Polyamines are small organic polycations characterized by aliphatic hydrocarbon chains bearing two or more primary amino groups, rendering them positively charged at physiological pH, and they are ubiquitous in prokaryotic and eukaryotic cells across all living organisms.[1] The principal polyamines in mammalian cells are putrescine (a diamine), spermidine (a triamine), and spermine (a tetramine), with putrescine serving as the precursor for the others.[2] First observed in human semen by Antonie van Leeuwenhoek in 1678, these compounds were later identified as essential regulators of cellular processes.[1]Polyamines are primarily synthesized endogenously through a tightly regulated biosynthetic pathway starting from the amino acid L-ornithine, which is decarboxylated by the rate-limiting enzyme ornithine decarboxylase (ODC) to form putrescine; subsequent addition of aminopropyl groups from decarboxylated S-adenosylmethionine yields spermidine and spermine.[2] They can also be obtained exogenously from dietary sources such as soybeans, wheat germ, and cheese, or produced by gut microbiota, with estimated daily intake in humans around 300 µmol.[3] Intracellular polyamine levels are maintained at millimolar concentrations through a balance of synthesis, catabolism (via polyamine oxidases producing reactive oxygen species), and transport mechanisms involving specific carriers.[4]These molecules exert diverse biological functions, including stabilization of DNA and RNA structures through electrostatic interactions with negatively charged nucleic acids, facilitation of protein synthesis and post-translational modifications (such as hypusination of eukaryotic initiation factor 5A), and modulation of ion channels, gene expression, and autophagy.[2] Polyamines are indispensable for cell proliferation and differentiation, with their depletion arresting growth in mammalian cells, and they also influence apoptosis, immune responses, and stress adaptation.[4] In mammalian physiology, spermidine in particular has been linked to promoting longevity in model organisms like yeast and flies, potentially through induction of autophagy, though polyamine levels decline with aging in humans.[1]Dysregulation of polyamine metabolism contributes to various pathophysiological states, including elevated levels in cancers such as colorectal, prostate, and ovarian tumors, where they support tumor growth, invasion, and immunosuppression in the tumor microenvironment.[4] Dysregulation of polyamine metabolism, often involving altered levels, is associated with cardiovascular diseases and neurodegenerative disorders like Alzheimer's and Parkinson's, highlighting their dual roles in health maintenance and disease progression.[2] Recent studies as of 2024 have linked higher dietary polyamine intake to lower risk of cardiovascular disease mortality.[5] Therapeutic strategies targeting polyamine pathways, such as inhibition of ODC with difluoromethylornithine (DFMO), have shown promise in reducing tumor burden and improving outcomes in high-risk neuroblastoma and other malignancies.[1]
Chemical Properties and Structure
Definition and General Structure
Polyamines are a class of small organic compounds defined by the presence of two or more primary amino groups (-NH₂), typically linked by short aliphatic hydrocarbon chains of 3 to 4 carbons between the nitrogen atoms. These polycations are ubiquitous in living organisms and play essential roles in cellular processes, though synthetic variants also exist. The general formula for the simplest polyamines, known as diamines, is H₂N-(CH₂)ₙ-NH₂, where n commonly equals 3 or 4; for instance, putrescine, the most basic polyamine, corresponds to n=4 with the structure H₂N-(CH₂)₄-NH₂.[6][2][7]Higher polyamines extend this architecture with additional amino groups, forming linear chains in most natural cases—such as triamines like spermidine (H₂N-(CH₂)₃-NH-(CH₂)₄-NH₂) or tetraamines like spermine (H₂N-(CH₂)₃-NH-(CH₂)₄-NH-(CH₂)₃-NH₂)—though branched structures occur in some organisms and synthetic designs. At physiological pH (around 7.4), the amino groups undergo protonation, resulting in a net positive charge that renders polyamines polycationic and enables interactions with negatively charged biomolecules like nucleic acids. This protonated state is nearly complete for primary and secondary amines under cellular conditions.[7][8][9]The historical discovery of polyamines traces back to putrescine, first isolated in 1885 from putrefying meat by German physician Ludwig Brieger as a decomposition product of proteins, with its structure confirmed via synthesis by chemist Albert Ladenburg in 1886. Spermine was initially observed as crystalline deposits in human semen in 1678 by microscopist Antonie van Leeuwenhoek, but its precise chemical structure was not elucidated until 1924 through synthetic efforts by researchers including Otto Rosenheim. Basic nomenclature categorizes these compounds by amino group count: diamines (two groups, e.g., putrescine), triamines (three, e.g., spermidine), and tetraamines (four, e.g., spermine).[10][11][12][13][2]
Physical and Chemical Properties
