  21. Lindstad RI, McKinley-McKee JS "Methylglyoxal and the polyol pathway. Three-carbon compounds are substrates for sheep liver sorbitol dehydrogenase". FEBS Lett. 1993; 330:31–5.
  22. Cheng HM, González RG "The effect of high glucose and oxidative stress on lens metabolism, aldose reductase, and senile cataractogenesis". Metab. Clin. Exp., 1986; 10–4.
  23. Gersing, S., Cagiada, M., Gebbia, M. et al. A comprehensive map of human glucokinase variant activity. Genome Biol, 2023; 24: 97.
  24. Greene ME, Blumberg B, McBride OW, Yi HF, Kronquist K, Kwan K, Hsieh L, Greene G, Nimer SD. "Isolation of the human peroxisome proliferator activated receptor γ cDNA: expression in hematopoietic cells and chromosomal mapping". Gene Expr. 1995; 4: 281–99.
  25. Elbrecht A, Chen Y, Cullinan CA, Hayes N, Leibowitz M, Moller DE, Berger J "Molecular cloning, expression and characterization of human peroxisome proliferator activated receptors γ1 and γ2". Biochem. Biophys. Res. Commun.,1996; 224.
  26. Michalik L, Auwerx J, Berger JP, Chatterjee VK, Glass CK, Gonzalez FJ, Grimaldi PA, Kadowaki T, Lazar MA, O'Rahilly S, Palmer CN, Plutzky J, Reddy JK, Spiegelman BM, Staels B, Wahli W. “International Union of Pharmacology. LXI. Peroxisome proliferator-activated receptors” Pharmacol. Rev. 2006;58: 726–41.
  27. Embi N, Rylatt DB, Cohen P, “Glycogen synthase kinase-3 from rabbit skeletal muscle: separation from cyclic-AMP-dependent protein kinase and phosphorylase kinase” Eur J Biochem., 1980;107:519 –527.
  28. Kanzariya NR, Patel RK, Patel NJ. Antidiabetic and vasoprotective activity of lithium: Role of glycogen synthase kinase-3. Indian J Pharmacol. 2011;43(4):433-6.
  29. Gruzman A, Babai G, Sasson S. Adenosine Monophosphate-Activated Protein Kinase (AMPK) as a New Target for Antidiabetic Drugs: A Review on Metabolic, Pharmacological and Chemical Considerations. Rev Diabet Stud. 2009;6(1):13-36.
  30. Nagappan A, Shin J, Jung MH. Role of Cannabinoid Receptor Type 1 in Insulin Resistance and Its Biological Implications. Int J Mol Sci. 2019,29;20(9):2109.
  31. Livanova NB, Chebotareva NA, Eronina TB, Kurganov BI "Pyridoxal 5′ Phosphate as a Catalytic and Conformational Cofactor of Muscle Glycogen Phosphorylase b". Biochemistry, 2002;10:1089–1998.
  32. Annie Bourdeau, Cytoplasmic protein tyrosine phosphatases, regulation and function: the roles of PTP1B and TC-PTP, Current Opinion in Cell Biology, 2005; 17:203–209.
  33. Alan Cheng, Michel L. Tremblay, CHAPTER 119 - Insulin Receptor PTP: PTP1B, Editor(s): Ralph A. Bradshaw, Edward A. Dennis, Handbook of Cell Signaling, Academic Press, 2003, 729-732,
  34. Kerru N, Singh-Pillay A, Awolade P, Singh P. Current anti-diabetic agents and their molecular targets: A review. Eur J Med Chem. 2018, 25; 152:436-488.
  35. Xiaodi Zhao, Dohee Ahn, Gibeom Nam, Jihee Kwon, Songyi Song, Min Ji Kang, Hyejin Ahn, Sang J. Chung, Identification of Crocetin as a Dual Agonist of GPR40 and GPR120 Responsible for the Antidiabetic Effect of Saffron, Nutrients, 2023; 15, 22, 4774.
  36. Dallatana A, Cremonesi L, Trombetta M, Fracasso G, Nocini R, Giacomello L, Innamorati G. G Protein-Coupled Receptors and the Rise of Type 2 Diabetes in Children. Biomedicines. 2023; 11(6):1576.
