Octreotide is a synthetic cyclic octapeptide analogue of the endogenous hormone somatostatin, designed to mimic its inhibitory effects on hormone secretion while possessing a longer duration of action.[1] It is primarily indicated for the treatment of acromegaly, where it reduces elevated levels of growth hormone (GH) and insulin-like growth factor 1 (IGF-1) in patients who do not respond adequately to surgery, radiation, or other therapies such as bromocriptine.[2] Additionally, octreotide is used to alleviate severe symptoms associated with neuroendocrine tumors, including diarrhea and flushing episodes in carcinoid syndrome and profuse watery diarrhea in vasoactive intestinal peptide (VIP)-secreting tumors (VIPomas).[3][4]By selectively binding to somatostatin receptors (primarily subtypes 2 and 5), octreotide inhibits the release of GH from the anterior pituitary, as well as insulin, glucagon, and various gastrointestinal hormones, thereby modulating splanchnic blood flow and reducing secretory activity in the endocrine and exocrine pancreas.[1] Off-label applications include treatment of thyrotrophin-secreting pituitary adenomas (thyrotropinomas) to control symptomatic hypersecretion (though it does not cure underlying tumors), chemotherapy-induced diarrhea, variceal bleeding in cirrhosis, and certain insulinomas or glucagonomas, reflecting its broad utility in conditions involving peptide hormone excess.[1]Octreotide is available in multiple formulations to accommodate different clinical needs, including immediate-release solutions for subcutaneous or intravenous administration (in strengths of 50 mcg, 100 mcg, 200 mcg, 500 mcg, or 1000 mcg per mL), long-acting release (LAR) depot injections for intramuscular use (10 mg, 20 mg, or 30 mg), which allow for monthly dosing after initial stabilization with the short-acting form, and an oral delayed-release capsule (Mycapssa; 20 mg or 40 mg) approved in 2020 for long-term maintenance treatment of acromegaly in patients who have responded to and tolerated octreotide injection.[3][4][5] Brand names include Sandostatin, Sandostatin LAR Depot, and Bynfezia Pen, with the drug first approved by the U.S. Food and Drug Administration in 1988 for acromegaly and subsequently expanded for tumor-related indications.[2]
Medical uses
Acromegaly
Acromegaly is a rare endocrine disorder characterized by excessive secretion of growth hormone (GH), typically resulting from a pituitary adenoma, leading to elevated insulin-like growth factor-1 (IGF-1) levels and progressive somatic overgrowth.[6]Octreotide, a somatostatin analog, plays a key role in managing acromegaly by suppressing GH release from pituitary tumors, thereby reducing IGF-1 levels and alleviating associated symptoms. In clinical practice, it normalizes IGF-1 levels in approximately 50-70% of patients, with biochemical control (GH <2.5 ng/mL and normal IGF-1) achieved in 42-57% across pivotal trials.[7][8] Additionally, octreotide induces tumor size reduction in 20-50% of cases, with median volume decreases of 36.2% observed after 48 weeks of long-acting release (LAR) therapy in treatment-naïve patients.[7][8]Standard dosing regimens for octreotide in acromegaly begin with subcutaneous (SC) administration of 50 mcg three times daily for 2 weeks, followed by maintenance doses of 100-500 mcg SC three times daily, though doses exceeding 300 mcg/day seldom provide further benefit.[9] For the LAR formulation, patients transition to 20 mg intramuscularly every 4 weeks for 3 months, with adjustments to 10-30 mg monthly based on GH/IGF-1 response and clinical status; doses up to 40 mg may be used in select cases.[7]Pivotal multicenter trials, such as a 1995 study involving 103 patients treated for up to 30 months, demonstrate octreotide's efficacy in symptom relief, with improvements in headache, perspiration, fatigue, and joint pain reported in 83-95% of participants, alongside reductions in soft tissue swelling (e.g., finger circumference decrease at 12 months).[10] Effective management requires regular monitoring of IGF-1 and GH levels every 2-4 weeks initially and periodically thereafter to guide dose titration and assess treatment response.[9][7]
Neuroendocrine tumors
Octreotide is indicated for the long-term management of severe diarrhea and flushing episodes associated with metastatic carcinoid tumors, as well as profuse watery diarrhea in vasoactive intestinal peptide-secreting tumors (VIPomas).[11] In carcinoid syndrome, it reduces symptoms such as flushing, diarrhea, and wheezing by mimicking somatostatin and inhibiting the release of bioactive substances like serotonin and histamine from tumor cells.[12] For VIPomas, octreotide effectively controls secretory diarrhea by suppressing excessive vasoactive intestinal peptide (VIP) secretion, thereby alleviating profound fluid and electrolyte losses.[11]Initial dosing for these indications typically involves subcutaneous administration of 100-600 mcg per day in 2-4 divided doses (often 100-600 mcg three times daily), titrated based on symptom response over 2 weeks.[11] Patients may then transition to long-acting repeatable (LAR) formulations, such as 20 mg intramuscularly every 4 weeks, with adjustments to 10 mg or 30 mg as needed for optimal control; doses exceeding 30 mg are not recommended.[11] Clinical trials have shown substantial symptom improvement, with meta-analyses indicating response rates of 66% for overall symptoms, 65% for diarrhea, and 72% for flushing in patients with carcinoid syndrome treated with octreotide.[13]Beyond symptom palliation, octreotide exhibits antitumor effects through binding to somatostatin receptors (SSTRs) on neuroendocrine tumor cells, which inhibits cell proliferation and angiogenesis via downstream signaling pathways.[14] The PROMID trial, a placebo-controlled phase III study in patients with well-differentiated metastatic midgut neuroendocrine tumors, demonstrated that octreotide LAR (30 mg monthly) significantly prolonged median progression-free survival to 14.3 months compared to 6 months with placebo (hazard ratio 0.34, 95% CI 0.20-0.59).[15] This antiproliferative benefit was observed in both functioning and non-functioning tumors, with stable disease achieved in 67% of treated patients at 6 months.[15] Additionally, radiolabeled octreotide analogs are used for somatostatin receptor imaging to aid in tumor localization, as detailed in radiolabeled applications.[12]
