What is the sacredness of Semaglutide 99% White powder? ——Decrypting the core ingredients of the "weight loss miracle drug" from the perspective of pharmaceutical raw material experts

December 23, 2025

In recent years, the global pharmaceutical industry has sparked a revolution in the treatment of metabolic diseases led by GLP-1 receptor agonists. At the core of this wave, there is a shining star molecule called Semaglutide. As pharmaceutical raw material experts, we not only need to see the brilliance of finished pills, but also need to delve into their raw powder - the material basis that determines the fate of drugs. Semaglutide 99% White powder,this seemingly ordinary white powder, in fact, contains the essence of modern biopharmaceutical technology. From the complex molecular structure design to the precise synthesis process, it reflects the profound understanding of human life science. This article will take you deeper into the molecular level, uncovering the mystery of Semaglutide 99% White Powder and exploring how it has transformed from a laboratory crystallization into a flower of life that improves the health of millions of people worldwide.

Molecular structural characteristics: One gram of raw powder, countless wisdom

The molecular structure of Semaglutide 99% White Powder is a carefully designed "metabolic regulatory machine", and its uniqueness cannot be summarized by a simple amino acid sequence. From a chemical perspective, Semaglutide is a polypeptide chain consisting of 31 amino acids with a molecular weight of approximately 4113.58 Da. However, if that's all, it is just an ordinary member among many GLP-1 analogs. The true essence of its structure lies in a series of carefully designed chemical modifications, which together solve the three major bottlenecks of natural GLP-1 in clinical applications: short half-life, easy degradation, and inconvenient administration.

MF of semaglutide

Firstly, let's focus on the most striking structural feature of Semaglutide molecules - fatty acid side chain modification. On the 26th lysine residue, an 18 carbon diacid (octadecanedioic acid) side chain is covalently linked via a gamma glutamic acid linker. This design is not arbitrary, but based on a profound understanding of the binding mechanism of albumin. The concentration of human serum albumin in the bloodstream is extremely high (about 500-700 μ M), and it has multiple hydrophobic binding pockets. The fatty acid side chains of Semaglutide can precisely embed into these pockets, forming stable non covalent bonds. This combination produces a dual effect: on the one hand, it significantly increases the molecular radius of semaglutide, exceeding the glomerular filtration threshold (approximately 30 kDa), thereby avoiding rapid excretion through the kidneys; On the other hand, albumin acts as a "protective shield", reducing the chances of peptide chains being degraded by various proteases in the bloodstream. Experimental data shows that this modification extends the plasma half-life of Semaglutide from 1-2 minutes of natural GLP-1 to approximately 165 hours (nearly 7 days), which is the key to its ability to achieve weekly administration.

Secondly, Semaglutide exhibits a specific spatial conformation in its original powder state, which directly affects its biological activity. Through nuclear magnetic resonance (NMR) and X-ray crystallography studies, scientists have discovered that Semaglutide forms a stable alpha helix structure in solution, particularly in the 7-28 amino acid region. This spiral structure is not rigid and unchanging, but has a certain degree of dynamic flexibility, which allows it to bind to GLP-1 receptors in a multi-step, induced binding manner. It is interesting that the molecules in Semaglutide powder are not completely uniform, but there are multiple conformational isomers present. Advanced analytical techniques such as circular dichroism (CD) and Fourier transform infrared spectroscopy (FTIR) show that the content of alpha helices in Semaglutide powder is about 60% -70%, beta folding is about 10% -15%, and the rest is irregular curling. This conformational diversity may be related to its dynamic behavior in vivo.

Thirdly, the amino acid substitution strategy reflects the precision of structural optimization. Compared with natural GLP-1, Semaglutide replaced alanine with alpha aminoisobutyric acid (Aib) at position 8. Aib is a non protein derived amino acid with a unique steric hindrance effect that effectively resists the cleavage of dipeptidyl peptidase-4 (DPP-4). DPP-4 is the main degradation enzyme of natural GLP-1, specifically cleaving the peptide bond of proline or alanine at the second N-terminal position. Through this clever substitution, Semaglutide has increased the stability of DPP-4 by over 1000 times. In addition, the 34th arginine was replaced by lysine, which not only maintains the positive charge characteristic, but also provides a suitable site for the connection of fatty acid side chains.

