Novel therapies with precision mechanisms for type 2 diabetes mellitus - Nature

Page created by Walter Curry
 
CONTINUE READING
Reviews

                                 Novel therapies with precision
                                 mechanisms for type 2 diabetes
                                 mellitus
                                 Leigh Perreault         1   ✉, Jay S. Skyler2 and Julio Rosenstock3

                                 Abstract | Type 2 diabetes mellitus (T2DM) is one of the greatest health crises of our time and
                                 its prevalence is projected to increase by >50% globally by 2045. Currently, 10 classes of drugs
                                 are approved by the US Food and Drug Administration for the treatment of T2DM. Drugs in
                                 development for T2DM must show meaningful reductions in glycaemic parameters as well as
                                 cardiovascular safety. Results from an increasing number of cardiovascular outcome trials using
                                 modern T2DM therapeutics have shown a reduced risk of atherosclerotic cardiovascular disease,
                                 congestive heart failure and chronic kidney disease. Hence, guidelines have become increasingly
                                 evidence based and more patient centred, focusing on reaching individualized glycaemic goals
                                 while optimizing safety, non-​glycaemic benefits and the prevention of complications. The bar
                                 has been raised for novel therapies under development for T2DM as they are now expected to
                                 achieve these aims and possibly even treat concurrent comorbidities. Indeed, the pharmaceutical
                                 pipeline for T2DM is fertile. Drugs that augment insulin sensitivity, stimulate insulin secretion or
                                 the incretin axis, or suppress hepatic glucose production are active in more than 7,000 global
                                 trials using new mechanisms of action. Our collective goal of being able to truly personalize
                                 medicine for T2DM has never been closer at hand.

                                Formal diagnostic criteria for diabetes mellitus were         speculation that some agents, such as thiazolidinedi-
                                first introduced by the National Diabetes Data Group          ones, might actually increase cardiovascular risk10. On
                                in 1979 (ref.1) and by the World Health Organization in       account of these concerns, in 2008, the FDA mandated
                                1980 (ref.2). Expanded and refined over time, plasma glu-     cardiovascular safety studies be conducted for all new
                                cose thresholds for diagnosis remain in diagnostic criteria   medications approved for lowering plasma levels of glu-
                                today based on their predictive value for microvascular       cose in patients with T2DM. Knowledge gained from the
                                disease, specifically retinopathy1. Furthermore, land-        cardiovascular outcome trials of these newer agents has
                                mark trials have convincingly demonstrated a reduction        considerably expanded our understanding of T2DM and
                                in incidence of microvascular disease with decreasing         what can be done for patients. Specifically, glucagon-​like
                                levels of plasma glucose in both type 1 diabetes melli-       peptide 1 (GLP1) receptor agonists and sodium–
                                tus and type 2 diabetes mellitus (T2DM)3–7. Altogether,       glucose cotransporter 2 (SGLT2) inhibitors exert desir-
                                diabetes mellitus is largely conceived as a disease of ele-   able ‘off-​target’ non-​glycaemic effects (for example,
                                vated blood concentrations of glucose. The reduction          reductions in body weight and blood pressure), have
1
 University of Colorado
                                in HbA1c remains a central focus of care8 as well as the      improved safety profiles (for example, no incidences of
Anschutz Medical Campus,        benchmark used by the FDA to approve pharmaceuticals          hypoglycaemia, as compared with insulin and sulfony-
Aurora, CO, USA.                that lower plasma concentrations of glucose.                  lureas), reduce the risk of atherosclerotic cardiovascular
2
 Diabetes Research Institute,       Nevertheless, much ado has been made about how            disease and hospitalization for heart failure, and slow
University of Miami, Miami,     to safely decrease plasma concentrations of glucose in        the progression of diabetic kidney disease11–17. These
FL, USA.
                                people with T2DM, with health-​care providers citing          beneficial effects are all in addition to their ability to
3
 Dallas Diabetes Research       concerns over the potential for hypoglycaemia and             decrease plasma concentrations of glucose. Interestingly,
Center at Medical City,
Dallas, TX, USA.
                                weight gain and, most notably, cardiovascular safety8,9.      SGLT2 inhibitors can also reduce the number of hospi-
✉e-​mail: leigh.perreault@      The latter point arose from interventional randomized         talizations for heart failure in people without T2DM18,19.
cuanschutz.edu                  controlled trials in patients with T2DM, which repeat-        This finding suggests that the cardiovascular benefits are
https://doi.org/10.1038/        edly showed a failure to reduce cardiovascular risk by        provided by a mechanism that is independent from the
s41574-021-00489-​y             decreasing plasma levels of glucose4–7, together with         glucose-​lowering effects.

