Fact-checked by Grok 2 weeks ago

Monocarboxylate transporter 1

Monocarboxylate transporter 1 (MCT1), encoded by the SLC16A1 gene on chromosome 1p13.2, is a that catalyzes the proton-linked transport of monocarboxylates such as L-lactate, pyruvate, and across the plasma membrane in a bidirectional manner driven by concentration and pH gradients. As a member of the solute carrier family 16 (SLC16), MCT1 features a characteristic structure with 12 transmembrane helices, intracellular N- and C-termini, and a large intracellular loop between transmembrane domains 6 and 7, which is essential for its function. It requires association with ancillary proteins, such as CD147 (basigin) or embigin, for proper trafficking to and stability at the cell surface. MCT1 is widely expressed in various tissues, including erythrocytes, skeletal muscle, heart, brain, and tumors, where it plays critical roles in cellular energy metabolism and homeostasis by facilitating the influx or efflux of metabolic substrates. In the , MCT1 is pivotal in the astrocyte-neuron lactate shuttle by exporting produced by , which is then taken up by neurons primarily via MCT2 for oxidative metabolism, thereby supporting , formation, and neuronal energy demands during high activity. Dysregulation of MCT1 expression or function has been linked to metabolic disorders, such as erythrocyte lactate transporter defect, and it serves as a therapeutic target due to its overexpression in many cancers. In cancer, MCT1 contributes to the Warburg effect by enabling export from glycolytic tumor cells, acidifying the extracellular microenvironment to promote and suppress immune responses, while also supporting uptake in oxidative tumor cells and endothelial cells to fuel and . Its inhibition has shown promise in preclinical studies for disrupting tumor and enhancing the of chemotherapeutics, highlighting MCT1's dual role as a prognostic and drug target across malignancies like , , and . As of 2025, MCT1 inhibitors such as AZD3965 are in early-phase clinical trials.

Genetics and molecular biology

Gene structure

The SLC16A1 gene, which encodes the monocarboxylate transporter 1 (MCT1), is located on the short arm of human at the 1p13.2 cytogenetic band, with genomic coordinates spanning from 112,911,847 to 112,956,196 on the reverse strand (GRCh38 assembly). This gene covers approximately 44 kb of genomic DNA and consists of 5 s, with the first being noncoding and the first exceeding 26 kb in length. The coding sequence begins in 2, and the gene's structure reflects its role in the solute carrier family 16 (SLC16), a group of proton-linked transporters. The promoter region of SLC16A1 lacks a and includes potential binding sites for several transcription factors, such as factor-3β (HNF-3β), which regulates its tissue-specific expression. Mutational analysis has identified activating variants in this promoter, including a 25-bp insertion at position -24 relative to the transcription start site, that disrupt silencer elements and lead to in pancreatic beta cells, contributing to exercise-induced hyperinsulinemic . Evolutionarily, SLC16A1 exhibits high conservation across mammals, sharing 86% identity with its ortholog, underscoring its essential role in monocarboxylate transport. The SLC16 family, including SLC16A1, represents a distinct class of solute carriers with no close resemblance to other known transporter proteins, but distant homologs exist in . Pathogenic in SLC16A1 have been linked to rare genetic disorders affecting and body handling (see Pathological implications).

Protein isoforms

The SLC16A1 gene, located on chromosome 1p13.2, encodes the monocarboxylate transporter 1 (MCT1) protein, a member of the solute carrier family 16. The canonical isoform comprises 500 amino acids with a calculated molecular weight of 53,943 Da and an apparent molecular weight of approximately 49 kDa on SDS-PAGE. The protein features 12 predicted transmembrane helices arranged in an N-terminal intracellular and C-terminal intracellular topology, characteristic of the major facilitator superfamily. Alternative splicing of SLC16A1 transcripts yields multiple variants, including two principal protein-coding isoforms documented in human tissues. The canonical isoform (UniProt P53985-1) represents the full-length sequence essential for standard membrane integration, whereas isoform 2 (P53985-2) is a truncated variant of 458 arising from , which may impair plasma membrane targeting and functional localization. Ensembl annotations indicate up to 22 transcripts overall, though many are non-coding or tissue-specific, contributing to regulatory diversity without altering the core . MCT1 undergoes limited post-translational modifications, with potential N-glycosylation consensus sites at Asn-73 and Asn-256 in the first extracellular loop; however, functional studies demonstrate that these sites are not utilized, as PNGase F deglycosylation fails to shift the protein's electrophoretic mobility, confirming MCT1 as non-glycosylated. This lack of glycosylation distinguishes MCT1 from its ancillary partners and supports its stability in the membrane environment. Expression and proper subcellular trafficking of MCT1 depend on association with ancillary chaperone proteins, notably basigin (BSG, also known as CD147), which binds isoform 2 of BSG to stabilize MCT1 and promote its delivery to the plasma membrane; without this interaction, MCT1 remains retained intracellularly and non-functional. Embigin (EMB) serves a similar role in certain cell types, underscoring the reliance on these partners for biophysical maturation.