The solid-state characteristics of Semaglutide powder are also worth paying attention to. As a peptide active pharmaceutical ingredient, its crystal morphology directly affects the feasibility of the formulation process and the stability of the final product. Through powder X-ray diffraction (PXRD) analysis, Semaglutide raw powder usually exists in amorphous or partially crystalline form, which is closely related to its freeze-drying process conditions. Differential Scanning Calorimetry (DSC) shows that the glass transition temperature (Tg) of Semaglutide powder is about 120 ° C, indicating its good thermal stability. However, in high humidity environments, amorphous forms are prone to moisture absorption and reduce Tg, so the storage conditions of the raw powder must be strictly controlled for humidity.

In the early optimization of the structure of semaglutide, a systematic evaluation was conducted on fatty acid side chains of different lengths and structures. Experiments have found that when the length of the fatty acid chain is between 14-18 carbon atoms, the albumin binding ability and half-life extension effect reach the optimal balance. When it is less than 14 carbons, the binding force is insufficient; When it is longer than 18 carbons, it may increase the risk of immunogenicity. The final choice of 18 carbon diacid structure ensures sufficient lipophilicity to bind to albumin, while maintaining a certain hydrophilicity due to its dicarboxylic acid end, avoiding excessive aggregation. This choice was validated in subsequent preclinical studies: compared to analogs modified with 16 carbon monoacids, the half-life of semaglutide in crab eating monkeys was extended by about 40%, while the incidence of immune reactions was comparable.

Application field: "all rounder" from diabetes to metabolic syndrome

As an active pharmaceutical ingredient (API), Semalutide 99% White powder has expanded its application field from the initial treatment of type 2 diabetes to a broader spectrum of metabolic diseases, showing remarkable clinical value. This application expansion is not accidental, but based on its unique pharmacological effects and deep integration with the complexity of human metabolic diseases.

uses of semaglutide

Treatment of type 2 diabetes: cornerstone and breakthrough

The earliest and most mature application field of Semaglutide is the treatment of type 2 diabetes (T2DM). In this field, it is not only a tool for blood sugar control, but also a symbol of the transformation of disease management concepts. Traditional hypoglycemic drugs often focus on a single pathological link, while Semaglutide achieves a "multi pronged" approach to blood glucose control through multiple pathways of GLP-1 receptors.

The SUSTAIN clinical research project is the best proof of Semalutide's value in the field of diabetes. The SUSTAIN 6 trial is a 2-year cardiovascular outcome trial involving 3297 T2DM patients. The results showed that compared with the placebo group, the Semaglutide treatment group (1.0 mg per week) had a 1.5 percentage point decrease in glycated hemoglobin (HbA1c), and this effect remained stable for 104 weeks. More notably, the Semaglutide group reduced the risk of major adverse cardiovascular events (MACE) by 26% and the risk of non fatal stroke by 39%. These data not only confirm its potent hypoglycemic ability, but also establish its advantageous position in T2DM patients with concomitant cardiovascular disease.

From the perspective of its functional characteristics, Semaglutide's hypoglycemic effect is glucose dependent, that is, it only promotes the secretion of blood glucose regulators when blood glucose levels rise, and its effect weakens at normal blood glucose levels. This feature significantly reduces the risk of hypoglycemia. In the SUSTAIN 1 trial, the incidence of diagnosed hypoglycemia (blood glucose<3.1 mmol/L) in the Semaglutide 1.0 mg group was only 2.2%, which was not statistically different from the placebo group (1.8%).

Obesity Management: Reshaping the Weight Control Paradigm

If Semalutide is an important breakthrough in the field of diabetes, it is a revolutionary change in the field of obesity treatment. The STEP (Semaglutide Treatment Effect in People with Obesity) clinical program has completely changed the medical community's understanding of the effectiveness of drug weight loss.