364 | June 2021 | volume 17                                                                                                     www.nature.com/nrendo

                                                                  0123456789();:
Reviews

                                                                                               that indirectly affect glycaemia (for example, anti-​obesity
 Key points
                                                                                               therapies) or used for complications related to T2DM.
 • Type 2 diabetes mellitus (T2DM) is one of the greatest health crises of our time, and          Third, we excluded preclinical, non-​human phase
   the number of people with T2DM is projected to increase by >50% globally by 2045.           evaluations or therapies that failed to meet their
 • Despite our extensive armamentarium of current drug treatments for T2DM, >7,000             safety and/or efficacy end points (that is, terminated
   trials are registered around the world, many looking at ‘novel’ drug targets.               development programmes). Outcomes provided to
 • Mechanisms of action for novel drugs in the pipeline for T2DM include directly              ClinicalTrials.gov and publications available on PubMed
   targeting β-​cells, targeting the incretin axis, directly or indirectly affecting glucose   were used to corroborate the phase and status of trials,
   metabolism in the liver, and increasing insulin sensitivity.                                whenever possible.
 • In our judgement, compounds with the most promise include dual-​acting and                     Thus, this Review focuses on the identification of
   triple-​acting incretin mimetics owing to their glucose-​lowering capacity,                 potentially novel pharmaceuticals for T2DM that are
   non-​glycaemic benefits and safety.
                                                                                               in active or completed clinical trials and conducted in
 • The bar has been raised for novel therapies under development for T2DM; new                 people with T2DM, with decreasing HbA1c levels as the
   therapies are now expected to prevent cardiovascular and renal complications                primary outcome, specifically for the purpose of meeting
   independent of and in addition to their ability to decrease the plasma concentrations
                                                                                               regulatory approval.
   of glucose.