  37. Sue Chan, Glucagon Receptor, Editor(s): S.J. Enna, David B. Bylund, Pharm: The Comprehensive Pharmacology Reference, Elsevier, 2007;9: 1-7,
  38. Kasina SVSK, Baradhi KM. Dipeptidyl Peptidase IV (DPP IV) Inhibitors, 2023. In: StatPearls, Treasure Island (FL): StatPearls Publishing; 2024.
  39. Deacon CF, Physiology and pharmacology of DPP-4 in glucose homeostasis and the treatment of type 2 diabetes, front. Endocrinol., 2019; 10:80.
  40.  Faizan A. Beerwala, Shruti V. Kolambkar, Vishal S. Patil, Harish R. Darasaguppe, Nayeem A. Khatib, Vishwambhar V. Bhandare, Harsha V. Hegde, Subarna Roy, Decoding the alpha-amylase inhibitory activity of Garcinia indica Choisy by computational and experimental studies, South African Journal of Botany, 2024;165:14-29.
  41. Razavi-Nematollahi, L., Ismail-Beigi, F. Adverse Effects of Glycemia-Lowering Medications in Type 2 Diabetes. Curr Diab Rep, 2019; 19:132.
  42. https://londondiabetes.com/news-and-events/common-side-effects-of-diabetes medication.
  43. Ganesan K, Rana MBM, Sultan S. Oral Hypoglycemic Medications. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024.
  44. Blaslov, K., Naran?a, F. S., Kruljac, I., & Renar, I. P. Treatment approach to type 2 diabetes: Past, present, and future. World journal of diabetes, 2018; 9(12):209–219.
  45. Dowarah J, Singh VP. Anti-diabetic drugs recent approaches and advancements. Bioorg Med Chem. 2020; 28:115263.
  46. Thaer Idrees, Iris A Castro-Revoredo, Alexandra L Migdal, Emmelin Marie Moreno & Guillermo E Umpierrez., Update on the management of diabetes in long-term care facilities. BMJ Open Diabetes Research & Care, 2022; 10(4): 002705.
  47. Borse S, P, Chhipa A, S, Sharma V, Singh D, P, Nivsarkar M: Management of Type 2 Diabetes: Current Strategies, Unfocussed Aspects, Challenges, and Alternatives, Med Princ Pract. 2021;30(2):109-121.
  48. Gaba Monika, Singh Sarbjot, Gaba Pun, Dipeptidyl Peptidase-4 Inhibitors: a new approach in diabetes treatment, Int J. Drug Dev. & Res. 2009;1(1):146-156.
  49. Goyal R, Jialal I. Diabetes Mellitus Type 2. Book StatPearls Treasure Island, 2021.
  50. Nagaraju Kerru, Ashona Singh-Pillay, Paul Awolade, Parvesh Singh, Current antidiabetic agents, and their molecular targets: A review. Eur J Med Chem., 2018;152(25):436-488.
  51. Sapra A, Bhandari P. Diabetes Mellitus. StatPearls, Treasure Island (FL) 2022.
  52. Lv, Wei; Wang, Xianqing; Xu, Qian; Lu, Wencong Mechanisms and Characteristics of Sulfonylureas and Glinides, Current Topics in Medicinal Chemistry, 2020;20:37-56.
  53. Di Magno L, Di Pastena F, Bordone R, Coni S, Canettieri G. The Mechanism of Action of Biguanides: New Answers to a Complex Question. Cancers (Basel), 2022 30;14(13):3220
  54. Black C, Donnelly P, McIntyre L, Royle PL, Shepherd JP, Thomas S. Meglitinide analogues for type 2 diabetes mellitus. Cochrane Database Syst Rev. 2007 Apr 18.
  55. Kathryn M. Hurren & Marissa Waldman Dunham, thiazolidinediones a preferred drug treatment for type 2 diabetes, Expert Opinion on Pharmacotherapy, 2021, 22:2, 131-133.
  56. Akmal M, Wadhwa R. Alpha Glucosidase Inhibitors. [Updated 2022 Aug 12]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan.
  57. Liu S-K, Hao H, Bian Y, Ge Y-X, Lu S, Xie H-X, Wang K-M, Tao H, Yuan C, Zhang J, Zhang J, Jiang C-S and Zhu K., Discovery of new alfa-glucosidase inhibitors structure-based virtual screening and biological evaluation, front. Chem. 2021; 9:639279.