Polyamines are generally colorless, hygroscopic solids or low-melting liquids that display high water solubility attributable to hydrogenbonding between their amino groups and water molecules.[14][15] For instance, putrescine appears as a colorless oil or crystalline solid and is very soluble in water, while spermidine and spermine are colorless solids with solubilities exceeding 50 mg/mL and 100 mg/mL in water, respectively.[16][17] Their boiling points tend to increase with molecular chain length; putrescine, the simplest aliphatic polyamine, boils at 158.5 °C at atmospheric pressure.[14]The chemical properties of polyamines stem primarily from their multiple amino groups, which confer strong basicity with pKa values typically ranging from 8 to 11, enabling stepwise protonation to form polycations.[18] Specific examples include putrescine (pKa 9.04 and 10.50), spermidine (pKa 8.25, 9.71, and 10.90), and spermine (pKa 7.96, 8.85, 10.02, and 10.80).[19] This basicity facilitates the formation of salts with acids, such as the common dihydrochlorides. The lone pairs on nitrogen atoms also enable nucleophilic reactivity, allowing polyamines to undergo reactions with electrophiles like carbonyl compounds or alkyl halides. Additionally, polyamines exhibit chelating ability toward divalent metal ions, including Mg²⁺ and Ca²⁺, via coordination through their nitrogen atoms.[20] They remain relatively stable under neutral conditions but can degrade in strong acids or bases due to protonation-induced hydrolysis or deprotonation effects.[21]Spectroscopic characteristics provide insights into their structure and bonding. In ¹H NMR spectra, the methylene protons adjacent to amino groups (-CH₂-NH₂) resonate at chemical shifts of approximately 2.5-3.0 ppm, reflecting the electron-withdrawing influence of the nitrogen.[22]Infrared (IR) spectroscopy reveals characteristic N-H stretching bands for primary and secondary amines in the 3300-3500 cm⁻¹ region, often appearing as broad or multiple peaks due to hydrogen bonding.[23] These features are consistent across common polyamines like putrescine, spermidine, and spermine.
Types of Polyamines
Natural Polyamines
Natural polyamines are small, positively charged aliphatic molecules essential to cellular processes across living organisms, with the most prevalent forms being putrescine (1,4-diaminobutane), spermidine (N-(3-aminopropyl)butane-1,4-diamine), and spermine (N,N'-bis(3-aminopropyl)butane-1,4-diamine).[24]Cadaverine (1,5-diaminopentane) is another common diamine polyamine, particularly abundant in certain bacteria and plants.[24] Thermospermine, a tetraamine isomer of spermine, is predominantly found in plants, where it occurs from primitive algae to higher land plants.[25]These polyamines are ubiquitous in all domains of life, including prokaryotes (bacteria and archaea) and eukaryotes (fungi, plants, animals), reflecting their fundamental role in cellular architecture.[26] In mammals, total polyamine concentrations are typically around 0.1 to 1.5 mM in the brain, among the lower levels observed among mammalian tissues.[27][4] Spermidine and spermine are notably enriched in semen, with spermine concentrations up to 14 mM, contributing to its characteristic properties, and both are associated with ribosomes in various cell types.[28]Putrescine is a key polyamine in bacteria, derived from decarboxylation of arginine or ornithine.[29]Variations in polyamine profiles exist across species and environments; for instance, agmatine, formed from arginine, predominates in certain bacteria where it serves as a precursor or signaling molecule.[30] Sym-homospermidine, a triamine analog of spermidine, is characteristic of extreme thermophiles like Thermus thermophilus.[31] Polyamine levels also vary dynamically with cellular states, often elevating in rapidly dividing cells to support growth demands.[32]The presence of polyamines in bacteria, archaea, and eukaryotes underscores their evolutionary conservation, suggesting an ancient origin tied to early cellular stabilization of nucleic acids and proliferation.[33]
Synthetic Polyamines
Synthetic polyamines encompass a diverse class of compounds produced through chemical synthesis, primarily for industrial and technological purposes, differing from naturally occurring polyamines by their tailored structures and large-scale manufacturing processes.Ethylenediamine (H₂N-CH₂-CH₂-NH₂), the simplest ethyleneamine, is industrially produced via the ethylene dichloride (EDC) process, involving the reaction of 1,2-dichloroethane with aqueous ammonia under pressure at approximately 180°C, or alternatively through the reductive amination of monoethanolamine (MEA).[34] Higher homologs, such as diethylenetriamine (DETA), are obtained as byproducts in the same processes or through sequential amination steps. Global production capacity for ethyleneamines, including ethylenediamine, exceeds 300,000 tonnes per year as of 2023, with major producers like Dow and BASF contributing significantly to output.