Bleeding esophageal varices
Octreotide plays a critical role in the acute management of bleeding esophageal varices, a life-threatening complication of portal hypertension often seen in patients with liver cirrhosis. By mimicking somatostatin, it induces splanchnic vasoconstriction, inhibiting the release of vasodilatory hormones such as glucagon and directly constricting splanchnic arterioles, which reduces portal venous inflow and lowers portal pressure by 20-50%.[16] This hemodynamic effect helps stabilize the patient by decreasing variceal blood flow and promoting hemostasis prior to definitive endoscopic therapy.[17]The recommended dosing regimen for octreotide in this setting is an initial intravenous bolus of 50 mcg, followed by a continuous infusion of 50 mcg per hour for 2-5 days, or until bleeding is controlled and endoscopic treatment is completed. Emerging evidence from 2024-2025 studies suggests that shorter infusions (24 hours to 1 day) may be sufficient and noninferior to longer durations after successful endoscopic therapy, though guidelines as of 2024 recommend 2-5 days.[18][19][20] This approach allows for rapid onset of action, with peak effects within minutes of administration. Clinical guidelines from organizations such as the American Association for the Study of Liver Diseases (AASLD) endorse octreotide as a first-line vasoactive agent, particularly when combined with endoscopic variceal ligation or sclerotherapy, to optimize control of active bleeding and prevent early rebleeding.[21]Randomized controlled trials and meta-analyses have demonstrated that octreotide achieves initial hemostasis in approximately 80-90% of cases, comparable to vasopressin or terlipressin, but with a superior safety profile due to reduced risks of ischemic complications like myocardial infarction or limb ischemia.[17] Furthermore, evidence from these studies shows significant reductions in blood transfusion requirements (by an average of 1-2 units) and rebleeding rates within 5 days (from 32% with alternatives to 19% with octreotide), highlighting its efficacy in improving short-term outcomes without increasing overall mortality.[17][22]
Sulfonylurea-induced hypoglycemia
Octreotide plays an established off-label role in managing refractory hypoglycemia resulting from sulfonylurea overdose or factitious use, where it inhibits insulin release to address sulfonylurea-induced hyperinsulinemia.[23] Sulfonylureas stimulate excessive insulin secretion from pancreatic beta cells, leading to prolonged hypoglycemia that is often resistant to glucose infusions alone; octreotide counters this by binding to somatostatin receptors on beta cells, suppressing calcium influx and insulin exocytosis.[24] This targeted inhibition of stimulated insulin release distinguishes its application here from broader somatostatin effects detailed in pharmacodynamics.[23]Standard dosing for adults involves subcutaneous administration of 50–100 mcg every 6–12 hours, continued until blood glucose levels stabilize, typically for 12–24 hours after initial correction.[23] In pediatric cases, doses are adjusted to 1–2 mcg/kg subcutaneously every 6–12 hours, with close monitoring due to limited data.[24] This regimen is recommended as an adjunct to intravenous dextrose following initial hypoglycemia correction.[23]Clinical efficacy is supported by case series and trials demonstrating rapid normalization of blood glucose in approximately 80–90% of cases, outperforming glucose infusions alone by reducing recurrent hypoglycemic episodes and dextrose requirements.[24] For instance, a retrospective analysis of 13 patients showed hypoglycemic events decreasing from an average of 3.3 to 0.4 per patient after octreotide initiation, while a prospective trial in adults confirmed sustained euglycemia for 8 hours post-dose compared to placebo (p < 0.0001).[24] Early studies, such as a 1993 report on severe refractory cases, further evidenced its superiority, with all treated patients achieving stable glucose without rebound hypoglycemia.[25]Guidelines from toxicology societies, including the American College of Medical Toxicology, endorse octreotide as first-line adjunctive therapy for sulfonylurea-induced hypoglycemia when dextrose fails to prevent recurrence, based on accumulated case series evidence.[24] These series, spanning pediatric and adult overdoses since the early 2000s, consistently report fewer hypoglycemic events (e.g., 3.2 to 0.2 per patient, p = 0.008) and shorter hospital stays with its use.[24] An observation period of at least 12 hours after discontinuing octreotide and dextrose is advised to monitor for delayed recurrence.[23]In contrast to sulfonylurea-induced cases, octreotide's efficacy in endogenous hyperinsulinism, such as insulinomas, is less consistent, achieving hypoglycemia control in only about 50–70% of patients due to variable somatostatin receptor expression on tumor cells.[26]
Radiolabeled applications