  58. Shubham Kumar, Anu Mittal, Amit Mittal, a review upon medicinal perspective and designing rationale of DPP-4 inhibitors, Bioorganic & Medicinal Chemistry, 2021;46:116354.
  59. Kasina SVSK, Baradhi KM. Dipeptidyl Peptidase IV (DPP IV) Inhibitors. StatPearls Treasure Island (FL): StatPearls Publishing; 2024.
  60. Deacon, C.F. Dipeptidyl peptidase 4 inhibitors in the treatment of type 2 diabetes mellitus. Nat Rev Endocrinol, 2020;16, 642–653.
  61. Gallwitz B Clinical Use of DPP-4 inhibitors, front. Endocrinol., 2019; 10:389.
  62. Padda IS, Mahtani AU, Parmar M. Sodium-Glucose Transport Protein 2 (SGLT2) Inhibitors, In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022.
  63. Available from: https://www.ncbi.nlm.nih.gov/books/NBK576405/
  64. Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Curr Opin Endocrinol Diabetes Obes. 2017;24(1):73-79.
  65. Hinnen D. Glucagon-Like Peptide 1 Receptor Agonists for Type 2 Diabetes. Diabetes Spectr., 2017;30(3):202-210.
  66. Collins L, Costello RA. Glucagon-Like Peptide-1 Receptor Agonists, In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024.
  67. Adeghate E, Kalász H. Amylin analogues in the treatment of diabetes mellitus: medicinal chemistry and structural basis of its function. Open Med Chem J. 2011;5(2):78-81.
  68. Edelman S, Maier H, Wilhelm K, Pramlintide in the treatment of diabetes mellitus, bio drugs, 2008;22(6): 375-86.
  69. Tiziano De Ventura, Vinicio Zanirato, Recent Advances in the Synthesis of Sulfonylureas, 2021;8: 1201-1214.
  70. Sroor, F.M., Basyouni, W.M., Aly, H.F. et al. Design, synthesis and SAR of novel sulfonylurea derivatives for the treatment of Diabetes mellitus in rats. Med Chem Res, 2022;31:195–206.
  71. Temel, Halide Edip, Alt?ntop, Mehlika Dilek, Sever, Belgin, Özdemir, Ahmet and Akal?n Çiftçi, Gül?en. "In vitro evaluation of 2-pyrazoline derivatives as DPP-4 inhibitors" Turkish Journal of Biochemistry, 2023; 48 (1): 104-109.
  72. Manisha Nidhar, Shaziya Khanam, Priyanka Sonker, Priya Gupta, Archisman Mahapatra, Swaraj Patil, Brijesh Kumar Yadav, Rahul Kumar Singh, Ashish Kumar Tewari, click inspired novel pyrazole-triazole-persulfonimide & pyrazole-triazole-aryl derivatives; Design, synthesis, DPP-4 inhibitor with potential anti-diabetic agents, Bioorganic Chemistry, 2022; 120, 105586.
  73. Gupta A, Bhat HR, Singh UP. Discovery of novel 1,3,5-triazine derivatives as an antidiabetic agent in Wistar rats via inhibition of DPP-4. Future Med Chem. 2023;15(10):829-852.
  74. Guang-Jing Feng, Yang-Fan Guo, Yuming Tang, Min Li, Yufei Jia, Zhimeng Li, Shuangshuang Wang, Hongmei Liu, Yuzhou Wu, and Hai Dong, Design, Synthesis, and Biological Evaluation of Thioglucoside Analogues of Gliflozin as Potent New Gliflozin Drugs, Journal of Medicinal Chemistry, 2023;66 (17):12536-12543.
  75. Masahiko Seki, Sandeep Ramesharao Tapkir, Maheshwara Reddy Nadiveedhi, Subarna Jyoti Kalita, Shaheen Kasim Mulani, and Kazushi Mashima, Synthesis of SGLT2 Inhibitors by Means of Fukuyama Coupling Reaction, The Journal of Organic Chemistry 2023; 88 (21):15367-15373.
  76. Yoshiaki Kitamura, Mahmoud Kandeel, Erina Oba, Chiori Iwai, Keitaro Iritani, Nanako Nagaya, Reo Namura, Hiroshi Katagiri, Hiroshi Ueda, Yukio Kitade, A Diversifiable Synthetic Platform for the Discovery of New Carbasugar SGLT2 Inhibitors Using Azide–Alkyne Click Chemistry, Chemical and Pharmaceutical Bulletin, 2023;71(3): 240-249.