[35]Other notable synthetic polyamines include hexamethylenetetramine (urotropin), synthesized by the condensation of formaldehyde with ammonia in either liquid-phase or gas-phase processes, yielding a cage-like structure used in resin production and as a reagent.[36] Dendrimeric polyamines, such as poly(amidoamine (PAMAM) dendrimers, are constructed through iterative Michael addition and amidation reactions starting from an ethylenediamine core, resulting in highly branched, nanoscale architectures with precise control over size and functionality for advanced applications.[37]These compounds find widespread use as surfactants and chelating agents in detergents, where ethylenediamine derivatives form complexes like EDTA to bind metal ions and enhance cleaning efficacy.[38] They also serve as hardeners for epoxy resins, providing rapid curing at room temperature with low viscosity and good adhesion properties in coatings, adhesives, and composites.[39] In pharmaceuticals, synthetic polyamines act as vectors for drug delivery, leveraging their cationic nature to encapsulate and transport therapeutic agents.[37]Recent advancements since 2020 have focused on biodegradable synthetic polyamines, such as poly(β-amino esters) (PBAEs), designed as non-viral vectors for gene therapy; these polymers feature ester linkages in the backbone for controlled degradation, improving biocompatibility and transfection efficiency in targeted delivery systems.[40]
Biosynthesis and Metabolism
Biosynthetic Pathways
In eukaryotes, the biosynthesis of polyamines primarily occurs through a dedicated pathway starting with the decarboxylation of ornithine to form putrescine, catalyzed by the pyridoxal 5'-phosphate-dependent enzymeornithine decarboxylase (ODC), which is the rate-limiting step in this process.[2] Subsequent steps involve the transfer of aminopropyl groups from decarboxylated S-adenosylmethionine (dcSAM), generated by S-adenosylmethionine decarboxylase (SAMDC), to putrescine and spermidine. Spermidine synthase (SPDS) facilitates the formation of spermidine from putrescine and dcSAM, releasing 5'-methylthioadenosine (MTA) as a byproduct, as depicted in the reaction:\text{putrescine} + \text{dcSAM} \rightarrow \text{spermidine} + \text{MTA}Spermine synthase (SPMS) then converts spermidine to spermine via another aminopropyl transfer from dcSAM.[2]In prokaryotes, polyamine biosynthesis exhibits variations, particularly in the initial production of putrescine, where arginine decarboxylase (ADC) serves as an alternative to ODC, especially in bacteria and some plants, converting arginine to agmatine as an intermediate before hydrolysis to putrescine.[2] This ADC-dependent route is prominent in many bacterial species, such as Escherichia coli, and contributes to environmental adaptation, while the downstream aminopropyl transfer steps to form spermidine and spermine remain conserved and rely on SAMDC, SPDS, and SPMS.[2]Plants feature additional specialized pathways, including the synthesis of thermospermine, a tetraamine isomer of spermine, from spermidine via the enzyme encoded by the ACAULIS5 (ACL5) gene, which acts as a thermospermine synthase and is highly homologous to SPMS.[41] The expression of the ACL5 gene is tightly regulated to control thermospermine levels, influencing vascular development and stem elongation in Arabidopsis thaliana.[41]Polyamine biosynthesis is subject to stringent regulation, primarily through feedback inhibition by the end products themselves on key enzymes like ODC and SAMDC, preventing overaccumulation and maintaining cellular homeostasis.[42] In bacteria, polyamines further modulate gene expression at the translational level, enhancing the synthesis of a coordinated set of proteins involved in growth and stress response, collectively termed the polyamine modulon.[42]Recent studies have highlighted the potential of ODC inhibitors, such as α-difluoromethylornithine (DFMO), in disrupting polyamine biosynthesis in fungi, where ODC represents the sole pathway for putrescine production, offering insights into antifungal strategies against plant pathogens like Phytophthora infestans.[43]
Catabolism and Homeostasis
Polyamine catabolism primarily involves oxidative deamination mediated by specialized enzymes that degrade these molecules into lower-order polyamines, aldehydes, and reactive oxygen species. In mammals and other eukaryotes, flavin-dependent polyamine oxidases (PAOs), such as spermine oxidase (SMO) and polyamine oxidase (PAOX), catalyze the terminal catabolism of spermine and spermidine. For instance, SMO oxidizes spermine to spermidine, 3-aminopropanal, and hydrogen peroxide (H₂O₂) via the reaction:\text{spermine} + \text{O}_2 + \text{H}_2\text{O} \rightarrow \text{spermidine} + 3\text{-aminopropanal} + \text{H}_2\text{O}_2This process generates cytotoxic byproducts like 3-aminopropanal, which can form acrolein, contributing to oxidative stress if unregulated.[44] In plants and bacteria, copper-containing amine oxidases (CuAOs) perform similar oxidations on putrescine and other diamines, producing H₂O₂ and aldehydes such as 4-aminobutanal, which support cell wall reinforcement and pathogen defense.