Structure

Tertiary structure

The tertiary structure of monocarboxylate transporter 1 (MCT1), also known as SLC16A1, belongs to the major facilitator superfamily (MFS) of transporters and consists of 12 transmembrane α-helices (TMs) organized into two bundles of six helices each, forming a central substrate translocation pathway. The N- and C-termini are located intracellularly, with a large intracellular loop between TM6 and TM7. This was resolved through cryo-electron (cryo-EM) studies of MCT1 in with the chaperone basigin-2 (also known as CD147 or EMMPRIN), achieving resolutions of 3.0–3.3 . The core fold features an inverted topology typical of MFS proteins, with the TM bundles exhibiting pseudo-twofold symmetry that alternates between outward- and inward-facing conformations during the transport cycle. Key structural motifs include the substrate-binding pocket, which is centrally located and lined by residues from TM1, TM8, and TM10, such as Tyr34 and Lys38 (TM1), Asp309 and Arg313 (TM8), and Phe367 and Ser371 (TM10). This pocket accommodates monocarboxylates like , with the carboxylate group forming salt bridges to Lys38 and Arg313, while the hydroxyl group of interacts with Ser371. The proton-binding site is primarily mediated by Asp309 in TM8, which serves as the proton-coupling residue essential for symport activity; in the outward-open state, Asp309 is positioned to coordinate proton transfer. These motifs were elucidated from structures bound to (PDB: 6LZ0) or inhibitors mimicking substrate binding. MCT1 adopts distinct conformational states during transport: an outward-open conformation observed in structures with or inhibitors like AZD3965 (PDB: 6LYY) and BAY-8002 (PDB: 7CKR), where the substrate-binding site is accessible from the extracellular side and the intracellular gate is closed; and an inward-open conformation captured with the inhibitor 7ACC2 (PDB: 7CKO) or the Asp309Asn mutant (PDB: 7DA5), featuring an open intracellular vestibule and occluded extracellular access. These states reflect the rocker-switch mechanism of MFS transporters, enabling alternating access. Structural comparisons to homologs highlight conserved yet distinct features; for instance, human MCT1 shares the MFS fold with the bacterial monocarboxylate transporter SfMCT from Syntrophobacter fumaroxidans (PDB: 6HCL), but exhibits major differences in the outward-facing arrangement of TM helices, particularly in the positioning of the pocket relative to the . Similarly, MCT1 aligns closely with human MCT2 (PDB: 7BP3) in the inward-open state, with root-mean-square deviations of approximately 1.5 Å for core TMs, underscoring family-specific adaptations for proton-coupled transport.

Oligomerization and chaperones

Monocarboxylate transporter 1 (MCT1) can assemble into homodimers in the absence of ancillary proteins, with the dimer interface primarily mediated by interactions between transmembrane helices, including hydrophobic contacts and hydrogen bonds involving helices 5 and 6. This oligomeric state is inferred from structural analogies to related family members like MCT2, where cryo-EM reveals a homodimeric architecture stabilized by crossover of transmembrane helix 5 between subunits and inter-subunit cation-π interactions at the helix 5-6 region. However, physiological assembly favors heterodimeric complexes with chaperone proteins rather than homodimers, as these partners sterically hinder self-association and promote functional maturation. Essential chaperones such as basigin (also known as CD147) and embigin (gp70) are required for the proper folding, trafficking, and plasma membrane insertion of MCT1. These single-transmembrane glycoproteins form tight heterodimers with MCT1, facilitating its exit from the () and preventing retention in intracellular compartments. In the absence of basigin or embigin, MCT1 accumulates in the ER/Golgi network, resulting in minimal surface expression and loss of transport activity, as demonstrated by co-transfection studies in heterologous cells. Basigin is the predominant chaperone in most tissues, associating specifically with MCT1 (and MCT4) via its extracellular immunoglobulin-like domains and a single transmembrane , while embigin serves a similar role in select contexts like brain and muscle. Cryo-EM structures of the MCT1-chaperone complexes provide detailed insights into the dimer interfaces. In the MCT1-embigin heterodimer (resolved at 3.6 ), embigin's engages MCT1's 6 through extensive hydrophobic interactions, a Tyr-Arg , and a Glu-Arg , while also inducing conformational changes in 5 to straighten it and prevent homodimerization via steric occlusion at the TM8 region. Similarly, the MCT1-basigin complex (at 3.0–3.3 ) features basigin's transmembrane contacting MCT1's 6 via hydrophobic packing and a key between Glu218 (basigin) and Asn187 (MCT1), with the 5-6 bundle contributing to overall stability through intra- and inter-molecular like Arg143-Glu376-Asp380. These contacts ensure the chaperone remains bound at the plasma membrane, distinct from transient ER interactions. Association with chaperones and resulting oligomeric states significantly influence MCT1's transport properties. The MCT1 homodimer exhibits higher substrate affinity (e.g., L- K_d of 1.8 mM) and enhanced coupling efficiency compared to the chaperone-bound heterodimer, which transitions to a "decoupled" state with reduced affinity (K_d of 8.7 mM) and lower overall transport rates due to altered 5 dynamics and proton-substrate coordination. This modulation arises from chaperone-induced conformational shifts that prioritize localization over maximal kinetic efficiency, ensuring regulated lactate/pyruvate flux in tissues without compromising specificity.

Biochemical function

Transport mechanism

Monocarboxylate transporter 1 (MCT1) facilitates the translocation of monocarboxylates across the plasma membrane through an ordered bi-bi kinetic , in which a proton binds first to the transporter on the extracellular side, inducing a conformational change that subsequently allows binding of the monocarboxylate anion. This sequential binding is followed by a further conformational shift that translocates the proton-monocarboxylate complex across the membrane, with release occurring in reverse order on the intracellular side. The rate-limiting step in this process is the conformational change associated with reorientation of the loaded transporter. The transport is electroneutral, involving a 1:1 of protons to monocarboxylate anions, which ensures no net charge movement and renders the process primarily driven by the transmembrane gradient (ΔpH) rather than the . For , a key , the apparent Michaelis constant (K_m) is approximately 3-10 , reflecting moderate suited to physiological concentrations during metabolic stress. The kinetics show high affinity for protons, with the overall process tightly coupled to local proton availability. MCT1 supports both net flux and modes of transport, with homo- or hetero- (such as lactate-pyruvate swapping) occurring 5-10 times faster than net uptake or efflux due to the accelerated reorientation of the empty (proton-free) transporter back to the outward-facing conformation. This disparity arises because net transport requires slower return steps without bound substrates, making particularly efficient for maintaining intracellular during fluctuating levels. Activity of MCT1 exhibits strong pH dependence, with optimal function at mildly acidic extracellular (6.0-6.5), where proton availability enhances and translocation rates while lowering the apparent K_m for monocarboxylates. At neutral or alkaline , transport is markedly inhibited due to reduced proton , underscoring the transporter's role in acid-base coupled handling.