  77. CenJun Li, Chao fan, Chunchao Wang, Jianchao Chen, Chengjun Wu, TieMin Sun, Synthesis, crystal structure, DFT calculation and confirmation of absolute configuration of empagliflozin, Journal of Molecular Structure, 2024;1301:137331.
  78. Asim Najmi, Md Shamsher Alam, Neelaveni Thangavel, Manal M. E. Taha, Abdulkarim M. Meraya, Mohammed Albratty, Hassan A. Alhazmi, Waquar Ahsan, Anzarul Haque & Faizul Azam, Synthesis, molecular docking, and in vivo antidiabetic evaluation of new benzylidene-2,4-thiazolidinediones as partial PPAR-γ agonists, Scientific Reports, 2023; 13(1) 2045-2322.
  79. Amin S, Sheikh KA, Iqubal A, Ahmed Khan M, Shaquiquzzaman M, Tasneem S, Khanna S, Najmi AK, Akhter M, Haque A, Anwer T, Mumtaz Alam M. Synthesis, in-Silico studies and biological evaluation of pyrimidine based thiazolidinedione derivatives as potential anti-diabetic agent. Bioorg Chem. 2023; 134:106449.
  80. Novel hybrids of thiazolidinedione-1,3,4-oxadiazole derivatives: synthesis, molecular docking, MD simulations, ADMET study, in vitro, and in vivo anti-diabetic assessment. Mahendra Gowdru Srinivasa, Jagdish Gopal Paithankar, Sumit Rao Saheb Birangal, Aravinda Pai, Vasudev Pai, Shridhar N. Deshpandea and B. C. Revanasiddappa. RSC Adv., 2023;13:1567-1579.
  81. Huneif, M.A.; Mahnashi, M.H.; Jan, M.S.; Shah, M.; Almedhesh, S.A.; Alqahtani, S.M.; Alzahrani, M.J.; Ayaz, M.; Ullah, F.; Rashid, U.; et al. New Succinimide–Thiazolidinedione Hybrids as Multitarget Antidiabetic Agents: Design, Synthesis, Bioevaluation, and Molecular Modelling Studies. Molecules 2023;28: 1207.
  82. Neda Shakour, Amirhossein Sahebkar, Gholamreza Karimi, Maryam Paseban, Aida Tasbandi, Fatemeh Mosaffa, Zahra Tayarani-Najaran, Razieh Ghodsi, Farzin Hadizadeh. Design, synthesis and biological evaluation of novel 5-(imidazolyl-methyl) thiazolidinediones as antidiabetic agents, Bioorganic Chemistry, 2021; 15:105162.
  83. Singh G, Singh R, Monga V, Mehan S. 3,5-Disubstituted-thiazolidine-2,4-dione hybrids as antidiabetic agents: Design, synthesis, in-vitro and in vivo evaluation. Eur J Med Chem. 2024, 15; 266:116139.
  84. Mengyue Li, Jinping Sun, Bingwen Liang, Xiaofeng Min, Jinhui Hu, Rihui Wu, Xuetao Xu, Thiazolidine-2,4-dione derivatives as potential α-glucosidase inhibitors: Synthesis, inhibitory activity, binding interaction and hypoglycemic activity, Bioorganic Chemistry, 2024;144:107177.
  85. Minoo Khalili Ghomi, Navid Dastyafteh, Mohammad Nazari Montazer, Milad Noori, Somayeh Mojtabavi, Mohammad Ali Faramarzi, Seyedeh Mahdieh Hashemi & Mohammad Mahdavi: Synthesis, in vitro potency of inhibition, enzyme kinetics and in silico studies of quinoline-based α-glucosidase inhibitors, Scientific Reports, 2024; 14(501). 
  86. Zahid Ali, Wajid Rehman*, Liaqat Rasheed, Abdullah Y. Alzahrani, Nawab Ali, Rafaqat Hussain, Abdul-Hamid Emwas, Mariusz Jaremko, and Magda H. Abdellattif: New 1,3,4-Thiadiazole Derivatives as α-Glucosidase Inhibitors: Design, Synthesis, DFT, ADME, and in Vitro Enzymatic Studies, American Chemical Society, 2024, 9(7):7480–7490.