                                                                                               Identified novel mechanisms of action
                                  In pursuit of precision medicine, guidelines for peo-        Once the aforementioned criteria were applied, we iden-
                               ple with T2DM are now principally focused on reaching           tified 43 compounds with highly novel mechanisms of
                               individualized glycaemic targets, while optimizing safety,      action in development for T2DM. Collectively, their
                               non-​glycaemic benefits, and the prevention of micro-           glucose-​lowering mechanisms could be grouped into
                               vascular and macrovascular complications for individual         four over-​arching physiological effectors of diabetes
                               patients who are at risk of specific complications20. Novel     mellitus. First are drugs that stimulate insulin secre-
                               therapies in development will not only need to show             tion directly from β-​cells (Fig. 1). These include both
                               meaningful reductions in glycaemic parameters but               pancreas-​selective and pancreas–liver dual-​activating
                               will need added value to meet these increased standards.        glucokinase activators (GKAs) and G-​protein-​coupled
                               Furthermore, to be competitive against the current ther-        receptor 40 (GPCR40) agonists (Table 1). Second are
                               apies in use, new drugs must distinguish themselves with        drugs that utilize the incretin axis (Figs 1,2). These include
                               additional attributes such as contributing to increased         agonists of the GLP1 receptor and glucose-​dependent
                               weight loss, having no increased risk for hypoglycaemia,        insulinotropic polypeptide (GIP) receptor, GLP1–gluca-
                               or utilizing improved drug delivery systems and routes          gon receptor agonists, triple GLP1–GIP–glucagon
                               of administration that might decrease the frequency of          receptor agonists, oxyntomodulin, and agonists of
                               use. This Review generates a comprehensive list of the          G-​protein-​coupled receptor 119 (GPCR119) (Table 2).
                               novel therapies for T2DM currently in development and           Third are drugs that directly or indirectly decrease
                               discusses their potential for improving care for patients.      hepatic glucose production or increase hepatic glucose
                                                                                               uptake (Fig. 2). These drugs include glucagon receptor
                               Review criteria                                                 antagonists, antisense oligonucleotide inhibitors specific
                               To compile the most comprehensive list of promising,            for glucagon receptor mRNA, dual amylin–calcitonin
                               novel therapies for T2DM, the US National Institute             receptor agonists (DACRAs) and liver-​selective GKAs
                               of Health Clinical Trials database was searched from            (Table 3). Fourth and finally are drugs that improve
                               November 26, 2019, to March 31, 2020, using the condi-          insulin sensitivity (Fig. 3). These include an antisense
                               tion or disease term ‘type 2 diabetes’. This query yielded      oligonucleotide inhibitor for protein tyrosine phos-
                               7,484 registered trials worldwide, which were examined          phatase 1B (PTP1B) mRNA, fibroblast growth factor 21
                               individually. The following three exclusion criteria were       (FGF21) analogues, a diacylglycerol acetyl transferase 1
                               applied. First, we excluded drugs in existing classes.          (DGAT1) inhibitor and an enterocytic microsomal tri-
                               Therapies considered ‘novel’ were any medical phar-             glyceride transfer protein (MTP) inhibitor. Additional
                               maceutical therapy not currently approved for T2DM.             drugs in this category include novel selective peroxi-
                               Hence, we excluded new drugs in development within              some proliferator-​activated receptor (PPAR) agonists,
                               existing classes (for example, new dipep­tidyl peptidase        a GLUT4 facilitative transporter stimulator, a nuclear
                               4 inhibitors) as well as advances in devices or technol-        factor-​κB (NF-​κB) inhibitor, a selective allosteric acti-
                               ogies related to currently approved medications (for            vator of LYN kinase, a nicotinic α7 receptor ligand,
                               example, oral insulin and inhaled GLP1 receptor ago-            a ghrelin analogue, a ghrelin–growth hormone (GH)
                               nists). Importantly, advances in delivery devices and           receptor agonist and an activin II-​B receptor modulator
                               technologies for people with T2DM are expected to lead          as well as a drug with an unknown mechanism of action
                               to improved medication adherence and persistence for            (imeglimin) (Table 4).
                               both existing and emerging therapies. An example of
                               such devices is ITCA650, a mini-​osmotic pump capable           Novel agents in development
                               of continuous subcutaneous delivery of exenatide for up         The year 2021 marks the 100th anniversary of the
                               to 6 months21.                                                  discovery of insulin — the very first pharmacological
                                   Second, we excluded therapies tested for their other        treatment for diabetes mellitus. Over those 100 years,
                               physiological properties (for example, insulin sensitiza-       10 distinct classes of drugs have been approved by the
                               tion but not glucose lowering), tested in disease states        FDA for the treatment of T2DM, most of which were

naTure RevIews | EnDoCRinology                                                                                                volume 17 | June 2021 | 365

                                                                 0123456789();:
Reviews

                                Oxyntomodulin
                                                 Glucagon
                          GLP1                   receptor
                        receptor                 agonists
                        agonists                                                                           Meglitinides
                                                                             Glucose
                        GLP1                                 Glucagon                                                Sulphonylureas
                                                             receptor       GLUT1/2
                        receptor
                                                                                                                                        KATP channel
  GIPR           GIPR                                                                                                                   (SUR1-Kir6.2)4
                                                             Glucokinase             Glucokinase
 agonists                                                     activators

                                                                                     Glucose
                                   cAMP            PKA                                                                     ATP
                                                                                     metabolism                                                   K+
            GPCR119                                              Pro-insulin
                                                                 biosynthesis                     Mitochondrion
GPCR119
agonists                                      Piccolo                                                                            Membrane
                                           RIM2 RAB3A                                                                            depolarization

                                   EPAC2                                          Insulin
                                                                                                                                              Ca2+
                                                       Insulin
                                                       granule                                       IP3          PLC

                                                                                                                                            L-type Ca2+
                                   RAP1A                                                  Ca2+- dependent                                   channel
                                                                                          and calmodulin-
                                                                                          dependent proteins
                                                                                                                                                          GPCR40
                                                                                            PKC                  DAG                                      agonists
                                                    Translocation

                                                                                                                          GPCR40
                                                                            Priming