Substrate specificity

Monocarboxylate transporter 1 (MCT1) exhibits specificity for a range of short-chain monocarboxylates, facilitating their proton-coupled symport across membranes. Primary physiological substrates include L-lactate, pyruvate, and such as D-3-hydroxybutyrate and acetoacetate. These molecules are transported with affinities in the millimolar range, reflecting MCT1's role in handling metabolic intermediates during conditions like exercise or . The affinity for L-lactate is typically characterized by a K_m of 3–10 mM, with values around 4.5–4.7 mM reported in erythrocyte and models. Pyruvate is transported with a K_m of approximately 1 mM, though measurements vary from 0.7 to 2 mM depending on the expression system. For , D-3-hydroxybutyrate has a K_m of about 3–12.5 mM, while acetoacetate shows slightly lower affinity at around 5–10 mM. These K_m values indicate comparable transport efficiencies for and pyruvate under physiological concentrations.
SubstrateK_m (mM)Source
L-Lactate3–10PubMed 8557697
Pyruvate0.7–2PubMed 7818477
D-3-Hydroxybutyrate3–12.5PubMed 8779821
Acetoacetate5–10PubMed 8779821
MCT1 demonstrates , preferentially transporting the L-enantiomer of with a K_m approximately 5–6 times lower than for D-lactate (27–28 mM). Pyruvate, being achiral, lacks enantiomeric preference. Secondary substrates include like butyrate and aromatic monocarboxylates such as nicotinate, both with affinities in the low millimolar range (e.g., nicotinate K_m ~4 mM). Certain drugs, including , interact weakly as substrates or competitive inhibitors with K_i > 1 mM. High-affinity inhibitors like AR-C155858, with an IC_{50} or K_i of ~2 nM, compete directly at the substrate-binding site, overlapping with monocarboxylate recognition. This overlap underscores the structural conservation of the binding pocket for both endogenous substrates and select xenobiotics.

Physiological roles

Tissue distribution

Monocarboxylate transporter 1 (MCT1), encoded by the SLC16A1 gene, exhibits high expression across multiple tissues, reflecting its role in monocarboxylate homeostasis. In humans, MCT1 mRNA levels are particularly elevated in , with median transcripts per million (TPM) values of 23.8 according to GTEx data, followed by heart (11.9 TPM) and tissues (2.2 TPM). Protein expression, confirmed via , is prominent in cardiac myocytes and oxidative fibers, such as type I fibers. MCT1 is also abundantly expressed in erythrocytes, where it facilitates lactate efflux, and in the , particularly the colon mucosa, supporting nutrient absorption. In the liver, MCT1 localizes to hepatocytes, aiding in body and handling. Within the , MCT1 is detected in , , and select neuronal populations, with strong immunostaining in glial cells associated with blood vessels. Functional MCT1 expression in these sites often correlates with basigin (CD147) co-localization, which is essential for its plasma membrane trafficking. Developmentally, MCT1 expression in the brain increases postnatally, peaking around day 15 to support body utilization during when glucose demands rise. This upregulation occurs primarily in the blood- barrier endothelium and , transitioning from low embryonic levels. Expression patterns of MCT1 are conserved between humans and mice, with comparable high levels in heart, , and .

Metabolic functions

Monocarboxylate transporter 1 (MCT1), encoded by the SLC16A1 gene, plays a central role in the shuttle system, facilitating the efflux of from glycolytic cells such as and white fibers, and its uptake into oxidative tissues like and cardiac myocytes for conversion to pyruvate and subsequent oxidation in the tricarboxylic acid cycle. In the , MCT1 enables the transfer of produced by during to neurons, where it serves as an to support synaptic activity and processes such as formation. Similarly, in the heart, MCT1 mediates influx from circulating sources or adjacent glycolytic cells like fibroblasts, enhancing ATP production during periods of high demand, such as exercise. This bidirectional transport maintains and supports metabolic between cell types under physiological conditions. Recent research has shown that MCT1 regulates in , influencing exercise activity and endurance (as of 2024). MCT1 also contributes to ketone body handling, particularly during fasting states when hepatic ketogenesis increases to provide alternative fuels. In the liver, MCT1 facilitates the export of ketone bodies, such as β-hydroxybutyrate, from hepatocytes into the bloodstream, enabling their distribution to peripheral tissues. This transport is crucial for peripheral uptake in energy-demanding organs, including the , where MCT1 expressed on endothelial cells and supports ketone body delivery across the blood-brain barrier. In neonates, during when glucose availability is limited, MCT1-mediated ketone body transport becomes especially vital for energy , as ketone bodies serve as primary substrates for lipid synthesis and neuronal function. Through its proton-linked symport mechanism, MCT1 couples monocarboxylate transport with H⁺ flux, contributing to homeostasis in tissues like . At rest, MCT1 promotes H⁺ influx to stabilize , while during exercise onset, it facilitates H⁺ efflux alongside export, helping to buffer extracellular generated by glycolytic activity. Studies in MCT1 haploinsufficient mice demonstrate that reduced MCT1 expression leads to greater acidification at the start of , underscoring its role in rapid proton extrusion and maintenance of contractile function. Beyond endogenous substrates, MCT1 has a minor role in clearance, particularly in the liver, where it transports certain drugs like valproic acid across membranes to support their and elimination. This function aids in the hepatic handling of monocarboxylate-like pharmaceuticals, though it is secondary to its primary metabolic roles.

Regulation

Transcriptional and epigenetic control

The expression of the SLC16A1 gene, encoding monocarboxylate transporter 1 (MCT1), is tightly regulated at the transcriptional level by several key transcription factors responsive to physiological and environmental cues. alpha (PPAR-α) activates SLC16A1 transcription in response to , promoting MCT1 upregulation in tissues such as liver, , and intestine to facilitate fatty acid-derived body transport; this effect is absent in PPAR-α knockout models. (AMPK), activated during exercise-induced energy stress, induces SLC16A1 mRNA and protein expression in fast-twitch , aiding clearance and metabolic adaptation. Epigenetic mechanisms further modulate SLC16A1 expression, particularly through and modifications. Hypomethylation of the SLC16A1 promoter in cancer cells correlates with increased MCT1 mRNA levels, while hypermethylation silences expression; treatment with demethylating agents like 5-aza-2'-deoxycytidine restores transcription. acetylation at the SLC16A1 promoter, mediated by the acetyltransferases p300 and (CBP), promotes relaxation and gene activation, as observed in pancreatic beta-cells where p300 enrichment enhances MCT1 expression under metabolic stress. MicroRNAs also exert post-transcriptional control over SLC16A1. miR-29a binds to the 3' (UTR) of SLC16A1 mRNA, suppressing MCT1 protein expression during processes, including in muscle and pancreatic beta-cells, thereby fine-tuning monocarboxylate in response to developmental signals.