  87. Yousefnejad, F., Mohammadi-Moghadam-Goozali, M., Sayahi, M. H., Halimi, M., Moazzam, A., Mohammadi-Khanaposhtani, M., Mojtabavi, S., Asadi, M., Faramarzi, M. A., Larijani, B., Amanlou, M., & Mahdavi, M.  Design, synthesis, in vitro, and in silico evaluations of benzo[d]imidazole-amide-1,2,3-triazole-N-arylacetamide hybrids as new antidiabetic agents targeting α-glucosidase. Scientific Reports, 2023;13(1): 12397.
  88. Emadi, M., Halimi, M., Moazzam, A., Hosseini, S., Mojtabavi, S., Faramarzi, M. A., Ghadimi, R., Moghadamnia, A. A., Nasli-Esfahani, E., Mohammadi-Khanaposhtani, M., & Mahdavi, M., Design, synthesis, in vitro anti-α-glucosidase evaluations, and computational studies of new phthalimide-phenoxy-1,2,3-triazole-N-phenyl (or benzyl) acetamides as potential anti-diabetic agents, Scientific Reports, 2023;13(1): 10030. 
  89. Tariq, H. Z., Saeed, A., Ullah, S., Fatima, N., Halim, S. A., Khan, A., El-Seedi, H. R., Ashraf, M. Z., Latif, M., & Al-Harrasi, A., Synthesis of novel coumarin–hydrazone hybrids as α-glucosidase inhibitors and their molecular docking studies. RSC Advances, 2023;13(37):26229–26238.
  90. Yuanzhen Dong, Jinhua Zhang, Hongjiang Xu, Hengqiao Shen, Qin Lu, Jun Feng, Zhengyan Cai, Design of a Novel Long-Acting Dual GLP-1/GIP Receptor Agonist, Bioorganic & Medicinal Chemistry, 2024;100: 117630.
  91. Jinhua Zhang, Hongjiang Xu, Jianguang Lu, Yuanzhen Dong, Jun Feng. Design, synthesis, and biological evaluation of a potential long-acting glucagon-like peptide-1 (GLP-1) analog. Bioorganic & Medicinal Chemistry, 2023; 85:117291.
  92. Maryam Ehsasatvatan & Bahram Baghban Kohnehrouz. Designing and computational analyzing of chimeric long-lasting GLP-1 receptor agonists for type 2 diabetes. Scientific Reports, 2023; 13:17778.
  93. Khyati Girdhar, Shilpa Thakur, Pankaj Gaur, Abhinav Choubey, Surbhi Dogra, Budheswar Dehury, Sunil Kumar, Bidisha Biswas, Durgesh Kumar Dwivedi, Subrata Ghosh, Prosenjit Mondal, Design, synthesis, and biological evaluation of a small molecule oral agonist of the glucagon-like-peptide-1 receptor, Journal of Biological Chemistry, 2022;298(5):101889.
  94. Suresh Thareja, Sant Kumar Verma, Akhlesh Kumar Jain, Manoj Kumar, Tilak Raj Bhardwaj, Rational design and synthesis of novel biphenyl thiazolidinedione conjugates as inhibitors of protein tyrosine phosphatase 1B for the management of type 2 diabetes, Journal of Molecular Structure, 2023;1274(1):134546.
  95. Farid M. Sroor, Wahid M. Basyouni, Hanan F. Aly, Sanaa A. Ali & Azza F. Arafa. Design, synthesis, and SAR of novel sulfonylurea derivatives for the treatment of Diabetes mellitus in rats. Med Chem Res., 2022; 31:195–206.
  96. Shah M, Jan MS, Sadiq A, Khan S, Rashid U. SAR and lead optimization of (Z)-5-(4-hydroxy-3-methoxybenzylidene)-3-(2-morpholinoacetyl) thiazolidine-2,4-dione as a potential multi-target antidiabetic agent, Eur J Med Chem. 2023;5(258)115591.
  97. Dr. Raja Biswas, Paul, K., Mukherjee, A. K., Bhattacherjee, I., and Mondal, S., “A comparative assessment of efficacy and safety of oral antidiabetic drug combinations, metformin plus pioglitazone versus metformin plus glimepiride in type-2 diabetes mellitus” in Indian journal of pharmacology, 2008.