Radiolabeled octreotide derivatives are employed in both diagnostic imaging and targeted radionuclide therapy for somatostatin receptor (SSTR)-positive tumors, particularly neuroendocrine tumors (NETs). For diagnostic purposes, indium-111 pentetreotide (Octreoscan) is a radiolabeled somatostatin analogue used in scintigraphy to localize primary and metastatic NETs that express somatostatin receptors. This agent binds to SSTR subtypes 2 and 5, enabling detection via single-photon emission computed tomography (SPECT) or planar imaging, with a reported success rate of 86.4% in identifying tumors compared to conventional methods like CT or MRI in clinical studies involving over 300 patients.[27]In therapeutic applications, lutetium-177 dotatate (Lutathera), an octreotide derivative conjugated to the beta-emitting radionuclide lutetium-177, is utilized in peptide receptor radionuclide therapy (PRRT) for adults and pediatric patients 12 years and older with SSTR-positive gastroenteropancreatic NETs (GEP-NETs), including those of midgut origin. In April 2024, FDA approval was expanded to pediatric patients 12 years and older. Lutathera delivers targeted radiation to tumor cells expressing high levels of SSTR2, leading to DNA damage and tumor regression while minimizing exposure to healthy tissues. The standard dosing regimen involves intravenous administration of 7.4 GBq (200 mCi) every 8 weeks for a total of four cycles, accompanied by an amino acid solution infusion (containing L-lysine and L-arginine) starting 30 minutes before, during, and for at least 3 hours after each dose to reduce renal radiation exposure by approximately 47%. The NETTER-2 trial (2024) showed that Lutathera plus octreotide LAR as first-line therapy for advanced Grade 2/3 GEP-NETs prolonged progression-free survival compared to high-dose octreotide alone.[28][29][30][31]The efficacy of Lutathera in PRRT was demonstrated in the phase 3 NETTER-1 trial, a randomized study of 229 patients with progressive, SSTR-positive midgut NETs, where addition of Lutathera to standard octreotide long-acting release therapy significantly prolonged median progression-free survival to 28.4 months compared to 8.4 months with octreotide alone (hazard ratio 0.21, 95% CI 0.13-0.33, P<0.001). At 20 months, 65% of patients in the Lutathera arm remained progression-free versus 11% in the control arm, with an objective response rate of 18% versus 3%. Patient selection for PRRT relies on confirmation of high SSTR2 expression, typically assessed via gallium-68 dotatate positron emission tomography (PET)/CT, which identifies suitable candidates by demonstrating uptake greater than or equal to normal liver in target lesions.[32][28][33]
Safety profile
Contraindications
Octreotide is contraindicated in patients with known hypersensitivity to octreotide or any of its components, as well as to other somatostatin analogues.[34][35] This absolute contraindication stems from the risk of severe allergic reactions, including rare reports of anaphylaxis and anaphylactoid shock following administration.[34][36]Relative contraindications include conditions requiring caution. For pregnancy, limited available data from case reports and postmarketing experience with octreotide use in pregnant women (mostly first trimester, 100-300 mcg/day) are insufficient to inform a drug-associated risk for major birth defects and miscarriage; no congenital malformations have been reported. Animal reproduction studies showed no adverse developmental effects at doses up to 7-13 times the maximum recommended human dose, though transient growth retardation occurred in rat offspring at lower doses.[37][1]Use during breastfeeding requires consideration of the developmental and health benefits of breastfeeding along with the mother's clinical need for octreotide and potential adverse effects on the breastfed infant; there are no data on its presence in human milk, but animal studies show low concentrations in rat milk (milk/plasma ratio 0.009).[37][38][39]Octreotide may improve fertility in premenopausal women by reducing GH/IGF-1 levels, so effective contraception is advised.[37]Octreotide suppresses thyroid-stimulating hormone (TSH) secretion, which may result in hypothyroidism, particularly with chronic use. It should be used with caution in patients with thyroid disorders; baseline and periodic thyroid function tests (e.g., TSH and free T4 levels) are recommended.[37][11][40]Special considerations apply to patients with short gut syndrome, where octreotide is generally avoided due to its inhibition of gastrointestinal hormones, which can impair intestinal adaptation and increase the risk of nutrient malabsorption.[41][42]Both the FDA and EMA guidelines emphasize these restrictions in product labeling, warning against use in hypersensitive individuals and urging careful evaluation in pregnancy, lactation, thyroid disorders, and conditions involving gut malabsorption to prevent serious complications.[34][35]
Adverse effects
Octreotide commonly causes gastrointestinal adverse effects, which occur in 30% to 61% of patients and are typically mild to moderate and transient. These include diarrhea, loose stools, nausea, and abdominal discomfort or pain.[1] Steatorrhea may also develop due to altered fat absorption, potentially leading to malabsorption and deficiencies in fat-soluble vitamins or vitamin B12 in some cases.[43]Endocrine-related side effects are frequent, particularly hyperglycemia from suppression of insulin secretion, affecting 16% of acromegalic patients, and hypoglycemia in 3%. Hypothyroidism develops in 10% to 15% of patients on chronic therapy, often subclinical, necessitating baseline and periodic thyroid function assessments such as TSH and free T4 levels.[44][1]Other notable adverse effects include gallbladder abnormalities like cholelithiasis or cholecystitis, occurring in 10% to 30% with long-term use (up to 63% in acromegaly), bradycardia in 25% of patients, and local pain at injection sites in approximately 8%. Serious effects are rare but can involve cardiac conduction abnormalities, such as arrhythmias (9%) or complete atrioventricular block, particularly with intravenous administration.[44][1]Management of these adverse effects involves dose adjustments to minimize gastrointestinal symptoms, regular monitoring of blood glucose, cardiac function via ECG in at-risk patients, and gallbladder ultrasound for long-term users. Ursodeoxycholic acid is effective for dissolving octreotide-induced gallstones in many cases, while thyroid hormone replacement addresses hypothyroidism, and vitamin supplementation treats deficiencies from steatorrhea.[44][43]