Post-translational modifications

MCT1 is subject to several post-translational modifications that influence its trafficking, stability, and kinetics. represents a primary regulatory mechanism, with the protein containing multiple potential sites for kinases such as p38 (MAPK). Activation of the p38 MAPK pathway has been shown to increase the maximal velocity (V_max) of MCT1-mediated by approximately two-fold without altering the Michaelis constant (K_m), likely by promoting translocation to the surface. Similarly, cAMP-dependent signaling induces of MCT1, leading to its internalization from the plasma membrane into caveolae and early endosomes, thereby reducing surface expression and activity. Glycosylation does not occur on MCT1 itself, as the protein lacks consensus N-linked sites; however, its proper folding, trafficking, and function depend on association with the ancillary protein basigin (CD147/BSG), which undergoes extensive N-linked at three extracellular residues. This of basigin is essential for the formation of the MCT1-basigin heterodimer, enabling efficient delivery of MCT1 to the plasma membrane and optimal monocarboxylate transport. Disruption of basigin impairs this and reduces MCT1 surface expression. Ubiquitination regulates MCT1 turnover by targeting the protein for endosomal-lysosomal . Plasma membrane-localized MCT1 undergoes ubiquitination, which facilitates its sorting into the degradative pathway, thereby controlling steady-state levels and preventing excessive accumulation under varying metabolic demands. This process is particularly relevant under , where increased ubiquitination contributes to MCT1 downregulation, modulating handling and cellular balance in stressed environments such as tumors or inflamed tissues. Overall, these modifications fine-tune MCT1 function without altering transcriptional control, such as the upregulation of MCT1 expression by AMPK activation.

Pathological implications

Genetic disorders

Monocarboxylate transporter 1 (MCT1), encoded by the SLC16A1 gene located on chromosome 1p13.2, is associated with several rare inherited disorders arising from pathogenic variants that disrupt its function or . These conditions primarily manifest as metabolic disturbances due to impaired monocarboxylate , including , pyruvate, and , across cell membranes. The disorders are autosomal recessive or dominant, with clinical presentations ranging from recurrent to exercise-induced and . MCT1 deficiency (MCT1D; OMIM #616095) is an autosomal recessive disorder caused by biallelic loss-of-function mutations in SLC16A1, such as homozygous nonsense variants like c.549G>A (p.Trp183Ter), leading to absent or severely reduced MCT1 protein expression and activity. This impairs ketone body uptake into cells, particularly in tissues reliant on oxidation during , resulting in recurrent severe ketoacidotic crises often triggered by illness, , or stress. Symptomatic presentation typically begins in infancy with episodes of profound , vomiting, dehydration, and hypoglycemia, sometimes accompanied by neurological symptoms like seizures or developmental delay; confirmation involves functional assays demonstrating absent erythrocyte MCT1 activity and via Sanger sequencing or next-generation sequencing to identify the causative variants. The condition is extremely rare, with at least 17 cases reported globally as of mid-2025, predominantly in consanguineous families. Familial hyperinsulinemic hypoglycemia type 7 (HHF7; OMIM #610021) represents an autosomal dominant form linked to heterozygous gain-of-function mutations in the SLC16A1 promoter region, such as a T-to-G 202 bp upstream of the translation start site (-202T>G), which abolishes a and induces ectopic MCT1 expression in pancreatic beta cells. Normally restricted from beta cells, MCT1 presence here facilitates pyruvate influx during , triggering inappropriate insulin release and subsequent post-exercise , which may cause syncope, seizures, or loss of consciousness. Symptoms are exercise-specific, with normal fasting glucose levels, and diagnosis relies on genetic identification of promoter variants alongside provocative testing like anaerobic exercise challenges. Only a limited number of families—fewer than 10—have been documented since its initial description in 2004. Erythrocyte lactate transporter defect (ELTD; OMIM #245340), also known as symptomatic deficiency in , is a rare autosomal dominant condition caused by heterozygous loss-of-function mutations in SLC16A1, exemplified by variants like c.1307_1309del (p.Gly436del), which abolish efflux from erythrocytes and . This accumulation of intracellular causes due to fragility, as well as exercise- and heat-induced muscle cramps, pain, stiffness, fatigue, and elevated levels, without overt . Presentation often occurs in childhood or following physical exertion, with diagnosis confirmed by reduced erythrocyte rates (typically <50% of normal) and Sanger sequencing revealing the mutations. Fewer than 10 cases have been reported, highlighting its rarity and challenges in recognition.

Role in cancer

Monocarboxylate transporter 1 (MCT1), encoded by , is frequently overexpressed in various solid tumors, including , , and , where it supports aberrant metabolic demands under hypoxic conditions. This overexpression is often driven by , which transcriptionally upregulates MCT1 in response to low oxygen levels, promoting a glycolytic shift in tumor cells. A systematic review of 16 cancer types found MCT1 overexpression associated with poor prognosis in most, with a meta-analysis indicating high MCT1 expression correlates with reduced disease-free survival (HR 1.54, 95% CI: 1.28–1.85, p<0.001). High levels generally predict worse outcomes in and . MCT1 plays a critical role in sustaining the Warburg effect, the hallmark aerobic glycolysis of cancer cells, by facilitating the efflux of lactate and protons from glycolytic tumor cells. This export prevents intracellular acidification, allowing continued high-rate glycolysis for rapid proliferation and biomass production, while the released lactate serves as an energy substrate for neighboring oxidative tumor cells or stromal components in a metabolic symbiosis. In this manner, MCT1 enables tumors to maintain an optimal cytosolic pH despite excessive lactate generation, thereby supporting tumor progression and survival under nutrient-limited conditions. Beyond metabolic support, MCT1 contributes to immune evasion by promoting acidification of the tumor microenvironment (TME) through lactate export, which impairs antitumor immune responses. Elevated extracellular lactate and low pH suppress T-cell proliferation, cytokine production, and effector functions, including cytotoxic activity against tumor cells, while fostering immunosuppressive cells like regulatory T cells and M2 macrophages. This TME remodeling creates a barrier to effective immunosurveillance, enhancing tumor escape from adaptive immunity. Recent preclinical studies from 2025 highlight 's therapeutic vulnerability in enhancing immunotherapy. In melanoma models, pharmacological blockade of with inhibitors like BAY-8002 rewired dendritic cell metabolism, restoring pro-inflammatory cytokine secretion (e.g., , ) and boosting CD4+ and CD8+ T-cell activation, proliferation, and effector functions. This metabolic intervention reduced immunosuppressive cytokine levels (e.g., , ) and improved anti-tumor T-cell fitness, synergizing with checkpoint inhibitors to increase tumor regression in glycan-altered TME settings. Such findings underscore inhibition as a strategy to overcome TME-mediated resistance to immunotherapy.