                                                                                Docking

                              Fig. 1 | Drugs that stimulate insulin secretion. Drug classes with new drugs in development are shown (blue ovals) as
                              well as the pathways that they act on in β-​cells to stimulate insulin secretion. New drugs include glucokinase activators
                              (GKAs), which augment glucose-​stimulated insulin secretion. Glucokinase (green oval) converts glucose into glucose-6-​
                              phophate. By contrast, G-​protein-​coupled receptor 40 (GPCR40) agonists act to increase free fatty acid-​stimulated insulin
                              secretion. New drugs that target the incretin axis are also under development, for example, dual and triple incretin mimetics,
                              such as glucagon-​like peptide 1 (GLP1) receptor agonists with a glucose-​dependent insulinotropic polypeptide receptor
                              (GIPR) agonist and/or a glucagon receptor agonist, oxyntomodulin analogues, and G-​protein-​coupled receptor 119
                              (GPCR119) agonists. Drugs that act on the incretin axis stimulate glucose-​dependent insulin secretion through cAMP
                              signalling. Also shown are the established type 2 diabetes mellitus therapeutics sulfonylurea and meglitinide (orange
                              ovals), which bind to SUR1 (part of the ATP-​sensitive Kir6.2 potassium channel), increase calcium influx and increase
                              insulin secretion. Adapted from ref.106, Springer Nature Limited.

                              approved in the past 20 years22. Despite the explosion               deficiency occurs when the pancreas makes insuffi-
                              in therapeutic options for people with T2DM, the pipe-               cient insulin, often in the context of insulin resistance.
                              line for novel therapies in T2DM remains robust and                  Interestingly, of the >400 genetic associations with
                              quite prolific. Companies developing each new therapy                T2DM, the majority govern β-​cell function23. Genetic
                              strive for theirs to be novel, first in class and better than,       discovery has delivered novel mechanistic insights into
                              or complimentary to, existing treatments. The goals of               disease pathophysiology, including new ways to stim-
                              novel therapies are to help patients reach their indi-               ulate insulin secretion directly from β-​cells (Table 1;
                              vidualized glycaemic targets while optimizing safety,                Fig. 1).
                              non-​glycaemic benefits and the prevention of compli-
                              cations for individual patients who are at risk of specific          Glucokinase activators. GKAs are a novel therapy that
                              complications.                                                       directly targets β-​cells, with at least 11 drugs under
                                                                                                   development (one in phase III, four in phase II and six
                              Drugs that directly target β-​cells                                  in phase I clinical trials) (Table 1). Glucokinase, which
                              Insulin deficiency is a defining feature of T2DM.                    facilitates the phosphorylation of glucose to glucose-6-​
                              Absolute insulin deficiency occurs when the pancreas                 phosphate, functions as the ‘glucose sensor’ of the body,
                              no longer makes insulin, whereas a relative insulin                  maintaining plasma concentrations of glucose within a

366 | June 2021 | volume 17                                                                                                               www.nature.com/nrendo

                                                                    0123456789();:
Reviews

                      narrow range (4–6 nM)24. The action of glucokinase is              concentration of triglycerides27–30. Nevertheless, safety
                      restricted to glucose-​sensitive and glucose-​responsive           issues could be avoidable through careful patient
                      tissues such as the liver and pancreas. As such, the GKAs          selection31 and perhaps liver-​s elective GKAs might
                      in development have been designed to target both liver             avoid the increased risk of hypoglycaemia; however, liver
                      and pancreas (dual) or to be more specific, targeting              safety will need to be clearly demonstrated32.
                      only one tissue (selective)25. Glucokinase activation in
                      the liver stimulates hepatic glucose uptake and inhib-             G-​protein-​coupled receptor 40. GPCR40 (also known
                      its hepatic glycogenolysis (Fig. 2), whereas glucokinase           as free fatty acid receptor 1; FFAR1) agonists have gen-
                      activation in the pancreatic β-​cell stimulates insulin            erated considerable interest as a novel mechanism for
                      secretion26. Dual GKAs can be further subdivided by                direct β-​cell stimulation. Agonists of GPCR40 act in the
                      their enzyme kinetics, which can render them more or               β-​cell to induce free fatty acid (FFA)-​stimulated insulin
                      less potent to stimulate insulin secretion, thereby under-         secretion33 and two agents have advanced into phase II
                      scoring the customizability of the molecules. Historically,        trials. These compounds act as cooperative, allosteric
                      GKAs have not thrived in development, limited by their             modulators of GPCR40 that rely on the ample circulat-
                      lack of durability, risk of inducing hypoglycaemia, liver          ing levels of FFAs to potentiate glucose-​dependent insu-
                      toxicity and ability to cause an increase in the plasma            lin secretion34. A previous GPCR40 agonist compound