Drug interactions
Pharmacokinetic interactions
Octreotide, a somatostatin analog administered primarily via subcutaneous or intravenous routes, exhibits pharmacokinetic interactions primarily through its effects on gastrointestinal motility and absorption rather than direct metabolic interference. These interactions can alter the bioavailability and systemic exposure of co-administered drugs, as evidenced by pharmacokinetic studies measuring changes in area under the curve (AUC) and peak plasma concentrations.A notable interaction occurs with cyclosporine, where octreotide administration can reduce cyclosporine blood levels, likely due to slowed gastrointestinal absorption from octreotide-induced inhibition of gastric emptying and intestinal transit. This reduction in cyclosporine exposure necessitates close monitoring of cyclosporine concentrations and potential dose adjustments to maintain therapeutic efficacy, particularly in transplant patients.[2]Similarly, co-administration of octreotide increases the bioavailability of bromocriptine, a dopamine agonist, by up to 40%, as shown in pharmacokinetic trials where bromocriptine AUC was significantly elevated without affecting its elimination half-life. This interaction arises from octreotide's interference with oral absorption mechanisms, recommending monitoring of bromocriptine effects and potential dose adjustments when used concurrently.[1]Octreotide also impairs the absorption of certain oral medications by reducing gastrointestinal motility, leading to decreased AUC and prolonged time to peak concentration for these agents. Pharmacokinetic studies confirm these effects are transient and reversible upon discontinuation of octreotide, but monitoring is advised for drugs with narrow therapeutic indices.As a peptide analog, octreotide does not significantly interact with cytochrome P450 (CYP450) enzymes, avoiding induction or inhibition of hepatic metabolism pathways observed with many small-molecule drugs. This lack of CYP450 involvement minimizes risks of broader pharmacokinetic alterations in polypharmacy scenarios.
Pharmacodynamic interactions
Octreotide exhibits pharmacodynamic interactions with antidiabetic medications primarily through its suppression of insulin and glucagon secretion via somatostatin receptor activation, which can alter glucose homeostasis. In patients treated with insulin, octreotide may enhance the risk of hypoglycemia by reducing endogenous insulin counterregulation and improving peripheral insulin sensitivity, potentially decreasing insulin requirements by up to 50% in conditions like insulinoma or type 2 diabetes.[1] Similarly, concomitant use with sulfonylureas, which stimulate insulin release from pancreatic beta cells, can lead to additive insulin suppression, exacerbating hypoglycemia if doses are not adjusted, as evidenced by case reports of refractory hypoglycemia in sulfonylurea overdose managed with octreotide to inhibit excessive insulin secretion.[45][34]Initial administration of octreotide often induces hyperglycemia due to acute inhibition of insulin release, while long-term use may stabilize or improve glycemic control but introduces risks of prolonged hypoglycemia in diabetic patients on oral antidiabetics.[1][46] Clinical guidelines, including those from the FDA, recommend frequent blood glucose monitoring and dose adjustments for antidiabetic agents upon initiating octreotide therapy to mitigate these effects, with reported incidences of hypoglycemia at 3% and hyperglycemia at 16% in treated populations.[34][1]With beta-blockers, octreotide can potentiate bradycardia and hypotension through shared inhibitory effects on cardiac output and vascular tone, as both agents reduce heart rate and blood pressure via distinct but complementary pathways.[47] Dosage adjustments for beta-blockers may be necessary, particularly in patients with acromegaly where octreotide-induced bradycardia occurs in up to 25% of cases.[1][48]No major pharmacodynamic interactions have been identified with chemotherapy agents, though caution is warranted in neuroendocrine tumor patients receiving octreotide alongside cytotoxic therapies, with monitoring for additive gastrointestinal or hormonal effects recommended based on clinical consensus.[49][50]
Pharmacology
Mechanism of action
Octreotide is a synthetic cyclic octapeptide analogue of the native hormone somatostatin, designed to mimic its physiological effects while overcoming limitations of the endogenous peptide.[51] Native somatostatin has a very short plasma half-life of 1–3 minutes due to rapid enzymatic degradation, but octreotide incorporates D-amino acids (such as D-phenylalanine and D-tryptophan) and a disulfide bridge for cyclization, which confers resistance to peptidases and extends its half-life to approximately 90–120 minutes.[52] This structural modification allows for more sustained therapeutic activity compared to the native hormone.[53]Octreotide binds with high affinity to specific somatostatin receptor subtypes, particularly SSTR2 (IC50 ≈ 0.6 nM) and SSTR5 (IC50 ≈ 7 nM), which are predominantly expressed on endocrine cells and certain tumors.[54] These receptors belong to the family of G-protein-coupled receptors (GPCRs) that couple to inhibitory G proteins (Gi/o). Upon agonist binding, octreotide activates these G proteins, which inhibit adenylate cyclase enzyme activity.[51]The resulting decrease in intracellular cyclic AMP (cAMP) levels disrupts downstream signaling pathways that regulate hormone exocytosis, leading to suppressed secretion of multiple hormones targeted by somatostatin, including growth hormone from the pituitary, insulin and glucagon from pancreatic islets, gastrin from gastric G cells, and vasoactive intestinal peptide (VIP) from intestinal cells.[53] This mechanism underlies octreotide's role in modulating endocrine hypersecretion in various clinical contexts.[51]