Therapeutic targeting

Inhibitors and activators

Monocarboxylate transporter 1 (MCT1) is modulated by various pharmacological inhibitors, with potent synthetic compounds targeting its proton-coupled mechanism. AZD3965 represents a highly selective MCT1 , exhibiting an IC50 of approximately 1.7 nM for in cells overexpressing MCT1, while showing greater than 100-fold selectivity over MCT4. This compound has been developed for potential anticancer applications due to its ability to block efflux in tumor cells. Similarly, syrosingopine acts as a dual of MCT1 and MCT4, with an IC50 of about 2.4 μM for MCT1-mediated , though it is approximately 60-fold more potent against MCT4 (IC50 40 nM). Natural and early-developed inhibitors also affect MCT1 activity, often with lower potency and less selectivity. , a found in various plants, inhibits MCT1 with an IC50 of around 25 μM, competing with substrates like for binding. α-Cyano-4-hydroxycinnamate (CHC), a classic non-selective inhibitor, exhibits a Ki of approximately 10 μM for MCT1, though its efficacy varies across isoforms and it impacts multiple MCT family members. These agents have been instrumental in early studies elucidating MCT1 function. Recent screening efforts have identified repurposed drugs, including non-steroidal anti-inflammatory drugs (NSAIDs) such as , as MCT1 inhibitors with IC50 values in the low micromolar range. Direct activators of MCT1 are scarce, but indirect modulation occurs through upstream signaling pathways. AMPK agonists such as 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) enhance MCT1 expression and activity via phosphorylation-dependent mechanisms, increasing protein levels in muscle tissues following treatment. This upregulation supports improved handling during metabolic stress. Regarding binding mechanisms, most MCT1 inhibitors, including AZD3965 and , competitively occupy the central substrate-binding pocket within the transporter's , directly competing with monocarboxylates like . In contrast, syrosingopine inhibits the coupled transport of and H+ ions, though its precise binding mechanism on MCT1 remains to be fully elucidated. These distinct modes underscore the potential for isoform-specific targeting in therapeutic design.

Clinical applications

Monocarboxylate transporter 1 (MCT1) has emerged as a target in cancer therapy due to its role in facilitating efflux from tumor cells, supporting the effect. The selective MCT1 AZD3965 underwent a phase I dose-escalation trial (NCT01791595) in patients with advanced solid tumors, enrolling 40 participants who received doses ranging from 5 to 30 mg once daily or 10 to 15 mg twice daily. The trial, completed in 2018 with results reported in 2023, demonstrated that AZD3965 was generally well tolerated, with the most common adverse events being grade 1 or 2 and ; no dose-limiting toxicities occurred at the recommended phase II dose of 15 mg twice daily. Although no objective responses were observed, 12 patients achieved stable disease, indicating modest antitumor activity in this heavily pretreated population. Preclinical studies have explored combinations of MCT1 inhibition with to enhance efficacy. For instance, AZD3965 has shown potential synergy with PD-1 inhibitors by increasing immune cell infiltration, such as dendritic cells and natural killer cells, in tumor models like B-cell lymphoma xenografts, suggesting improved antitumor immune responses when MCT1 is blocked. Ongoing aims to translate these findings into clinical settings for cancers reliant on MCT1-mediated . In metabolic disorders, MCT1 deficiency, caused by mutations in the SLC16A1 gene, manifests as recurrent episodes of severe and , particularly during or illness. Management focuses on supportive measures to prevent and treat these crises, including prompt administration of oral carbohydrates or intravenous 10% dextrose in saline to correct and . Aggressive fluid and correction are essential during acute episodes, as body utilization is impaired due to defective MCT1 transport. While approaches remain exploratory for this rare condition, current strategies emphasize early recognition and preventive dietary interventions to avoid triggers like prolonged . MCT1 overexpression in tumors has been leveraged for , particularly with prodrugs that exploit this transporter for selective uptake. For example, 3-bromopyruvate (3-BP), an alkylating agent that inhibits , is transported into via MCT1, enabling its use in tumor-specific metabolic disruption; formulations like microencapsulated 3-BP (ME3BP-7) further enhance targeting in MCT1-high pancreatic ductal models, leading to rapid while sparing normal tissues. Similar strategies with MCT1-dependent prodrugs, such as dicarboxylate conjugates of 5-fluorouracil, have demonstrated improved oral and colon selectivity in preclinical evaluations. As a , MCT1 expression levels in tissues correlate with aggressive disease features and outcomes. High MCT1 expression is associated with larger tumor size, higher histological grade, increased of recurrence, and shorter across subtypes, including . This prognostic value positions MCT1 as a potential tool for , guiding decisions on therapies in clinical practice.