[45] These enzymatic activities ensure the turnover of excess polyamines, preventing toxic accumulation while recycling nitrogen for biosynthesis.[46]Cellular polyamine homeostasis is maintained through intricate regulatory mechanisms that balance synthesis, degradation, export, and uptake. Polyamine transport systems, including ATP-binding cassette (ABC) transporters and solute carrier (SLC) proteins like SLC18B1 (vesicular polyamine transporter), facilitate the influx and efflux of polyamines to fine-tune intracellular levels.[47] A key feedback loop involves ornithine decarboxylase antizyme (AZ), which binds and inhibits ornithine decarboxylase (ODC), the rate-limiting enzyme in polyamine biosynthesis, while also promoting ODC degradation via the proteasome and suppressing polyamine uptake.[48] Additionally, spermidine/spermine-N¹-acetyltransferase (SSAT) acetylates spermidine and spermine at the N¹ position, marking them for export or subsequent oxidation by PAOX, thereby regulating polyamine pools and preventing overload.[49] These mechanisms collectively ensure polyamine concentrations remain within a narrow physiological range essential for cell viability.The polyamine cycle integrates biosynthesis, acetylation, oxidation, and recapture to sustain dynamic equilibrium. Acetylated polyamines produced by SSAT are either exported or oxidized back to putrescine and spermidine, allowing recapture into biosynthetic pathways and nitrogen recycling. H₂O₂ generated during catabolism acts as a signaling molecule, triggering stress responses such as antioxidant defense activation and programmed cell death in plants under abiotic stresses like drought or salinity. Dysregulation of this cycle leads to pathological states; for example, cancer cells often exhibit polyamine overaccumulation due to upregulated transport and reduced catabolism, fueling proliferation. As of 2025, targeting polyamine catabolism has shown promise in enhancing ferroptosis to suppress tumor growth.[50] A 2019 study highlights SSAT upregulation as a compensatory response in tauopathy models of neurodegeneration, mitigating polyamine stress but potentially exacerbating oxidative damage if unchecked.[51][52][53][54]
Biological Functions
Cellular and Molecular Roles
Polyamines play essential roles in cellular and molecular processes, primarily due to their polycationic nature at physiological pH, which enables strong electrostatic interactions with negatively charged biomolecules. Intracellular concentrations of polyamines, such as putrescine, spermidine, and spermine, typically range from 0.1 to 5 mM, varying by cell type and proliferative state, with higher levels in rapidly dividing cells like those in the intestinal epithelium or immune cells.[55][56] These concentrations allow polyamines to stabilize macromolecules and modulate key pathways, supporting cellular homeostasis and function.A primary function of polyamines is the stabilization of nucleic acids through electrostatic binding to the phosphate backbone of DNA and RNA, which neutralizes negative charges and promotes structural compaction. For DNA, this interaction facilitates chromatin condensation, reducing electrostatic repulsion and enabling compact forms essential for packaging and protection.[57][58] In RNA, polyamines similarly stabilize structures; spermidine, in particular, binds to 16S rRNA in the small ribosomal subunit, aiding in the correct folding of helix 44 near the decoding center and maintaining ribosome integrity during translation.[59][60]Polyamines also drive cell growth and proliferation by influencing translation and ion channel activity. They are critical for the post-translational modification of eukaryotic initiation factor 5A (eIF5A), where spermidine donates a butylamine group to form hypusine at a specific lysine residue, enabling eIF5A to facilitate translation elongation, particularly of polyproline motifs, and linking polyamine levels directly to protein synthesis rates.[61][62] Additionally, intracellular polyamines like spermine modulate ion channels, including NMDA receptors, where they act as positive allosteric modulators to enhance channel opening and calcium influx, thereby regulating neuronal excitability and synaptic plasticity.[63][64]In regulating cell death pathways, polyamines exhibit anti-apoptotic effects by inhibiting caspase activation, particularly caspase-3, which prevents proteolytic cascades leading to programmed cell death.[65] Conversely, spermidine promotes autophagy, a cytoprotective process, by inhibiting the acetyltransferase activity of EP300, which deacetylates autophagy-related proteins like ATG5, ATG7, and ATG12, enhancing autophagosome formation; this mechanism was first elucidated in the early 2010s and has been corroborated in recent clinical trials assessing spermidine's autophagy-inducing potential in humans.[66][67][68] Beyond these, polyamines contribute to cytoplasmic pH buffering, where their protonation states help maintain intracellular pHhomeostasis, supporting enzyme activity and cell survival under stress.[69] They also exert antioxidant effects by directly scavenging hydrogen peroxide and other reactive oxygen species, mitigating oxidative damage to cellular components.[70][71]