                       Table 1 | Drugs that directly stimulate β-​cells
                       Compound           Mechanism of action                    Trial     Results                                     Ref. or clinical
                                                                                 phase                                                 trial number
                       GKAs
                       HMS5552            Dual GKA (pancreas and liver);         III       24 people with T2DM randomized              31,103

                       (dorzagliatin)     GKA in pancreatic β-​cells                       to dorzagliatin 75 mg QD or BID for
                                          augments glucose-​stimulated                     28 days; HbA1c –1.22% with QD and
                                          insulin secretion; GKA in liver                  –0.79% with BID
                                          increases hepatic glucose
                       AZD1656            uptake; together, they maintain        II        458 people with T2DM randomized             30,104

                                          plasma levels of glucose at                      to AZD1656 20 mg QD, 40 mg QD,
                                          4–6 nM                                           titrated from 10 mg to 140 mg
                                                                                           QD, titrated from 20 mg to 200 mg QD,
                                                                                           glipizide titrated from 5 mg to 20 mg
                                                                                           QD or placebo for 4 months; HbA1c
                                                                                           –0.80% in the AZD1656 titrated
                                                                                           groups but waned over time
                       RO4389620                                                 Ib        15 people with T2DM randomized              27,105

                       (piragliatin)                                                       to triple crossover of piragliatin
                                                                                           25 mg, 100 mg or placebo; glucose
                                                                                           AUC during an OGTT at the 100 mg
                                                                                           dose was less than that observed at
                                                                                           the 25 mg dose, which was less than
                                                                                           observed with placebo
                       PSN-821                                                   I         No published data                           NCT01386099
                       DS-7309                                                   I         No published data                           NCT01862939,
                                                                                                                                       NCT01956305
                       PB-201                                                    I         No published data                           NCT03973515
                       BMS-820132                                                I         No published data                           NCT01290575,
                                                                                                                                       NCT01105429
                       LY2608204                                                 I         Safety established in phase I trial         NCT01247363
                       Globalagliatin                                            I         No published data                           NCT03414892,
                                                                                                                                       NCT03171623
                       PF04937319         Systemic partial GKA (weak             II        639 people with T2DM randomized             32

                                          pancreatic effects only)                         to PF04937319 50 mg QD or 100 mg
                                                                                           QD for 12 weeks; HbA1c –0.45%
                                                                                           at 100 mg QD; PK/PD, safety and
                                                                                           add-​on studies appear promising
                       GPCR40 agonists
                       MK 8666            GPR40 is highly expressed in           II        63 people with T2DM randomized              33

                                          pancreatic β-​cells; its activation              to MK 8666 50 mg QD, 150 mg
                                          by fatty acids amplifies                         QD, 500 mg QD for 14 days; dose
                                          glucose-​dependent insulin                       dependent 31–54 mg/dl decrease
                                          secretion                                        in fasting plasma glucose
                       JTT-851                                                   II        No published data                           NCT01699737
                       AUC, area under the curve; BID, twice daily; GKA, glucokinase activator; GPCR40, G-​protein-​coupled receptor 40; OGTT, oral
                       glucose tolerance test; PD, pharmacodynamics; PK, pharmacokinetics; QD, daily; T2DM, type 2 diabetes mellitus.

naTure RevIews | EnDoCRinology                                                                                            volume 17 | June 2021 | 367

                                                        0123456789();:
Reviews

                                           Liver                                                                               Islet

                                                                                                                      Oxyntomodulin             Glucagon
                                                                                                                                                receptor
                                                                                                        GPCR119                                 agonists