References

  1. [1]
    SLC16A1 solute carrier family 16 member 1 [ (human)] - NCBI
    Aug 19, 2025 · The N-terminal signature motif on the transporter MCT1 is critical for CD147-mediated trafficking. The polymorphism T1470A of the SLC16A1 gene ...
  2. [2]
    The monocarboxylate transporter family--Structure and functional ...
    Nov 30, 2011 · Monocarboxylate transporters (MCTs) catalyze the proton-linked transport of monocarboxylates such as L-lactate, pyruvate, and the ketone bodies ...
  3. [3]
    Monocarboxylate transporters in the brain and in cancer - PMC
    This review focuses on the brain and on cancer. In the brain, MCTs control the delivery of lactate, produced by astrocytes, to neurons, where it is used as an ...
  4. [4]
    SLC16A1 Gene - GeneCards | MOT1 Protein | MOT1 Antibody
    SLC16A1 (Solute Carrier Family 16 Member 1) is a Protein Coding gene. Diseases associated with SLC16A1 include Erythrocyte Lactate Transporter Defect and ...
  5. [5]
    Entry - *600682 - SOLUTE CARRIER FAMILY 16 ... - OMIM
    The SLC16A1 gene encodes a monocarboxylate transporter (MCT1) that mediates the movement of lactate and pyruvate across cell membranes.
  6. [6]
    Gene: SLC16A1 (ENSG00000155380) - Summary - Homo_sapiens
    Gene: SLC16A1 ENSG00000155380 . Description. solute carrier family ... Gene Synonyms. MCT, MCT1. Location. Chromosome 1: 112,911,847-112,957,593 reverse strand.
  7. [7]
    Physical Exercise–Induced Hypoglycemia Caused by Failed ...
    Jul 26, 2007 · These studies show that promoter-activating mutations in EIHI induce SLC16A1 expression in b cells, where this gene is not usually transcribed, ...<|separator|>
  8. [8]
    The making of a potent L-lactate transport inhibitor - PubMed Central
    Sep 6, 2021 · We report the rational, structure-guided identification of potent, nanomolar affinity inhibitors acting on an L-lactate-specific SLC16 homologue from the ...
  9. [9]
    Monocarboxylate transporter 1 - Homo sapiens (Human) | UniProtKB
    Nov 30, 2010 · MCT1 is a major lactate exporter (By similarity). Plays a role in cellular responses to a high-fat diet by modulating the cellular levels of ...
  10. [10]
    SLC16A1 (solute carrier family 16, member 1 (monocarboxylic acid ...
    Feb 1, 2010 · HGNC. SLC16A1 ; LOCATION. 1p13.2 ; LOCUSID. 6566 ; ALIAS. HHF7,MCT,MCT1,MCT1D ; FUSION GENES. Show Gene Fusions ...<|separator|>
  11. [11]
  12. [12]
  13. [13]
    Embigin facilitates monocarboxylate transporter 1 localization to the ...
    Sep 13, 2022 · Cell-surface ancillary proteins basigin (CD147) or embigin (gp70) act as chaperones facilitating membrane trafficking of MCTs (Poole and ...Missing: ER | Show results with:ER
  14. [14]
    Cooperative transport mechanism of human monocarboxylate ...
    May 15, 2020 · To understand the cooperative transport mechanism, we determined the cryo-EM structure of MCT2 at a resolution of 3.8 Å. The density map ...
  15. [15]
    CD147 is tightly associated with lactate transporters MCT1 and ...
    We conclude that CD147 facilitates proper expression of MCT1 and MCT4 at the cell surface, where they remain tightly bound to each other.Missing: ER | Show results with:ER
  16. [16]
  17. [17]
    Lactate transport in skeletal muscle — role and regulation of ... - NIH
    The mechanism of transport involves an ordered sequential mechanism whereby a proton first binds to the transporter and then the lactate anion. This is ...Missing: Km | Show results with:Km
  18. [18]
    AR-C155858 is a potent inhibitor of monocarboxylate transporters ...
    Using the rate of L-lactate transport and the concentration of MCT1 we calculated the turnover number (kcat) to be 12.2±1.1 s−1 at 6 °C. Since we have ...
  19. [19]
  20. [20]
    [PDF] A Kinetic Model of the Monocarboxylate Transporter MCT1 and its ...
    The model consists of six states arranged in cyclic fashion and features an ordered, mirror- symmetric, binding mechanism, where binding and unbinding of the ...<|control11|><|separator|>
  21. [21]
    The kinetics, substrate, and inhibitor specificity of the ... - PubMed
    Km values for L-lactate (4.7 mM), D-lactate (27 mM), D,L-2-hydroxybutyrate (3.3 mM), L-3-hydroxybutyrate (12.7 mM), and acetoacetate (6.1 mM) were very similar ...
  22. [22]
    The kinetics, substrate and inhibitor specificity of the ... - PubMed
    Dec 15, 1994 · The Km values for L-lactate, pyruvate and D-lactate were found to be 4.54, 0.72 and 27.5 mM respectively, similar to values found previously ...
  23. [23]
    Substrate and inhibitor specificities of the monocarboxylate ...
    Of physiological interest, the ketone bodies D-beta-hydroxybutyrate and acetoacetate had K(m) values of 10 and 5.4 mM, respectively. Vmax values were similar to ...
  24. [24]
    Monocarboxylate transporters in the brain and in cancer
    MCT1 preferentially facilitates the uptake of lactic acid and operates in an ordered process that starts when a proton binds to K38 at the extracellular surface ...Missing: His- | Show results with:His-
  25. [25]
    Tissue expression of SLC16A1 - Summary - The Human Protein Atlas
    No readable text found in the HTML.<|control11|><|separator|>
  26. [26]
    Immunohistochemical analysis of MCT1, MCT2 and MCT4 ... - NIH
    MCT1 is widely expressed in different tissues (Halestrap & Price, 1999) ... Our results were that MCT1 protein is present throughout oxidative fibres ...
  27. [27]
    Monocarboxylate Transporter 1 - an overview | ScienceDirect Topics
    Monocarboxylate transporter 1 (MCT1) transports ketone bodies (from fatty acid metabolism in the liver) and lactate (released in skeletal muscles during ...
  28. [28]
    Monocarboxylate Transporter 1 (MCT1) in Liver Pathology - MDPI
    Membrane monocarboxylate transporter 1 (SLC16A1/MCT1) plays an important role in hepatocyte homeostasis, as well as drug handling.
  29. [29]
    Monocarboxylate Transporter 1 May Benefit Cerebral Ischemia via ...