Role in DNA Repair
Polyamines play a critical role in the DNA damage response, particularly by promoting homology-directed repair (HDR) of double-strand breaks (DSBs). Spermidine and spermine, the predominant cellular polyamines, enhance the formation and stability of RAD51 nucleoprotein filaments on single-stranded DNA (ssDNA), facilitating the search for homologous sequences and strand invasion during HDR.[72] This stimulation occurs through polyamines' ability to promote the capture of homologous duplex DNA by the RAD51-ssDNA filament, thereby increasing synaptic complex assembly and DNA strand exchange activity.[72] Additionally, polyamines stabilize interactions between BRCA2 and RAD51, supporting presynaptic filament assembly without altering BRCA2 expression levels.[72]At the molecular level, polyamines exert these effects via cationic bridging of DNA strands, leveraging their positively charged amine groups to neutralize phosphate backbones and condense DNA structures, which aids in aligning repair substrates.[72] The binding affinity of polyamines to DNA typically falls in the range of 10-50 μM (Kd), enabling physiological concentrations to influence repair dynamics without excessive aggregation.[73] Polyamines do not significantly affect non-homologous end joining (NHEJ), as demonstrated by reporter assays showing no change in NHEJ efficiency upon polyamine modulation.[72]Beyond HDR, polyamines contribute to DNA repair by protecting against oxidative damage. Spermine acts as a free radical scavenger, directly quenching reactive oxygen species (ROS) such as hydroxyl radicals to prevent base modifications and strand breaks induced by oxidative stress.[70] Spermidine and spermine differentially mediate this protection, with spermine showing stronger inhibition of ROS-mediated DNA oxidation in cellular models.[74]Experimental evidence underscores these roles: depletion of polyamines using the ornithine decarboxylase inhibitor difluoromethylornithine (DFMO) impairs HDR in a dose-dependent manner and sensitizes cells to ionizing radiation by increasing DSB accumulation and apoptosis.[72] This sensitization manifests as reduced cell proliferation and heightened genomic instability, highlighting polyamines' necessity for radiation resistance and genome maintenance.[72]
Functions in Plants and Microorganisms
In plants, thermospermine plays a critical role in regulating xylem development by suppressing excessive differentiation of xylem vessels. The ACL5 gene encodes thermospermine synthase, and its loss-of-function mutants exhibit dwarfism accompanied by overproliferation of xylem tissues, highlighting thermospermine's function in maintaining vascular balance during growth.[41] Additionally, putrescine and spermidine contribute to abiotic stress tolerance, particularly under drought and salt conditions, by enhancing reactive oxygen species (ROS) scavenging through upregulation of antioxidant enzymes like superoxide dismutase and catalase.[75] These polyamines also inhibit leaf senescence by preserving chlorophyll and protein levels, thereby extending photosynthetic activity and delaying age-related tissue degradation.[76]In microorganisms, polyamines support adaptation to environmental stresses. In bacteria such as Escherichia coli, polyamines like putrescine and spermidine protect against acid stress by inducing glutamate decarboxylase activity, which helps maintain intracellular pH, and by modulating membrane permeability through porin blockade to stabilize cellular integrity.[77]Polyamines also mediate symbiotic interactions between plants and microorganisms. In rhizobia-legume symbioses, polyamines such as putrescine and spermidine are essential for nodule formation and nitrogen fixation, where exogenous application enhances nodulation under stress and supports bacteroid differentiation within root nodules.[78] Similarly, in algae, spermidine protects photosynthetic machinery from oxidative damage, maintaining photosystem II integrity and enhancing biomass yield under high light or CO₂ stress by bolstering antioxidant defenses. In microalgae, polyamines including spermidine are associated with light-harvesting complex II and chlorophyll proteins, with levels increasing under light stress to aid adaptation.[79]Recent research underscores evolving insights into polyamine functions in these systems. A 2023 preprint showed that spermine and spermidine inhibit or induce programmed cell death in Arabidopsis thaliana in a dose-dependent manner, contributing to plant defense responses.[80] In 2025, investigations into the gut microbiome revealed that bacterial polyamines, produced de novo by commensal species, influence host physiology by altering epithelial barrier function and immune homeostasis through microbial-host metabolic crosstalk.[81] Notably, polyamine profiles differ between plants and bacteria: plants exhibit higher levels of thermospermine, which is integral to developmental signaling, whereas agmatine predominates in many bacterial species as a precursor and stress protectant.[82]