The STEP 1 trial is a 68 week randomized double-blind study that recruited 1961 adults with a body mass index (BMI) ≥ 30 kg/m ² or ≥ 27 kg/m ² and at least one obesity related complication. The results were shocking: the average weight loss in the Semaglutide group (2.4 mg per week, higher than the treatment dose for diabetes) reached an astonishing 14.9%, while that in the placebo group was only 2.4%. This means that a patient with an initial weight of 100 kilograms can lose an average of nearly 15 kilograms. More notably, over one-third of patients achieved or exceeded a weight loss of 20%, a goal that was previously unattainable with any weight loss medication.

From the perspective of its mechanism of action, Semaglutide's weight loss effect stems from multiple synergistic effects: firstly, it enhances satiety signals through the central nervous system (especially the hypothalamus), reducing food intake; The second is to delay gastric emptying and increase the duration of satiety; Thirdly, it may increase energy consumption. PET-CT studies have shown that treatment with semaglutide can specifically activate appetite regulating centers such as the hypothalamic arcuate nucleus and solitary tract nucleus, while inhibiting brain activity related to food rewards such as the amygdala. This dual effect of central and peripheral functions makes its weight loss effect surpass traditional appetite suppressants.

Non alcoholic fatty liver disease (NAFLD): emerging treatment frontiers

With the deepening of research, the potential of Semaglutide in the treatment of NAFLD and non-alcoholic fatty liver disease (NASH) is gradually emerging. NAFLD is the most common chronic liver disease worldwide, affecting approximately 25% of the global population, and there are currently no approved drug therapies.

A 72 week phase II clinical trial provided strong evidence for this. This study included 320 NASH patients with liver fibrosis confirmed by biopsy, who were randomly assigned to the group receiving semaglutide 0.1 mg, 0.2 mg, 0.4 mg once daily or placebo. The results showed that 59% of patients in the 0.4 mg group achieved NASH remission without worsening of liver fibrosis, compared to only 17% in the placebo group. In addition, the proportion of liver fibrosis improvement (at least one stage) reached 43% in the Semaglutide 0.4 mg group, significantly higher than the 33% in the placebo group. These data indicate that Semaglutide not only improves hepatic steatosis and inflammation, but may also directly act on hepatic stellate cells to inhibit fibrosis progression.
From a pathophysiological perspective, the benefits of Semaglutide for NAFLD/NASH may stem from multiple mechanisms: weight loss and visceral fat are the main factors, but not the only ones. Animal studies have shown that GLP-1 receptor agonists can directly act on liver cells, reducing the expression of fatty acid synthesis genes (such as SREBP-1c) while increasing fatty acid oxidation. In addition, they can inhibit the activation of hepatic stellate cells and reduce collagen deposition. This multi-target effect gives

Cardiovascular benefits: value beyond blood glucose control

The cardiovascular protective effect of Semaglutide may be one of its most important clinical characteristics. This protective effect is partly due to its comprehensive improvement on traditional cardiovascular risk factors (blood glucose, blood pressure, blood lipids, weight), but there may also be direct effects independent of these factors.

Experimental studies have shown that GLP-1 receptors are widely expressed in the heart and vascular system. Semaglutide can enhance myocardial contractility by activating GLP-1 receptors in cardiomyocytes, increasing levels of cyclic adenosine monophosphate (cAMP), and possibly inhibiting cardiomyocyte apoptosis through the protein kinase A (PKA) pathway. At the vascular level, it can promote the expression of endothelial nitric oxide synthase (eNOS), improve endothelial function, and inhibit the formation of atherosclerotic plaque. Animal models have shown that Semaglutide treatment can reduce the aortic plaque area of apolipoprotein E knockout mice by more than 30%, and this effect can be observed even in mice with no significant changes in body weight.