    Apr 25, 2022 · This review concludes that MCT1, in cerebral ischemia, may improve lactate transport from glial cells to neurons, which subsequently alleviates cellular damage.
  30. [30]
    Cell-specific expression pattern of monocarboxylate transporters in ...
    It was observed that, at both the mRNA and the protein levels, astrocytes strongly expressed MCT1 but had very little if any MCT2. By contrast, neurons had high ...
  31. [31]
    Article New Insights into the Lactate Shuttle: Role of MCT4 in the ...
    Dec 20, 2019 · When Bsg is knocked out, MCT1 and MCT4 fail to traffic to the plasma membrane and are targeted for degradation. Specific deletion of Bsg in ...
  32. [32]
    Developmental switch in brain nutrient transporter expression in the rat
    This study investigated the temporal and regional expression of MCT1 and MCT2 mRNA and protein in the BBB and the nonvascular brain during postnatal development ...
  33. [33]
    Expression of monocarboxylate transporter mRNAs in mouse brain
    During development, a peak in the expression of both transporters occurs around postnatal day 15, declining rapidly by 30 days at levels observed in adults.
  34. [34]
    Perinatal and early postnatal changes in the expression of ...
    We studied the immunocytochemical expression of the monocarboxylate transporters MCT1 and MCT2 in the rat brain between embryonic day (E) 16 and postnatal day ( ...
  35. [35]
    Role of Monocarboxylate Transporters in Drug Delivery to the Brain
    One of the important roles of MCT1 is the unidirectional transport of L-lactate (influx or efflux) which depends on the intracellular and extracellular lactate ...
  36. [36]
    Inhibition of MCT 1 & 4 reduces exercise capacity in mice
    Sep 6, 2022 · In the current study, we examined whether selective inhibitors of MCT1 and MCT4 alter the exercise capacity and lactate metabolism.Abstract · INTRODUCTION · RESULTS · DISCUSSIONMissing: upregulation | Show results with:upregulation
  37. [37]
    Lactate metabolism in human health and disease - Nature
    Sep 1, 2022 · Lactate shuttle mediated by MCT1 and other subtypes establishes intracellular connections and is involved in the synergistic metabolism between ...
  38. [38]
    Lactate Transporters Mediate Glia-Neuron Metabolic Crosstalk in ...
    MCTs are a family of proton-linked plasma membrane transporters that allow the passage of monocarboxylates, including lactate, pyruvate, and ketone bodies ( ...Differentially Expressed... · Lactate Transporter... · Brain Energy Metabolism and...<|separator|>
  39. [39]
    Role of Human Monocarboxylate Transporter 1 (hMCT1) and 4 ...
    Sep 4, 2023 · MCT1 and MCT4 play a crucial role in the maintenance of intracellular pH in tumor cells by transporting monocarboxylic acids (such as lactate, pyruvate and ...
  40. [40]
    The Hepatic Monocarboxylate Transporter 1 (MCT1) Contributes to ...
    Apr 14, 2021 · Our observations suggest an important role of ketone bodies and its transporter Mct1 in FAA under caloric and temporal food restriction.
  41. [41]
    A SLC16A1 Mutation in an Infant With Ketoacidosis and ... - Frontiers
    Jul 17, 2019 · We report a variant in the SLC16A1 gene encoding MCT1 in a neonate with recurrent acidosis, hypoglycemia, and neurological sequelae. Individuals ...
  42. [42]
    Cerebral ketone body metabolism - PubMed
    Ketone bodies (KBs) are an important source of energy for the brain. During the neonatal period, they are also precursors for the synthesis of lipids ...
  43. [43]
    Role of MCT1 and CAII in skeletal muscle pH homeostasis ...
    Mar 2, 2017 · The purpose of this study was to investigate the effects of a partial suppression of monocarboxylate transporter (MCT)-1 on skeletal muscle ...
  44. [44]
    Monocarboxylate Transporter 1 (MCT1) in Liver Pathology - PubMed
    Feb 26, 2020 · Membrane monocarboxylate transporter 1 (SLC16A1/MCT1) plays an important role in hepatocyte homeostasis, as well as drug handling.
  45. [45]
  46. [46]
  47. [47]
  48. [48]
  49. [49]
  50. [50]
    Phosphorylation of MCT-1 by p44/42 MAPK is required for its ...
    Oct 2, 2006 · The MCT-1 protein is predicted to have numerous putative phosphorylation sites including, a p44/p42 mitogen-activated protein kinases (MAPK) and ...
  51. [51]
    [PDF] regulation of monocarboxylate transporter 1 (mct1) by
    phosphorylate MCT1, but rather reduces the phosphorylation of MCT1. Because the phosphorylation status of membrane proteins can control their surface.
  52. [52]
    Regulation of Mct1 by cAMP-dependent internalization in rat brain ...
    In this report, we demonstrate that cAMP induces the dephosphorylation and internalization of Mct1 from the plasma membrane into caveolae and early endosomes.
  53. [53]
    Interaction of the Erythrocyte Lactate Transporter (Monocarboxylate ...
    It could be argued that the new MCT-containing band is a result of cross-linking MCT1 to itself, forming oligomers. This seems unlikely for several reasons.
  54. [54]
    Links between CD147 Function, Glycosylation, and Caveolin-1
    Cell surface CD147 shows remarkable variations in size (31-65 kDa) because of heterogeneous N-glycosylation ... MCT1 and MCT4 and facilitates their cell surface ...
  55. [55]
    Regulation of Monocarboxylic Acid Transporter 1 Trafficking by ... - NIH
    MCT1 on the plasma membrane undergoes ubiquitination, and the ubiquitinated MCT1 is sorted into the endosomal/lysosomal trafficking system for degradation.Missing: 248 | Show results with:248
  56. [56]
    Effect of AMPK activation on monocarboxylate transporter (MCT)1 ...
    AMPK is a possible candidate for regulating exercise-induced changes in MCT expression, because it is known to regulate various proteins related energy ...Missing: SLC16A1 | Show results with:SLC16A1
  57. [57]
    Monocarboxylate Transporter 1 Deficiency and Ketone Utilization