Pharmacodynamics
Octreotide inhibits the secretion of various gastroenteropancreatic hormones, including gastrin, vasoactive intestinal peptide (VIP), secretin, motilin, and pancreatic polypeptide, which in turn reduces pancreatic enzyme secretion and biliary flow.[55] This suppression also decreases gastrointestinal motility and gallbladder contractility, contributing to decreased bile release.In terms of hemodynamic effects, octreotide induces vasoconstriction in the splanchnic and portal venous vasculature through contraction of vascular smooth muscle, thereby reducing splanchnic blood flow without causing systemic hypertension. These localized vascular changes are mediated primarily by somatostatin receptor subtypes 2 and 5.[1]Octreotide exerts endocrine effects by suppressing the growth hormone (GH)/insulin-like growth factor-1 (IGF-1) axis, leading to reduced GH and IGF-1 levels. It also modulates the balance of insulin and glucagonsecretion, often resulting in mild hyperglycemia due to predominant inhibition of insulin release.The drug demonstrates antitumor activity through antiproliferative effects, including the induction of apoptosis in cells expressing somatostatin receptors (SSTR), particularly via SSTR2 activation which triggers tyrosine phosphatases and inhibits pathways like PI3K/Akt.[56] This mechanism has been observed in neuroendocrine tumor cells and pituitary adenomas with high SSTR density.[56]Dose-response relationships vary by effect; endocrine suppression, such as GH inhibition, often occurs at lower doses (e.g., 50–500 mcg three times daily), while gastrointestinal effects like hormonesecretion inhibition may require higher doses (e.g., 100–600 mcg daily).
Pharmacokinetics
Octreotide is administered via subcutaneous, intravenous, or intramuscular routes, with the long-acting release (LAR) formulation designed for intramuscular injection to provide sustained release. Subcutaneous administration results in rapid and complete absorption, achieving peak plasma concentrations within 0.4 hours for a 100 mcg dose, with bioavailability approaching 100% compared to intravenous administration. The intramuscular LAR formulation exhibits a relative bioavailability of 60-63% versus subcutaneous injection, featuring an initial transient peak within 1 hour followed by a plateau phase reached after 2-3 weeks, enabling monthly dosing.[57][11][58]The volume of distribution at steady state is approximately 0.27 L/kg (13.6 L in healthy volunteers), with higher values (up to 21.6 ± 8.5 L) observed in patients with acromegaly; about 65% of octreotide is bound to plasma proteins, primarily lipoproteins and albumin. Octreotide minimally crosses the blood-brain barrier due to its peptide nature and efflux transport mechanisms.[57][11][59]Metabolism occurs primarily through enzymatic degradation into smaller peptides by tissue peptidases, with no involvement of hepatic cytochrome P450 enzymes. The elimination half-life for the immediate-release formulation is 1.5-1.9 hours in healthy individuals, extending to 2.4-3.1 hours in renal impairment and 3.7 hours in liver cirrhosis; for the LAR formulation, plasma concentrations show an initial transient peak within 1 hour, followed by a decline over 3-5 days and a subsequent rise to a plateau phase maintained for 2-3 weeks with monthly dosing, achieving steady-state after repeated administrations.[57][11][58]Excretion involves renal elimination of about 32% of the dose unchanged in urine, with the remainder undergoing fecal clearance following metabolism; dose adjustments are recommended in patients with renal impairment to account for prolonged half-life and reduced clearance (approximately 4.5 L/h in dialysis patients versus 10 L/h in healthy subjects). Octreotide may briefly interact with cyclosporine by potentially reducing its clearance, though this is addressed in detail under pharmacokinetic interactions.[57][11][58]
Chemistry
Chemical structure
Octreotide is a synthetic cyclic octapeptide that serves as an analogue of the native hormone somatostatin-14, designed to mimic its core pharmacophore while incorporating modifications for improved stability and duration of action. The molecule consists of eight amino acids with a disulfide bridge forming the cyclic structure, enabling a compact conformation essential for biological activity. This design was first detailed in the synthesis of SMS 201-995 (octreotide) by Bauer et al. in 1982, where the peptide's architecture was optimized to selectively target somatostatin receptors.[60]The molecular formula of octreotide is C49H66N10O10S2, and its molecular weight is 1019.2 Da. The amino acid sequence is H-D-Phe-Cys-Phe-D-Trp-Lys-Thr-Cys-Thr-ol, with cyclization occurring via a disulfide bond between the cysteine residues at positions 2 and 7 (denoted as a 2→7 bridge). This sequence retains the critical Phe-D-Trp-Lys-Thr motif from somatostatin-14, which forms the pharmacophore responsible for receptor interactions.[61]Key structural modifications include the incorporation of two D-amino acids—D-phenylalanine at position 1 and D-tryptophan at position 4—to resist enzymatic degradation, and replacement of the C-terminal threonine with threoninol (Thr-ol), a reduced alcohol derivative that further enhances metabolic stability. These alterations allow octreotide to adopt a type II' β-turn conformation around the D-Trp-Lys residues, as revealed by crystallographic studies, which positions the pharmacophore for effective binding to somatostatin receptors. The overall structure features a distorted β-turn and variable side-chain orientations, contributing to its selectivity and potency.[61]