                                    Glucagon                                                  GIPR                                               Glucagon
                                    receptor              Glucagon                           agonists                                            receptor
                                   antagonists
                                                                                                                                                         DACRAs
                                                                     Glucose                                                   GLP1
    Glucagon receptor antisense                                                                                                receptor
     oligonucleotide inhibitors                                                Glucokinase       GIPR
                                                  Glucagon                      activators
                                                  receptor
                                                                                                           Augmented
                                                                                                           insulin
                                                                                                           secretion                          Amylin
                                                      GLUT1/2                                                                                 receptor
                                                                                                                                       Increased
             Glucagon                 Adenylate                                                                                        amylin
          receptor mRNA                cyclase                                                                                         secretion
                                                     Glucokinase
                                                                                                                                                          β-Cell
                                       cAMP
                                                         G-6-P                                    Amylin

                                                                                                                     Insulin
                                        PKA
                                                      Glycogen

                                  • Glycogenolysis
                                  • Gluconeogenesis              Hepatocyte

                                                                                                                  Glucagon
                                   Hepatic glucose                                                                secretion
                                   production

                                                                                                                                                         α-Cell

                              Fig. 2 | Drugs that decrease hepatic glucose production or increase hepatic glucose uptake. Glucagon receptor
                              antagonists and antisense oligonucleotide inhibitors for the glucagon receptor both act directly on hepatocytes to
                              decrease the hepatic glucose production stimulated by glucagon. By contrast, liver-​selective glucokinase activators
                              increase hepatic glucose uptake. Indirect effects on hepatic glucose metabolism are induced by drugs that act on β-​cells.
                              For example, dual amylin–calcitonin receptor agonists (DACRAs) and drugs targeting the incretin axis (dual or triple
                              incretin mimetics, oxyntomodulin analogues or G-​protein-​coupled receptor 199 (GPCR119) receptor agonists) induce
                              insulin and/or amylin secretion, which inhibits glucagon secretion from α-​cells. Decreased circulating levels of glucagon
                              result in less induction of hepatic glucose production. G-6-​P, glucose 6-​phosphate; GIPR, glucose-​dependent insulinotropic
                              polypeptide receptor; GLP1, glucagon-​like peptide 1.

                              in development (fasiglifam, also called TAK 875) showed           been associated with increased de novo lipogenesis
                              glucose-​lowering properties in phase IIa clinical trials;        and subsequent hepatic steatosis36. Furthermore, nat-
                              however, considerable liver toxicity in phase III trials          urally occurring polymorphisms in the glucokinase
                              led to the termination of its development programme35.            regulatory protein have been associated with elevated
                              Hence, continued vigilance towards safety will dominate           plasma levels of triglyceride, FFAs and VLDL cho-
                              the development of its successors as it is unclear how            lesterol in humans37–39. By contrast, studies in animal
                              these new molecules differ from fasiglifam.                       models show that the liver toxicity seen with GPCR40
                                                                                                agonists seems to be mediated through disrupted bile
                              Potential for liver toxicity. The liver toxicity associated       acid homeostasis40,41. Taking the liver toxicity data
                              with drugs that stimulate insulin secretion directly from         with GKAs and GPCR40 agonists together, it is clear
                              β-​cells is worth contemplating. Of note, both glucoki-           that directly stimulating insulin secretion from the
                              nase and GPCR40 are expressed in the liver as well as             β-​cell using drugs that also have actions in the liver
                              in β-​cells. In humans, naturally occurring mutations             could lead to untoward alterations in pathways of lipid
                              leading to hepatic glucokinase overexpression have                metabolism.

368 | June 2021 | volume 17                                                                                                               www.nature.com/nrendo

                                                                   0123456789();:
Reviews

                       Table 2 | Drugs that utilize the incretin axis
                       Compound            Mechanism of action             Trial Results                                                    Ref. or clinical
                                                                           phase                                                            trial number
                       Multi-​site receptor agonists for GLP1, GIP and/or glucagona
                       AVE2268             GLP1–GIP–glucagon               I         No published data                                      NCT00361738
                                           triple receptor agonist
                       HM12460A            GLP1–GIP–glucagon               I         No published data                                      NCT01724814,
                                           triple receptor agonist                                                                          NCT03332836
                       LY3298176           GLP1–GIP dual receptor          III       318 people with T2DM randomized to                     44