Roles in Health and Disease
Involvement in Cancer
Polyamines exhibit a complex involvement in cancer, primarily promoting tumor progression through dysregulated biosynthesis and metabolism while also presenting opportunities for therapeutic intervention via depletion strategies. Elevated ornithine decarboxylase (ODC) activity, the rate-limiting enzyme in polyamine synthesis, is observed in many human cancers, driving increased intracellular polyamine levels that support rapid cell proliferation and survival. This upregulation is frequently linked to the MYConcogene, which is overexpressed in about 70% of cancers and directly transcriptionally activates ODC, thereby enhancing polyamine production and contributing to oncogenic transformation. For instance, spermidine facilitates tumor cell proliferation by hypusinating eukaryotic translation initiation factor 5A (eIF5A), a process essential for protein synthesis and cell growth in malignant cells.[9]In specific cancers, such as prostate and breast tumors, plasma spermine levels have been measured in patients with benign and malignant disease, with limited elevation observed in some breast cancer cases but no significant differences or correlations with disease progression.[83] Polyamines also enhance metastasis by promoting epithelial-mesenchymal transition (EMT), a key process enabling cancer cells to acquire migratory and invasive properties; for example, increased putrescine availability has been shown to augment EMT through epigenetic modifications like H3K27 acetylation.[84] Mechanistically, dysregulated polyamine homeostasis leads to genomic instability, as excessive polyamines induce DNA damage and chromosomal aberrations, further fueling malignant evolution.[9] Additionally, oxidation products from polyamine catabolism, such as hydrogen peroxide (H₂O₂) and acrolein generated by spermine oxidase (SMOX), act as reactive species that cause oxidative stress and DNA lesions, paradoxically contributing to carcinogenesis despite their potential pro-apoptotic effects at high concentrations.[85]Conversely, polyamine depletion holds anti-tumor potential, as demonstrated by difluoromethylornithine (DFMO), an irreversible ODC inhibitor that reduces polyamine levels and inhibits tumor growth in preclinical and clinical settings.[9] ODC serves as a reliable biomarker for tumor prognosis, with high expression levels associated with aggressive phenotypes and worse outcomes across multiple cancer types.[86] In colorectal cancer, a phase III randomized, double-blind, placebo-controlled trial combining low-dose DFMO with sulindac (a COX inhibitor) significantly reduced the risk of recurrent adenomas by 70% (relative risk 0.30; 95% CI, 0.18-0.49), highlighting the efficacy of targeting polyamine metabolism for chemoprevention without substantial toxicity.[87] Recent advances as of 2025 include phase I trials of polyamine transport inhibitors like AMXT 1501 dicaprate, showing promise in reducing immunosuppression in the tumor microenvironment.[88] These findings underscore polyamines' pro-tumorigenic dominance in cancer but affirm their viability as therapeutic targets, particularly when combined with agents that address compensatory pathways like polyamine uptake or catabolism.[85]
Polyamines in Aging and Neurodegeneration
Polyamines, particularly spermidine, have been implicated in mitigating age-related decline through the induction of autophagy, a cellular process that promotes longevity across model organisms. Administration of spermidine extends lifespan in yeast, nematodes, flies, and mice by enhancing autophagic flux, which helps clear damaged cellular components and reduces oxidative stress associated with aging.[89] This mechanism involves hypusination of eIF5A and subsequent deacetylation of eIF2α, facilitating protein synthesis regulation critical for autophagosome formation.[90] In mammalian models, spermidine supplementation similarly preserves cardiac function and delays age-related pathologies, underscoring its geroprotective potential.[91]Human epidemiological data further support spermidine's role in healthy aging. A prospective cohort study involving over 800 participants found that higher dietary spermidine intake was associated with a 20-40% reduction in all-cause mortality over 20 years, with specific links to lowered cardiovascular disease risk through improved endothelial function and reduced inflammation.[92] More recent analyses from 2023 cohorts, including the Bruneck Study follow-up, confirmed that elevated spermidine levels from dietary sources correlate with decreased cardiovascular mortality, emphasizing the polyamine's translational relevance from preclinical to human longevity.[93]In the context of neurodegeneration, polyamines exhibit both neuroprotective and dysregulatory effects depending on disease state. Spermine acts as a positive allosteric modulator of NMDA receptors, enhancing glutamate signaling that is often impaired in Alzheimer's disease (AD); this modulation is altered in AD brain tissue, contributing to synaptic dysfunction and cognitive deficits.