Physical and chemical properties
Octreotide acetate is typically presented as a white to off-white lyophilized powder for pharmaceutical use.[62] This form is the acetatesalt of the synthetic cyclic octapeptide, which facilitates its reconstitution for injection.[63]The compound exhibits high solubility in aqueous media, with solubility exceeding 10 mg/mL in water at 25°C, as well as in glacial acetic acid and methanol.[64] It is freely soluble in solutions at pH 4-6, consistent with its behavior at acidic and physiological pH values, which supports its formulation for subcutaneous or intravenous administration.[65]Octreotide acetate is sensitive to light, heat, and moisture, requiring protection during storage and handling to maintain integrity.[66] It remains stable when stored at -20°C, and in solution, it is stable for up to 14 days at room temperature (20-30°C) if protected from light.[67]The pKa values of octreotide acetate are 7.02 and 10.15, corresponding to ionizable groups including the epsilon-amino group of lysine and other basic residues in the peptide structure.[65] These values influence its ionization state and solubility profile across physiological pH ranges. The logP value, indicative of lipophilicity, is approximately 1, which contributes to its ability to cross biological membranes despite its peptide nature.[68]
History
Development
Somatostatin, the natural peptide hormone that served as the basis for octreotide, was first isolated in 1973 by Paul Brazeau and colleagues at the Salk Institute, who demonstrated its role in inhibiting the secretion of growth hormone and other hormones from the pituitary gland. This discovery highlighted somatostatin's therapeutic potential for conditions involving excessive hormone secretion, such as acromegaly, but its extremely short plasma half-life of approximately 2-3 minutes posed significant challenges for clinical use.[69]To overcome these limitations, chemists at Sandoz Pharmaceuticals (now part of Novartis) in Basel, Switzerland, including Jean Pless and Wilfried Bauer, synthesized octreotide in 1979 as a stable octapeptide analog of somatostatin-14. The rationale focused on enhancing duration of action through key structural modifications, such as cyclization to form a more rigid structure and incorporation of D-amino acids (notably D-tryptophan at position 2), which increased enzymatic resistance and extended the half-life to about 90-120 minutes while preserving potent and selective inhibition of growth hormone release over insulin. These changes were informed by systematic analog screening starting in the mid-1970s, prioritizing selectivity for growth hormone over other somatostatin effects like gastrointestinal regulation.[70][69][71]Preclinical evaluation in the early 1980s involved animal studies in rats, dogs, and monkeys, where subcutaneous administration of octreotide demonstrated prolonged suppression of growth hormone secretion—up to 8-12 hours—far exceeding somatostatin's brief effect, with minimal impact on other hormones at therapeutic doses. These findings established octreotide's safety profile and efficacy in models of hypersecretion, paving the way for human investigation.[70][69]The first human trials began in the early 1980s under compassionate use protocols, targeting patients with acromegaly to assess growth hormone reduction and those with carcinoid syndrome for symptom relief from hormone excess. Initial phase I studies confirmed tolerability and rapid onset of action following subcutaneous injection. By 1984, phase II trials reported significant clinical benefits in acromegaly and carcinoid syndrome, establishing octreotide as a viable long-acting alternative to native somatostatin infusions.[69]
Regulatory approvals
Octreotide received its initial regulatory approvals in 1988. In the United States, the Food and Drug Administration (FDA) approved Sandostatin (octreotide acetate injection) for the symptomatic treatment of severe diarrhea and flushing episodes associated with metastatic carcinoid tumors where continuous infusion of a somatostatin analogue is necessary, as well as for reducing growth hormone levels in acromegaly patients who have not responded adequately to other treatments.[37]In Europe, octreotide was authorized on November 18, 1988, for similar indications, including acromegaly and carcinoid syndrome, with additional approval for the emergency management of bleeding gastro-oesophageal varices in cirrhotic patients.[72][73]Subsequent expansions broadened octreotide's applications. In 1998, the FDA approved Sandostatin LAR Depot, a long-acting repeatable intramuscular formulation, for the treatment of acromegaly and severe diarrhea/flushing in carcinoid syndrome patients who had responded to short-acting octreotide.[74] The EMA followed with similar authorization for the LAR formulation in acromegaly and neuroendocrine tumors. In 2020, the FDA approved Mycapssa, the first oral formulation of octreotide (delayed-release capsules), for long-term maintenance treatment of acromegaly in adults who have responded to and tolerated somatostatin analog therapy.[47] In December 2022, the EMA granted marketing authorization for Mycapssa in the European Union for the same indication; however, this authorisation was withdrawn on February 26, 2025.[75]Octreotide is approved in over 95 countries worldwide, with indications varying by region but generally encompassing acromegaly, carcinoid syndrome, VIPomas, and in some jurisdictions, variceal bleeding and TSH-secreting pituitary adenomas.[76] Recent updates include the EMA's approval on June 30, 2025, of Oczyesa, a once-monthly subcutaneous long-acting formulation of octreotide, for the treatment of acromegaly in adults, offering improved patient convenience over prior injectable options.[77]