  98. Ravi Kumar, Ponnusamy, Ravindran, Zinjarde, and Bhargava, “Evaluation of traditional Indian antidiabetic medicinal plants for human pancreatic amylase inhibitory effect in vitro”, Evidence-based Complementary and Alternative Medicine, vol. 2011; 2011.
  99. Roohallah Yousefi1,2. The Potential Application of Roselle Extracts (Hibiscus sabdariffa L.) in Managing Diabetes Mellitus. 1-11 http://aprh.journals.ekb.eg/
  100. Keyur Dudhat*, Anita Wanjari, Elijah Skarlus Doelakeh A Systematic Review: Antihyperglycemic Properties of Catharanthus roseus Linn. 1-8 https://www.jyoungpharm.org
  101. Harshvardhan  Pant a, Sandeep Negi a, Kumud Saklani b,*, Subhash Chandra c,*Lucia Raquel de Lima d, Henrique Douglas Melo Coutinho. Chemical composition and pharmacological activities of Ficus racemosa fruit methanolic extract.1-6 https://doi.org/10.1016/j.prenap.2025.100163
  102. Gabriella Alisha Sudarisman1, Sajuni Widjaja2, Puri Safitri Hanum3. Effect of Giving Papaya Leaf Extract (Carica Papaya L.) on Changes in Fasting Blood Glucose in Male Wistar Rats (Rattus Norvegicus) Induced by Streptozotocin. 1-6 https://doi.org/10.47191/ijmscrs/v5-i02-03
  103. Jinan M. Abugharsa Systematic Review: The Efficacy of Coriander,Cinnamon, Fenugreek, and Flaxseed, in Regulating Blood Sugar. 1-21 http://www.doi.org/10.62341/jima1954
  104. Unathi L. Mnge a, Corinne R. Ngnameko a,b, Veronica F. Salau a,Kolawole A. Olofinsan a, Abhay P. Mishra a, Motlalepula G. Matsabisa. Cinnamomum verum (Syn. C. zeylanicum) bark ethanolic extract inhibits carbohydrate digestive enzymes and enhances glucose uptake in 3T3-adipocytes: Insights from in vitro and computational perspectives. 1-12 https://doi.org/10.1016/j.sciaf.2025.e02539
  105. K. Satyanarayana1, K. Sravanthi2, I. Anand Shaker1, R. Ponnulakshmi3 Molecular approach to identify antidiabetic potential of Azadirachta indica.165-10 https://DOI:10.4103/0975-9476.157950
  106. Dipto Kumer Sarker1 | Pallobi Ray1 | Ashit Kumar Dutta2 | Razina Rouf3 |Shaikh Jamal Uddin. Antidiabetic potential of fenugreek (Trigonella foenum-graecum):A magic herb for diabetes mellitu. 1-26 https://DOI:10.1002/fsn3.4440
  107. shanmugapriya A.1, Jannathul Firdous2*, Karpagam T.1, Suganya V.1, Varalakshmi B. Anti-Diabetic and Free Radical Scavenging Activity of Phytochemicals from Caesalpinia bonducella. 1-10 https://doi.org/10.31632/ijalsr.2024.v07i03.015
  108. Juhaer Anjum1+, Maniza Muni1+, Nusrat Jahan Shawon2 , Fahima Aktar1 , Md. Mehadi Hasan3 , Mohammad Sofi3 , Md. Rafat Tahsin4 , Md Reaz Uddin5 , Abu Asad Chowdhury1 , Jakir Ahmed Chowdhury5 , Shaila Kabir1 , Md. Al Amin Sikder1 and Md. Shah Amran1 *. Exploration of the GLUT-4 Modulation Potential of Gynura procumbens as a Mechanism Behind its Antidiabetic Activity. 1-10 https://dx.doi.org/10.13005/bpj/3066
  109. Hui-Li Tan1, Kok-GanChan2, Priyia Pusparajah 1, Learn-HanLee1* and Bey HingGoh1* Gynura procumbens: An Overview of the Biological Activities. 1-10 https://doi.org/10.3389/fphar.2016.00052.

Photo
Dr. Manisha Nidhar
Corresponding author

Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Health Science Campus, Kochi, 682041, India. School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, 302017, India.