    Nov 13, 2014 · Genetic analysis in 96 patients suspected of having ketolytic defects yielded seven additional inactivating mutations in MCT1, both homozygous ...
  58. [58]
    A SLC16A1 Mutation in an Infant With Ketoacidosis and ...
    Jul 18, 2019 · Inactivating recessive homozygous or heterozygous mutations in the SLC16A1 gene were described in patients with recurrent ketoacidosis and ...Missing: 1307G> G436D
  59. [59]
    Mitochondrial bioenergetic is impaired in Monocarboxylate ...
    Jun 21, 2022 · Deficient MCT1 activity due to inactivating nucleotide changes in SLC16A1 has been recorded in patients suffering from a ketone metabolism ...<|control11|><|separator|>
  60. [60]
    610021 - HYPERINSULINEMIC HYPOGLYCEMIA, FAMILIAL, 7; HHF7
    Familial hyperinsulinemic hypoglycemia-7 (HHF7) is characterized by inappropriate insulin secretion, particularly during anaerobic exercise.
  61. [61]
    Mutations in MCT1 cDNA in patients with symptomatic ... - PubMed
    We identified 5 patients with subnormal erythrocyte lactate transport plus symptoms and signs of muscle injury on exercise and heat exposure.
  62. [62]
    245340 - ERYTHROCYTE LACTATE TRANSPORTER DEFECT
    - Caused by mutations in the solute carrier family 16, monocarboxylic acid transporter, member 1 gene (SLC16A1, 600682.0001). △ Close ...
  63. [63]
    Targeting lactate metabolism for cancer therapeutics - JCI
    For example, elevated levels of MCT1 have been detected in breast, colorectal, gastric, and cervical cancer as well as in neuroblastoma and glioma (63–68) ...
  64. [64]
    Monocarboxylate transporter 1 and 4 inhibitors as potential ...
    Aug 1, 2020 · Overexpression of MCT1 is found in variety of cancers including head and neck cancer, breast cancer, colon cancer, lung cancer ...<|separator|>
  65. [65]
    Prognostic Value of Monocarboxylate Transporter 1 Overexpression ...
    Results showed that MCT1 is an indicator of poor prognosis and decreased survival for cancer patients in sixteen types of malignancies.
  66. [66]
    Proton-coupled monocarboxylate transporters in cancer
    Nov 23, 2022 · Prognostic significance of lactate/proton symporters MCT1, MCT4, and ... K. M., et al. (2012). Targeting the lactate transporter MCT1 ...Mct And Tumor Metabolism · Mct And Energy Metabolism · Mcts And Targeted Drugs<|control11|><|separator|>
  67. [67]
    Role of monocarboxylate transporters in cancer immunology and ...
    Jul 30, 2025 · Monocarboxylate transporters (MCTs) affect cancer metabolism and the regulation of immune responses, making them targets for cancer therapy.
  68. [68]
    Impact of lactate on immune cell function in the tumor ... - Frontiers
    Lactate enters cells through MCT1 and upregulates MOESIN, stimulating the TGF-β/STAT3 pathway to enhance Treg cell development and immunological suppression.
  69. [69]
  70. [70]
    MCT1 lactate transporter blockade re-invigorates anti-tumor ... - Nature
    Jan 27, 2025 · Indeed, tumors create a suppressive metabolic microenvironment that supports the metabolism of tumor-promoting cells (regulatory CD4+ T [Treg] ...
  71. [71]
    The role of MCT1 in tumor progression and targeted therapy - NIH
    Jun 19, 2025 · Although MCT1 can transport multiple substrates, its primary physiological role is to mediate lactate entry into cells and, in conjunction with ...Mct1 Mediates Metabolic... · Mct1 Leads To... · Inhibition Of Mct1 Impedes...<|control11|><|separator|>
  72. [72]
    A Phase I Dose-escalation Study of AZD3965, an Oral ...
    We report results from the dose-escalation part of a first-in-human trial of AZD3965, a first-in-class MCT1 inhibitor, in advanced cancer. Patients and Methods:.
  73. [73]
    [PDF] LICENSING OPPORTUNITY: MCT-1 INHIBITOR AZD3965
    AZD3965-mediated inhibition of MCT1 in the Raji B cell lymphoma cells (MCT4-. /MCT1+) increased the abundance of dendritic cells (DC) and natural killer (NK).Missing: NCT01791595 | Show results with:NCT01791595<|separator|>
  74. [74]
    MCT1 deficiency - a rare disorder of the SLC16A1 gene
    A rare genetic disorder of the SLC16A1 gene causes MCT1 deficiency, which affects ketone metabolism. MCT1 deficiency was first reported in 2014. By 2016 ...Missing: p. G436D mutation
  75. [75]
    Heterozygous Monocarboxylate Transporter 1 (MCT1, SLC16A1 ...
    Nov 26, 2015 · Arg328Ter mutation and that for other truncating mutations in the SLC16A1 gene, differences in the sensitivity of the immunoblots performed in ...Missing: 1307G> G436D<|control11|><|separator|>
  76. [76]
    Tumor Energy Metabolism and Potential of 3-Bromopyruvate as an ...
    3-BrPA enters tumor cells via tumor-specific overexpression of monocarboxylate transporters (MCTs) (especially MCT1), followed by the inhibition of glycolysis ( ...Missing: prodrug | Show results with:prodrug
  77. [77]
    ME3BP-7 is a targeted cytotoxic agent that rapidly kills pancreatic ...
    May 20, 2025 · This study presents a valuable finding and developed ME3BP-7 as a novel microencapsulated formulation of 3BP, which specifically targets MCT1-overexpressing ...Missing: prodrug | Show results with:prodrug
  78. [78]
    A novel oral prodrug-targeting transporter MCT 1 - NIH
    Apr 25, 2019 · A novel oral prodrug-targeting transporter MCT 1: 5-fluorouracil-dicarboxylate monoester conjugates ... conjugates as prodrugs for colon cancer ...Missing: bromopyruvate | Show results with:bromopyruvate
  79. [79]
    Unveiling Metabolic Signatures as Potential Biomarkers in Common ...
    MCT1 overexpression was positively correlated with large tumour size, high-grade tumours, shorter progression-free survival, and cancer recurrence, irrespective ...
  80. [80]
    SLC16A1 - an overview | ScienceDirect Topics
    SLC16A1, also known as MCT1, is a proton-linked transporter for monocarboxylates like acetate, acetoacetate, and beta-hydroxybutyrate, located at 1p12.Missing: evolutionary | Show results with:evolutionary