[94] In Parkinson's disease (PD), overexpression of spermidine/spermine N1-acetyltransferase (SSAT) leads to depletion of higher-order polyamines like spermine, exacerbating α-synuclein aggregation and dopaminergic neuron loss, as observed in transgenic models and patient-derived cells.[95] Conversely, spermine demonstrates protective effects against tau pathology in AD by inhibiting higher-order tau aggregation and promoting fibril disassembly, thereby reducing neurofibrillary tangle formation in cellular assays.[96]Baseline polyamine concentrations in the brain range from 1 to 5 mM, with spermine and spermidine predominating in neurons and glia to support synaptic plasticity and ion channel function. Age-related declines in these levels, often exceeding 50% by late life, strongly correlate with neurodegenerative progression, including amyloid-β accumulation and mitochondrial dysfunction in AD and PD.[97] This depletion disrupts autophagy—detailed in cellular roles elsewhere—and exacerbates proteotoxic stress, linking polyamine homeostasis directly to brain aging trajectories.[98]Recent clinical advances highlight spermidine's therapeutic promise for cognitive health, though results are mixed. The SmartAge trial (NCT03094546), a randomized controlled phase 2b study in older adults with subjective cognitive decline, found no significant effect of 12 months of spermidine supplementation (1 mg/day from wheat germ extract) on episodic memory performance compared to placebo.[99] A 2025 review of interventional trials noted preliminary positive effects on cognition in three of four studies, suggesting potential benefits in select populations for slowing mild cognitive impairment progression.[100] Circulating acetylated polyamines have been associated with COVID-19 severity in cancer patients, but links to long COVID neuroinflammatory symptoms remain exploratory.[101]Therapeutically, spermidine supplementation from natural sources like wheat germ—containing up to 24 mg/100 g—offers a safe approach to counteract age-related declines without synthetic analogs.
Polyamine Analogues and Applications
Development of Analogues
The development of polyamine analogues began in the late 1980s, driven by the recognition that natural polyamines such as spermidine and spermine are essential for rapid cell proliferation in tumors, prompting efforts to create structural mimics that disrupt polyamine metabolism.[102] These analogues are designed to interfere with key enzymes in the polyamine pathway, including ornithine decarboxylase (ODC) and spermidine/spermine N1-acetyltransferase (SSAT), by incorporating modifications that enhance binding affinity or induce conformational changes. For instance, fluorination at the alpha position of ornithine yields irreversible ODC inhibitors, while alkyl substitutions on the polyamine backbone can upregulate SSAT activity, leading to polyamine catabolism and depletion of intracellular pools.[102] A notable example is N1,N12-bis(ethyl)spermine (BESpm), a symmetric analogue of spermine that competitively traps natural polyamines, displacing them from cellular targets and suppressing biosynthetic enzyme levels such as ODC.[103]Synthesis of these analogues typically starts from natural polyamine scaffolds like putrescine or spermine, modified through selective alkylation to introduce ethyl or other alkyl groups at terminal nitrogens, which alters their charge distribution and metabolic stability.[102] This approach allows for the creation of conformationally restricted variants, such as those with unsaturated bonds or cyclic elements, to optimize interactions with polyamine transport proteins or enzymes. To accelerate structure-activity relationship (SAR) studies, combinatorial libraries have been employed, using solid-phase synthesis to generate diverse alkylated derivatives screened for antitumor potency and enzyme inhibition.[104] These methods enable high-throughput evaluation, identifying leads with improved selectivity over natural polyamines.Prominent analogues include α-difluoromethylornithine (DFMO), an early ODC inhibitor featuring a difluoromethyl group that mimics the ornithine substrate and forms a covalent adduct with the enzyme's active site, effectively halting polyamine biosynthesis from the initial step.[105] Another key compound is PG-11047, an unsaturated analogue of spermine with ethyl substitutions and a cis double bond, designed to bind DNA more avidly than endogenous polyamines, thereby interfering with nucleic acid-dependent processes in proliferating cells.[106] By the 2020s, research has evolved toward advanced derivatives, incorporating features for targeted activation in tumor microenvironments, building on the foundational cancer-focused efforts of the 1980s.[102]Despite these advances, challenges persist, particularly the risk of toxicity arising from excessive polyamine depletion, which can disrupt normal cellular homeostasis and induce apoptosis or necrosis in non-target tissues.[107] Recent structural biology efforts have provided insights into polyamine enzyme inhibition to guide the design of more selective analogues to mitigate off-target effects.