Photo
Sooraj Sura
Co-author

Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS, Health Science Campus, Kochi, 682041, India.

Photo
Vipin Kumar
Co-author

Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India

Photo
Shabnam Khan
Co-author

School of Pharmaceutical Sciences, Jaipur National University, Jaipur, Rajasthan, 302017, India

Photo
Ashish Kumar Tewari
Co-author

Department of Chemistry, Institute of Science, Banaras Hindu University, Varanasi 221005, Uttar Pradesh, India

Sooraj Sura, Vipin Kumar, Shabnam Khan, Ashish Kumar Tewari, Dr. Manisha Nidhar*, Recent Progress of Antidiabetic Agents for The Treatment of Type 2 Diabetes; A Mini-Review, Int. J. of Pharm. Sci., 2025, Vol 3, Issue 5, 1664-1689 https://doi.org/10.5281/zenodo.15382305

More related articles
Pyrimidine Derivatives as Promising Anti-Leishmani...
Raja Waleed Sajjad, Hammad Nasir, Ahmad Nawaz, Muhammad Mueen, Mu...
A Review on: Polyherbal Hand wash...
Pathan Shain Mahemood, N. S. Pendbhaje, Sonali D. More, Smeeta L....
Formulation And Evaluation Of Herbal Anti-Bacteria...
Ms.Priyanka Raut, Mrs.Manisha Mishra, Gayatri Bawane, Ayushi Amba...
Review A Mucoadhesive drug delivery system...
Rutuja S. Pawar, Sukumar M. Lande, Rupali R. Bendgude, ...
Medicinal Chemistry of Anticancer Drugs...
Pavan Sable , Mangesh Randive, Amol Shirode, ...
Related Articles
Prevention Of Obesity - The First And Foremost Strategy For Mankind To Prevent L...
J.C. Balachandar, M. Jayachandran, Shiv Swaroop Singh IRS, Saanvi Agrawal, S. Irshad Ahamed, A. Ayes...
Formulation And Evaluation Of Capsule Of Nifedipine By Liquisolid Compact ...
Bhagyashri S. Jadhav, Dnyanda H. Kangude , Awantika N. Kale, S.D. Mankar, ...
A Brief Review on Regulatory Affairs: Ensuring Compliance, Safety, and Market Ac...
Kishor S. Arote, Darshan A. Salade, Nilesh V. Patil, ...
Formulation of Immune Booster Ficus Carica Tea Pack ...
Chetan Bajare, Riteek Raghuwanshi, Nandakishor Deshmukh, Dr. Swati Deshmukh, ...
Pyrimidine Derivatives as Promising Anti-Leishmanial Agents, Integrative Molecul...
Raja Waleed Sajjad, Hammad Nasir, Ahmad Nawaz, Muhammad Mueen, Muhammad Ziaullah, Saba Manzoor , Raj...
More related articles
Pyrimidine Derivatives as Promising Anti-Leishmanial Agents, Integrative Molecul...
Raja Waleed Sajjad, Hammad Nasir, Ahmad Nawaz, Muhammad Mueen, Muhammad Ziaullah, Saba Manzoor , Raj...
A Review on: Polyherbal Hand wash...
Pathan Shain Mahemood, N. S. Pendbhaje, Sonali D. More, Smeeta L. Chandre, ...
Formulation And Evaluation Of Herbal Anti-Bacterial Crack Heel Ointment Using Th...
Ms.Priyanka Raut, Mrs.Manisha Mishra, Gayatri Bawane, Ayushi Ambade, Alshapha Anjum Khan, Abhilasha ...
Pyrimidine Derivatives as Promising Anti-Leishmanial Agents, Integrative Molecul...
Raja Waleed Sajjad, Hammad Nasir, Ahmad Nawaz, Muhammad Mueen, Muhammad Ziaullah, Saba Manzoor , Raj...
A Review on: Polyherbal Hand wash...
Pathan Shain Mahemood, N. S. Pendbhaje, Sonali D. More, Smeeta L. Chandre, ...
Formulation And Evaluation Of Herbal Anti-Bacterial Crack Heel Ointment Using Th...
Ms.Priyanka Raut, Mrs.Manisha Mishra, Gayatri Bawane, Ayushi Ambade, Alshapha Anjum Khan, Abhilasha ...