Society and culture
Brand names and formulations
Octreotide is available under the primary brand name Sandostatin, marketed by Novartis, in immediate-release formulations for subcutaneous or intravenous administration. Sandostatin Injection is supplied as a clear, sterile solution in single-dose glass ampuls containing 50 mcg/mL, 100 mcg/mL, or 500 mcg/mL of octreotide (as octreotide acetate), with each 1 mL ampul also including lactic acid and mannitol as excipients.[37] For long-acting therapy, Sandostatin LAR Depot is provided as a lyophilized powder for injectable suspension in single-dose vials of 10 mg, 20 mg, or 30 mg strengths, intended for intramuscular gluteal injection after reconstitution with a diluent supplied in a pre-filled syringe; the formulation incorporates a biodegradable glucose star polymer matrix of D,L-lactic and glycolic acids copolymer for sustained release over approximately one month.[7]An oral formulation, Mycapssa (also by Chiasma, a Takeda company), consists of delayed-release capsules each containing 20 mg of octreotide (as octreotide acetate) coated with a proprietary delivery technology using a lipid-based excipient to enhance gastrointestinal absorption; it is prescribed for daily dosing ranging from 40 mg to 80 mg, typically administered as 20 mg or 40 mg twice daily with food.[78] Another immediate-release brand, Bynfezia Pen by Sun Pharmaceutical Industries, is available as a pre-filled, multi-dose pen delivering 50 mcg, 100 mcg, or 200 mcg per subcutaneous injection (equivalent to 0.5 mg/mL octreotide acetate solution), facilitating self-administration with a built-in dosing mechanism. It was discontinued in 2021 but re-approved and relaunched in 2024.[79]Generic versions of octreotide acetate are widely available, primarily as immediate-release injections in multi-dose vials or pre-filled syringes for subcutaneous use, with concentrations matching the branded Sandostatin (e.g., 100 mcg/mL or 200 mcg/mL in 1 mL or 5 mL volumes); these generics, approved by the FDA since 2010, include lactate buffer and are interchangeable with the reference product.[80] On October 1, 2024, Teva Pharmaceuticals launched the first generic equivalent to Sandostatin LAR Depot as octreotide acetate for injectable suspension in 20 mg kits, comprising a vial of lyophilized powder, a pre-filled diluentsyringe, vialadapter, and safety needle for monthly intramuscular administration.[81]Radiolabeled formulations based on octreotide derivatives are used for diagnostic and therapeutic purposes in neuroendocrine imaging and treatment. Octreoscan (by Curium Pharma), a diagnostic kit, prepares Indium In-111 pentetreotide (a DTPA-conjugated octreotide analog) as a sterile lyophilized powder in 10-vial kits for intravenous injection after radiolabeling, enabling somatostatin receptor scintigraphy.[27]
Lyophilized powder vials (10–30 mg) with diluent syringe
Intramuscular (gluteal)
Monthly dosing; microsphere suspension
Oral Capsules
Mycapssa
Delayed-release 20 mg capsules
Oral (with food)
Daily maintenance; lipid-enhanced absorption
Diagnostic Radiolabeled
Octreoscan
Kit for In-111 pentetreotide (lyophilized, ~111 MBq dose)
Intravenous
Somatostatin receptor imaging
Legal status
Octreotide is classified as a prescription-only medication throughout the world and is not scheduled as a controlled substance under any major international or national regulatory frameworks.[44][82]In the United States, the Food and Drug Administration (FDA) has designated octreotide as an orphan drug for the treatment of acromegaly and neuroendocrine tumors, including carcinoid syndrome, granting extended market exclusivity to incentivize development for these rare diseases. For instance, the oral formulation Mycapssa received orphan drug approval for long-term maintenance in acromegaly patients responsive to somatostatin analogs, with exclusivity extending until June 26, 2027.[83][84]Within the European Union, octreotide formulations such as Oczyesa are centrally authorized by the European Medicines Agency (EMA), facilitating consistent regulatory oversight and access across member states. Oczyesa (CAM2029), developed by Camurus AB, was approved by the EMA in July 2025 and launched in November 2025 as a ready-to-use, long-acting subcutaneous depot for monthly administration in acromegaly maintenance. Authorizations often incorporate pediatric investigation plans to evaluate potential use in children for relevant indications, though not all formulations have established pediatric efficacy or safety data.[77][85]Octreotide, as a high-cost biologic, is typically covered by health insurance for approved indications, with 99% of Medicare Part D plans providing reimbursement. Manufacturer-sponsored patient assistance programs, such as those from Novartis, offer the drug at no or reduced cost to eligible patients lacking adequate insurance coverage.[86][87]Off-label prescribing of octreotide is common, particularly for conditions like hypoglycemia in cases of hyperinsulinism or sulfonylurea overdose, and remains legally permissible in the United States as physicians exercise professional discretion outside FDA regulatory purview.[1][88]
Research directions
New formulations for acromegaly and neuroendocrine tumors