Neuroprotective Potential: An Unexpected Discovery

Preclinical studies and observational clinical data suggest that Semaglutide may have neuroprotective properties. In animal models of Alzheimer's disease and Parkinson's disease, GLP-1 receptor agonists have been shown to reduce neuroinflammation, inhibit tau protein phosphorylation, and alpha synuclein aggregation. These effects may be related to the widespread distribution of GLP-1 receptors in the central nervous system, particularly in the hippocampus and cortical regions.
A retrospective study based on nationwide registration data in Denmark provides preliminary support for this. The study analyzed about 120000 T2DM patients, and found that the incidence rate of dementia in patients using GLP-1 receptor agonists (including Semaglutide) was about 40% lower than that in patients using other hypoglycemic drugs. Although this is only observational evidence, it has prompted the initiation of a large-scale phase III clinical trial (EVOKE study) specifically evaluating the efficacy of Semaglutide in early Alzheimer's disease, with highly anticipated results.

From the perspective of raw materials, the value of Semaglutide raw powder lies not only in its current clinical applications, but also in the metabolic regulatory network interaction principles it reveals. These constantly expanding indications reflect the paradigm shift of modern drug development from "one drug, one target" to "one drug, multiple effects". Semaglutide raw powder is like a precise key that can simultaneously open multiple doors to metabolic health, which is precisely its core value as a strategic level active pharmaceutical ingredient.

Mechanism of Action: Molecular Dancer and Metabolic Symphony

The mechanism of action of Semaglutide 99% White Powder in the body is like a carefully choreographed molecular ballet, involving precise collaboration of multiple organs and systems. To fully understand this process, we need to start with molecular interactions and gradually expand to the entire physiological system.

Receptor binding and activation: precise molecular recognition

The first step in the action of Semaglutide 99% White Powder is specific binding to GLP-1 receptors. GLP-1 receptors belong to the B-class G protein coupled receptor (GPCR) family and have a typical seven fold transmembrane structure. Compared with natural GLP-1, Semaglutide exhibits unique binding kinetics.

Surface plasmon resonance (SPR) studies have shown that the binding affinity (Kd value) of Semaglutide to GLP-1 receptors is approximately 0.38 nM, while natural GLP-1 is 0.1 nM. Although the value is slightly lower, the dissociation rate of Semaglutide is significantly slower than that of natural GLP-1, which prolongs receptor activation time. More importantly, Semaglutide exhibits biased signaling characteristics - it preferentially activates the G α s/cAMP pathway and is relatively weak in recruiting β - arrestin. This bias may bring clinical advantages: the cAMP pathway mediates most of the metabolic benefits, while the β - arrestin pathway is associated with receptor desensitization and certain side effects. Experiments have shown that the cAMP accumulation efficiency induced by Semaglutide is about 80% of that of natural GLP-1, but the duration of action is prolonged by more than 5 times.

From a structural biology perspective, the binding of Semaglutide to GLP-1 receptors is a multi-step process. Cryo EM structural analysis showed that the N-terminus of Semaglutide (especially the 7th histidine) penetrated deep into the receptor transmembrane domain, forming a hydrogen bond network with key residues such as E128 and R190; The alpha helix (positions 13-20) of the middle segment extensively contacts the extracellular domain of the receptor; The C-terminus (especially the lysine modified by the 26th fatty acid) is located in the proximal region of the receptor membrane, which may affect the conformational dynamics of the receptor. This multi-point binding mode creates highly stable complexes with a half-life of tens of minutes, much longer than the seconds of natural GLP-1.

Pancreatic effect: intelligent blood glucose regulation

In pancreatic beta cells, Semaglutide triggers a series of cascade reactions by activating GLP-1 receptors. Firstly, G α s protein activates adenylate cyclase, leading to an increase in intracellular cAMP levels. CAMP subsequently activates two main pathways, protein kinase A (PKA) and exchange protein directly activated by cAMP (Epac).

The PKA pathway works by phosphorylating multiple target proteins: it phosphorylates ATP sensitive potassium channels (KATP), promotes their closure, causes cell membrane depolarization and voltage dependent calcium channel opening, and triggers glucose regulating hormone vesicle exocytosis through calcium ion influx. Meanwhile, PKA also phosphorylates gene transcription factors (such as PDX-1), enhancing blood glucose regulation biosynthesis. The Epac pathway mainly activates the small G protein Rap1 to promote vesicle mobilization and docking.