                       (tirzepatide)       agonist                                   tirzepatide 1 mg Q week, 5 mg Q week, 10 mg
                                                                                     Q week, 15 mg Q week, dulaglutide 1.5 mg Q
                                                                                     week or placebo for 26 weeks; HbA1c –1.06%,
                                                                                     –1.73%, –1.89%, –1.94%, –1.21%, –0.06%,
                                                                                     respectively
                       SAR425899           GLP1–glucagon dual              II        PK/PD established in 36 people with T2DM               46

                                           receptor agonist
                       MEDI0382            GLP1–glucagon dual              IIa       MEDI0382 lowered glucose AUC during a                  NCT03596177,
                                           receptor agonist                          MTT versus placebo; PK/PD, safety, add-​on             NCT03555994,
                                                                                     established in phase I trial                           NCT03550378,
                                                                                                                                            NCT03745937,
                                                                                                                                            NCT03385369,
                                                                                                                                            NCT02548585,
                                                                                                                                            NCT03341013,
                                                                                                                                            NCT03444584,
                                                                                                                                            NCT03645421,
                                                                                                                                            NCT03244800,
                                                                                                                                            NCT03235050,
                                                                                                                                            NCT04515849,
                                                                                                                                            NCT04208620
                       Oxyntomodulin analogueb
                       OPK88003            This oxyntomodulin              II        No published data                                      NCT03406377
                                           analogue has the
                                           potential actions of a
                                           GLP1–glucagon dual
                                           receptor agonist
                       GPCR119 agonist
                       MBX-2982            GPCR119 is highly               I         No published data                                      NCT01035879
                                           expressed in the human
                       DS-8500a            gastrointestinal tract          II        99 people with T2DM randomized to                      47,48

                                           (small intestinal L cells)                DS-8500a 10 mg QD, 75 mg QD or placebo
                                           and pancreas (β-​cells);                  for 28 days; 24 h mean weighted glucose
                                           the stimulation of                        measurements decreased by 0.74 mmol/l
                                           GPCR119 has the                           and 1.05 mmol/l for 10 mg and 75 mg doses,
                                           potential to promote                      respectively, relative to placebo
                                           glucose-​dependent
                                           insulin secretion
                       AUC, area under the curve; GIP, glucose-​dependent insulinotropic polypeptide; GLP1, glucagon-​like peptide 1; GPCR119,
                       G-​protein-​coupled receptor 119; MTT, meal tolerance test; PD, pharmacodynamics; PK, pharmacokinetics; QD, daily; Q week,
                       dosed once per week; T2DM, type 2 diabetes mellitus. aGLP1 is secreted by the L cells in the small bowel in response to nutrient
                       ingestion. Modulated through a complex neural plexus, it augments glucose-​dependent insulin secretion, suppresses glucagon
                       and induces satiety, resulting in weight loss. GIP is secreted by intestinal K cells with food ingestion, has the potential to augment
                       insulin secretion and has been found to be deficient in people with T2DM. Glucagon has lipolytic and thermogenic capabilities.
                       The glucose-​raising effects of glucagon are constrained when used in combination with GLP1. bOxyntomodulin, like GLP1
                       and glucagon, is a peptide product derived from the post-​translational modification of preproglucagon. It is secreted by the
                       enteroendocrine cells of the small intestine in response to nutrient ingestion and has been shown in vitro to be a natural chimera,
                       binding and activating both the GLP1 receptor and the glucagon receptor.

                      Drugs that utilize the incretin axis                                  of drugs that act on the incretin axis is further amplified
                      The indirect stimulation of β-​cells is achievable through            by the ability of incretins to induce weight loss42.
                      drugs that affect the incretin axis (Table 2; Fig. 2). By defi-
                      nition, incretin hormones (for example, GLP1 and GIP)                 Dual-​a cting or triple-​a cting incretin mimetics.
                      are hormones secreted by enteroendocrine cells in the                 Importantly and unlike GKAs and GPCR40 agonists,
                      gastrointestinal tract in response to the oral ingestion of           insulin secretion invoked by incretin mimetics is glu-
                      nutrients. These hormones act to decrease the plasma                  cose dependent; that is, the plasma concentrations of
                      concentrations of glucose by mediating delayed gastric                glucose must be elevated or rising for insulin secretion
                      emptying, augmenting insulin secretion and suppressing                to be stimulated. GLP1 receptor agonists have been
                      glucagon secretion. Of note, the glucose-​lowering ability            used in T2DM care for the past 15 years to decrease

naTure RevIews | EnDoCRinology                                                                                                 volume 17 | June 2021 | 369