Recent advancements in octreotide formulations aim to enhance patient compliance by reducing injection frequency and offering non-invasive options for treating acromegaly and neuroendocrine tumors (NETs). These developments focus on sustained-release systems that maintain therapeutic levels of octreotide while minimizing administration burdens compared to traditional intramuscular long-acting repeatable (LAR) formulations.[89][90]CAM2029, a once-monthly subcutaneous depot formulation of octreotide utilizing FluidCrystal technology, has demonstrated promising efficacy in acromegaly. In the phase III ACROINNOVA 1 trial (NCT04076462), CAM2029 achieved IGF-1 control rates superior to placebo, with 72.2% of patients normalizing IGF-1 levels (≤ upper limit of normal) at weeks 22 and 24, alongside improvements in symptoms and quality of life. The ACROINNOVA 2 open-label extension (NCT04125836) reported sustained biochemical control and symptom improvement over 52 weeks. In April 2025, the European Medicines Agency's CHMP issued a positive opinion for CAM2029 (branded as Oczyesa) for the treatment of acromegaly. This subcutaneous approach offers self-administration potential, contrasting with the intramuscular injections required for LAR, and pharmacokinetic analyses indicate approximately fivefold higher bioavailability compared to LAR, supporting its role as an alternative for long-term management.[90][91][92]For oral delivery, expansions of Mycapssa (delayed-release octreotide capsules) have emphasized long-term adherence in acromegaly patients previously responsive to injectable somatostatin analogs. As of the 2023 analysis of the MPOWERED open-label extension, sustained biochemical control was reported in 82.9% of patients at 36 months, with median adherence rates exceeding 95%, highlighting improved convenience over twice-daily dosing requirements despite gastrointestinal side effects. These findings underscore Mycapssa's utility for maintenance therapy, though variable absorption necessitates monitoring.[93][94]In NETs, the phase III XT-XTR008-3-01 trial evaluated high-dose octreotide LAR (up to 60 mg monthly) against peptide receptor radionuclide therapy (PRRT) with 177Lu-Dotatate. Reported in 2025, the trial showed PRRT superior in progression-free survival (median not reached vs. 5.8 months; hazard ratio 0.06, p<0.0001) and objective response rates (43.4% vs. 1.0%) in advanced grade 1-2 gastroenteropancreatic NETs, suggesting high-dose LAR as a benchmark but inferior to targeted radionuclide combinations for disease control.[95]Overall, these formulations yield benefits such as reduced injection frequency—once monthly for CAM2029 versus every 28 days for LAR—and improved quality-of-life scores, with acromegaly patients reporting up to 30% better treatment satisfaction in convenience domains. However, oral forms like Mycapssa face challenges with bioavailability variability (20-60% absorption), potentially requiring dose adjustments to ensure consistent IGF-1 suppression.[96][97]
Emerging indications
Octreotide has been investigated for several potential applications beyond its established uses in acromegaly, neuroendocrine tumors, and carcinoid syndrome. Research in the 2020s has primarily focused on its somatostatin receptoragonist properties to modulate hormonal pathways in conditions involving abnormal growth or vascular factors, though clinical translation remains limited.[1]In obesity, particularly cases linked to hypothalamic damage or hyperinsulinemia, octreotide has shown potential for appetite modulation through suppression of insulin and satiety gut hormones such as GLP-1 and PYY. A 2006 randomized, placebo-controlled trial in obese adults with insulin hypersecretion demonstrated significant weight loss with octreotide long-acting release (LAR) at 40 mg or 60 mg doses, attributed to reduced postprandial insulin and improved insulin sensitivity.[98] However, a 2003 double-blind trial in pediatric hypothalamic obesity found only weight stabilization and BMI maintenance rather than reduction, indicating mixed efficacy.[99] More recent efforts, such as a 2021 ongoing trial (NCT04871204), explore octreotide to mitigate postoperative weight loss by countering exaggerated gut hormone responses that impair nutritional recovery, but results in general obesity populations remain inconclusive with no large-scale 2020s studies confirming broad appetite suppression benefits.[100]For polycystic liver disease (PLD), octreotide exhibits promise in symptom control and cyst volume reduction by inhibiting cyclic AMP-mediated fluid secretion in cyst-lining cells. The phase 2/3 POSITANO trial, completed in 2025, evaluated subcutaneous octreotide depot (CAM2029) in symptomatic PLD patients and reported significant reductions in height-adjusted total liver volume (4.3%) and liver cyst volume (8.7%) compared to placebo after 24 weeks, alongside improvements in abdominal symptoms and quality of life.[101] Earlier phase 2 studies supported these findings, showing modest cystgrowth inhibition over 6-12 months, though long-term hepatic benefits require further validation.[102]In diabetic macular edema (DME), octreotide's investigational role stems from its antiproliferative effects and inhibition of vascular endothelial growth factor (VEGF) expression via somatostatin receptors on retinal cells. A phase 1 trial initiated in 2025 (NCT06881888) is assessing the safety and preliminary efficacy of intranasal octreotide delivery in DME patients, aiming to reduce macular thickening and improve visual acuity without invasive injections.[103] This approach builds on preclinical evidence of VEGF suppression but awaits clinical outcomes to establish feasibility.[104]As of November 2025, no new regulatory approvals for these indications have been granted by the FDA or EMA, with ongoing research emphasizing combination therapies—such as octreotide paired with VEGF inhibitors for DME or mTOR inhibitors for PLD—to enhance efficacy while minimizing side effects.[105] Preliminary explorations into growth hormone (GH) modulation for neurodevelopmental conditions like autism remain speculative and lack robust clinical data.