It is worth noting that the hypoglycemic hormone secretion effect of semaglutide has strict glucose dependence. This characteristic arises from the cross dialogue between glucose metabolism and cAMP signaling within beta cells. Only when the glucose level is high enough, the ATP/ADP ratio inside the cell will increase, causing the KATP channel to close and the cell to be in a "ready to secrete" state. At this point, the cAMP signal induced by Semaglutide can be effectively converted into Blood glucose regulator secretion. This dual regulatory mechanism acts as a safety lock, effectively preventing the occurrence of hypoglycemia.

In alpha cells, Semaglutide also works through the cAMP PKA pathway, but the effect is opposite: it inhibits Glucagon secretion. This inhibition may be partially achieved through paracrine mechanisms - β - cell secreted blood glucose regulators and gamma aminobutyric acid (GABA) can inhibit adjacent α - cells. In addition, Semaglutide may directly act on GLP-1 receptors in alpha cells, regulating the secretion mechanism of Glucagon through PKA phosphorylation. This bidirectional regulation of blood glucose regulators and Glucagon enables Semaglutide to finely maintain blood glucose homeostasis.

Central nervous system function: reprogramming of appetite and reward

The weight loss effect of Semaglutide mainly stems from its profound impact on the central nervous system. Unlike many large molecular peptides that cannot cross the blood-brain barrier, Semaglutide can enter the brain through multiple mechanisms: firstly, its fatty acid side chains may promote binding with lipoproteins in the blood and enter through receptor-mediated transport; Secondly, it can come into contact with incomplete blood-brain barriers in the organs surrounding the ventricles (such as the posterior region); Thirdly, it is possible to indirectly affect the central nervous system by transmitting signals through the vagus nerve.

Functional magnetic resonance imaging (fMRI) studies provide direct evidence of the central role in humans. In a randomized double-blind trial, obese subjects received a single dose of Semaglutide or placebo treatment and underwent fMRI scans while viewing images of high calorie foods. The results showed that the Semaglutide group had significantly reduced activation in the amygdala, insula, and frontal orbital cortex, which are associated with food rewards and cravings; And the activation of the prefrontal cortex is enhanced, which is responsible for cognitive control and decision-making. The change in this neural activity pattern reflects the shift of Semaglutide's appetite regulation from "instinctive drive" to "cognitive control".

At the molecular level, Semaglutide activates GLP-1 receptors in the hypothalamic arcuate nucleus, regulating two classic appetite regulating neuronal groups: promoting POMC neurons to release alpha melanocyte stimulating hormone (alpha MSH), which is a potent anorexia signal; Simultaneously inhibiting the activity of neuropeptide Y (NPY)/agouti associated protein (AgRP) neurons, which are important appetite promoting signal sources. This dual regulation breaks the "set point" of energy balance, allowing the body to adapt to lower weight levels.

Gastrointestinal Dynamics: Delaying Emptying and Feeling Full

The delaying effect of Semaglutide on gastric emptying is an important mechanism for generating satiety. By activating vagus nerve fibers and directly acting on GLP-1 receptors in gastric smooth muscle, it reduces the amplitude and frequency of gastric antral contractions while enhancing pyloric tension.

Cardiovascular and renal protection: multi organ benefits

The cardiovascular and renal protective effects of Semaglutide involve complex mechanisms. At the cardiac level, animal experiments have shown that Semaglutide can reduce myocardial infarction area and inhibit ischemia-reperfusion injury through the PI3K/Akt and MEK/ERK pathways. The molecular mechanism includes reducing mitochondrial permeability transition pore opening and inhibiting caspase-3 activation, thereby reducing cardiomyocyte apoptosis.

In the vascular system, the mechanism by which Semaglutide improves endothelial function includes increasing eNOS expression and activity, reducing reactive oxygen species (ROS) production, and inhibiting vascular cell adhesion molecule-1 (VCAM-1) expression. Clinical studies have shown that treatment with Semaglutide for 12 weeks can improve blood flow mediated vasodilation (FMD) in T2DM patients by 2.1%, which is direct evidence of improved endothelial function.