Therapeutic and Industrial Uses
Polyamines and their analogues have found significant applications in therapeutics, particularly through polyamine depletion strategies that target dysregulated metabolism in diseases. Eflornithine (DFMO), an irreversible inhibitor of ornithine decarboxylase, is a cornerstone treatment for second-stage human African trypanosomiasis (sleeping sickness), where it is used in combination with nifurtimox as the nifurtimox-eflornithine combination therapy (NECT); this regimen is listed on the World Health Organization's Model List of Essential Medicines for its efficacy in reducing treatment duration and improving outcomes in resource-limited settings.[108][109]In oncology, DFMO has gained traction for high-risk neuroblastoma, with FDA approval of oral eflornithine (branded as IWILFIN) in December 2023 to reduce relapse risk in pediatric and adult patients following standard therapy; clinical data from a phase II trial demonstrate improved 4-year event-free survival of 84% (versus 73% in external controls) as maintenance therapy following standard treatment including immunotherapy.[110][111] Polyamine-depleting combinations, such as DFMO with celecoxib, are under investigation in phase I studies for relapsed neuroblastoma, showing synergistic tumor suppression through ornithine decarboxylase inhibition and cyclooxygenase-2 blockade.[112] Additionally, a phase II trial of the polyamine analogue N1,N11-diethylnorspermine (DENSPM) in previously treated metastatic breast cancer reported no partial responses and stable disease in up to 40% of patients after initial cycles, though all patients progressed by 4 months, highlighting potential in combination regimens despite dose-limiting toxicities.[102][113]Beyond depletion, polyamine analogues exhibit targeted effects in specific cancers and neurological disorders. In estrogen receptor-positive breast cancer, analogues such as bis(ethyl)polyamines down-regulate estrogen receptor α expression and activity, reversing hormone-dependent growth in preclinical models and suggesting utility in overcoming endocrine resistance.[114] For epilepsy, endogenous polyamines like spermine modulate ion channels, including NMDA receptors and persistent sodium currents; therapeutic modulation via polyamine supplementation or analogues reduces hyperexcitability in rodent models, with potential to enhance anticonvulsant responses by restoring intracellular polyamine levels depleted in chronic epilepsy.[115][63]Spermidine, a natural polyamine, is increasingly used in supplements promoted for longevity, leveraging its role in autophagy induction; the global spermidine supplement market, valued at approximately USD 175 million in 2024, is projected to exceed USD 500 million by 2032 due to rising demand for anti-aging interventions supported by epidemiological data linking higher dietary spermidine intake to reduced mortality.[116]Industrially, polyamines serve as chelating agents in water treatment, where polyamine-modified clays and resins efficiently remove heavy metals like copper and lead from wastewater through strong coordination complexes, achieving adsorption capacities up to 200 mg/g in low-cost hybrid materials.[117] In drug delivery, polyethylenimine (PEI)-based polymers are widely employed for siRNA transfection, with 2024 optimizations incorporating lipid hybrids and microfluidic formulations enhancing delivery efficiency by 2-5 fold while reducing cytotoxicity in cancer cell lines.[118]Emerging applications include microbiome therapeutics, where bacterial polyamine modulators are being explored for irritable bowel syndrome (IBS); 2025 studies indicate that gut microbiota-derived polyamines influence intestinal barrier function and inflammation, with interventions targeting polyamine biosynthesis pathways showing promise in alleviating IBS symptoms through restored microbial homeostasis.[119] Overall, these uses underscore the versatility of polyamines, though ongoing phase II/III trials emphasize the need for refined dosing to balance efficacy and safety.