In terms of kidney protection, Semaglutide may exert its effects through multiple pathways: reducing glomerular pressure (by dilating afferent arterioles), inhibiting kidney inflammation and fibrosis, and directly protecting podocytes. The post hoc analysis of the SUSTAIN 6 trial showed that the Semaglutide group had a 36% reduced risk of developing or worsening kidney disease, and this effect remained significant after adjusting for changes in blood glucose and blood pressure, suggesting the possibility of a direct renal protective mechanism.

Metabolic reprogramming: an integrated effect beyond a single organ

The ultimate therapeutic effect of Semaglutide stems from its integrated regulation of the systemic metabolic network. By synchronously regulating feeding behavior, energy consumption, substrate metabolism, and multiple organ functions, it achieves the reconstruction of metabolic homeostasis.

From a systems biology perspective, the role of Semaglutide can be seen as a recalibration of the metabolic "control system". It enhances hormone sensitivity not only through peripheral effects, but also through central effects - hypothalamic GLP-1 receptor activation can enhance the sensitivity of liver and muscle hormones, possibly mediated by the autonomic nervous system. In addition, Semaglutide can promote browning of white adipose tissue, increase mitochondrial biogenesis and fatty acid oxidation, and this metabolic reprogramming may play a key role in long-term weight maintenance.

The true brilliance of Semaglutide's mechanism of action lies in its spatiotemporal specificity. Due to its binding properties, albumin forms a "circulating reservoir" in the body, slowly releasing active molecules. This pharmacokinetic characteristic enables it to maintain a stable receptor occupancy rate, avoiding fluctuations in concentration peaks and valleys, thereby producing stable and long-lasting pharmacological effects. From the perspective of pharmaceutical raw material experts, this perfect connection from molecular design to systemic effects is a model of modern rational drug design and the core value of Semaglutide powder as an efficient API.

mechanism of action of semaglutide

Research direction: The future is here, exploration is endless

The research on Semaglutide 99% White Powder is far from limited to current clinical applications, and global research teams are pushing its scientific boundaries from multiple dimensions to explore new possibilities. These research directions may not only expand their therapeutic spectrum, but also deepen our understanding of metabolic regulatory networks.

Development of new dosage forms

The development of oral formulations represents a more significant technological advancement. The launch of Semaglutide oral tablets is a milestone event in the field of peptide drug delivery, but its bioavailability is only about 1%, which has prompted researchers to seek further optimization.

The current research focuses on new strategies to improve oral bioavailability. One is the application of new absorption enhancers: in addition to the already used SNAC, other compounds that promote cross cellular transport are being evaluated, such as medium chain fatty acid salts, surfactants, and tight junction modulators. Preclinical studies have shown that certain novel enhancers can increase the oral bioavailability of Semaglutide to 3-5%, which is several times higher than the current level.

The second is the exploration of nanocarrier systems. New carriers such as lipid nanoparticles (LNP), polymer nanoparticles, and self microemulsifying drug delivery systems can protect semaglutide from gastrointestinal degradation, promote lymphatic transport, and bypass first pass effects. Animal experiments have shown that the relative bioavailability of chitosan nanoparticles loaded with semaglutide in rats can reach 15% of that of injections, which is a significant improvement.

New indication expansion: possibilities beyond metabolic diseases

In addition to established metabolic diseases, the potential of semaglutide in other fields is being actively explored.

Neurodegenerative diseases are one of the most concerning new directions. Two phase III studies evaluating the efficacy of oral semaglutide in early Alzheimer's disease suggest that semaglutide can exert neuroprotective effects through multiple pathways: reducing beta amyloid deposition and tau protein hyperphosphorylation, inhibiting neuroinflammation, and promoting neurotrophic factor expression. Animal models have shown that treatment with semaglutide can reduce hippocampal beta amyloid plaques in APP/PS1 transgenic mice by 40% and improve cognitive function test performance.

Combination therapy: maximizing synergistic effects

The combination application of semaglutide with other drugs may produce synergistic effects, which is currently one of the research hotspots.

The combination with sodium glucose cotransporter 2 inhibitor (SGLT2i) has attracted much attention. These two drugs act through different but complementary pathways: semaglutide primarily reduces glucose input (suppresses appetite, delays emptying), while SGLT2 inhibitors increase glucose output (promotes urinary glucose excretion). The SUSTAIN 9 trial showed that the addition of Somautide in patients who had used SGLT2i could reduce HbA1c by an additional 1.4 percentage points and weight by an additional 4.7 kg. More importantly, this combination might provide "dual cardiac protection" - Somaglutide to reduce atherosclerosis events, and SGLT2i could improve the outcome of heart failure.

From these research directions, it can be seen that the scientific journey of semaglutide powder is far from over. Every application expansion, every mechanism discovery, and every process improvement is expanding the potential boundaries of this extraordinary molecule. As pharmaceutical raw material experts, we have witnessed its glory now and look forward to its promising future.

Conclusion

The story of Semaglutide 99% White Powder is a microcosm of modern drug development - from a deep understanding of natural hormones to rational design based on structure; From exploring a single target to understanding systemic effects; From the breakthrough of diabetes treatment to the revolution of metabolic syndrome management. This gram of white powder embodies the wisdom of structural biology, the ingenuity of medicinal chemistry, the innovation of formulation science, and the validation of clinical medicine. It is not only a product of the pharmaceutical industry, but also a strategic weapon for humans to combat metabolic diseases. With the continuous deepening of research, we have reason to believe that Semaglutide and its derived molecules will continue to expand the boundaries of treatment, bringing healthier and better lives to hundreds of millions of patients worldwide. From the perspective of pharmaceutical raw materials, this is not just a raw material drug, but also a perfect intersection of scientific imagination and human health needs.

Xi'an Faithful BioTech Co., Ltd. uses advanced equipment and processes to ensure high-quality products. We produce high-quality raw Semaglutide 99% White powder that meet international drug standards. Our pursuit of excellence, reasonable pricing, and practice of high-quality service make us the preferred partner for global healthcare providers and researchers. If you need to conduct scientific research or production of Semaglutide, please contact our technical team through sales12@faithfulbio.com.

Reference

Lau, J., Bloch, P., Schäffer, L., Pettersson, I., Spetzler, J., Kofoed, J., ... & Knudsen, L. B. (2015). Discovery of the once-weekly glucagon-like peptide-1 (GLP-1) analogue semaglutide. Journal of Medicinal Chemistry, *58*(18), 7370-7380.

Marso, S. P., Bain, S. C., Consoli, A., Eliaschewitz, F. G., Jódar, E., Leiter, L. A., ... & Vilsbøll, T. (2016). Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. New England Journal of Medicine, *375*(19), 1834-1844.

Wilding, J. P., Batterham, R. L., Calanna, S., Davies, M., Van Gaal, L. F., Lingvay, I., ... & Kushner, R. F. (2021). Once-weekly semaglutide in adults with overweight or obesity. New England Journal of Medicine, *384*(11), 989-1002.

Newsome, P. N., Buchholtz, K., Cusi, K., Linder, M., Okanoue, T., Ratziu, V., ... & Harrison, S. A. (2021). A placebo-controlled trial of subcutaneous semaglutide in nonalcoholic steatohepatitis. New England Journal of Medicine, *384*(12), 1113-1124.

Gabery, S., Salinas, C. G., Paulsen, S. J., Ahnfelt-Rønne, J., Alanentalo, T., Baquero, A. F., ... & Jelsing, J. (2020). Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight, *5*(7), e133429.

Davies, M., Pieber, T. R., Hartoft-Nielsen, M. L., Hansen, O. K. H., Jabbour, S., & Rosenstock, J. (2017). Effect of oral semaglutide compared with placebo and subcutaneous semaglutide on glycemic control in patients with type 2 diabetes: a randomized clinical trial. JAMA, *318*(15), 1460-1470.

Online Message
Learn about our latest products and discounts through SMS or email