                                                         0123456789();:
Reviews

                              plasma concentrations of glucose and body weight43 and,             rather than the supplementation of GLP1 and GIP is
                              from 2016, to provide protection from cardiovascular                being tested using analogues of GPCR119; one drug
                              disease13,14. Expanded use of the incretin axis includes            is in phase I trials and another is in phase II47,48.
                              the development of molecules that act as both GLP1                      The well-​established ability of glucagon to stimulate
                              receptor agonists and GIP receptor agonists and/or as               hepatic glucose production makes it a counterintuitive
                              glucagon receptor agonists. Such dual or triple agonist             choice as a treatment for T2DM. The rationale for inte-
                              drugs aim to achieve even greater efficacy for glucose              grating the pharmacology of GLP1 and glucagon relies
                              lowering, greater body weight loss and perhaps greater              upon the action of GLP1 to restrain the hyperglycae-
                              cardiovascular protection in patients with T2DM than                mic effect of glucagon while adding a centrally medi-
                              provided by single agonists.                                        ated anorectic action to synergize with the lipolytic and
                                  In addition to their role in treating T2DM, dual-​              thermogenic capabilities of glucagon with the aim to
                              acting or triple-​a cting agonists have the potential               substantially decrease body weight49.
                              to attain independent indications for obesity, sleep
                              apnoea, renal insufficiency, non-​alcoholic fatty liver             Oxyntomodulin analogues. Oxyntomodulin, like GLP1
                              disease (NAFLD) and non-​alcoholic steatohepatitis in               and glucagon, is a peptide product derived from the
                              people with or without T2DM. Currently, drugs in clin-              post-​t ranslational modification of preproglucagon.
                              ical trials include one dual-​acting GLP1–GIP receptor              Oxyntomodulin is secreted by the enteroendocrine
                              agonist in phase III44, two dual-​acting GLP1–glucagon              cells of the small intestine in response to nutrient inges-
                              receptor agonists in phase II45,46, and two triple-​acting          tion and has been shown in vitro to be a natural chi-
                              GLP1–GIP–glucagon receptor agonists in phase I trials               mera, binding and activating both GLP1 and glucagon
                              (Table 2). The clinical rationale for combining these               receptors50. The dual stimulation of GLP1 and gluca­
                              dual-​acting or triple-​acting peptides is mainly to poten-         gon receptors is currently being tested for T2DM using
                              tiate the weight loss effects as it is unlikely that the HbA1c      an analogue of oxyntomodulin (one drug in phase II
                              reductions observed will be much greater than the                   trials)50. Similar to dual-​acting GLP1–glucagon recep-
                              1.5% or even 2% that have already been achieved with                tor agonists, oxyntomodulin exerts its glucoregulatory
                              compounds like semaglutide (approved) or tirzepatide                actions independent of but in addition to considerable
                              (phase III), respectively. In addition, the stimulation             weight loss50.

                               Table 3 | Drugs that modulate hepatic glucose metabolism
                               Compound                    Mechanism of action             Trial        Results                              Ref. or clinical
                                                                                           phase                                             trial number
                               Glucagon receptor antagonist
                               LGD-6972                    Inhibits glucagon action        II           PK/PD established                    54

                               REMD-477                                                    I–II         No published data                    NCT02455011
                               Antisense oligonucleotide inhibitor for the glucagon receptor
                               ISIS-449884 (GCGRRx)        Inhibits hepatic glucagon       II           Three phase II randomized,           55

                                                           receptor expression and                      double-​blind studies exposed
                                                           hepatic glycogenolysis                       people with T2DM to
                                                                                                        ISIS-449884 50–200 mg SC
                                                                                                        weekly for 13–26 weeks; HbA1c
                                                                                                        –0.9% to 2.0% with the various
                                                                                                        doses and a dose-​dependent
                                                                                                        increase in transaminases was
                                                                                                        observed
                               DACRAs
                               KBP-042                     Calcitonin stimulates           II           No published data                    NCT03230786
                                                           amylin secretion; amylin
                               KBP089                      lowers glucose levels           I            No published data                    NCT03907202
                                                           through delayed gastric
                                                           emptying and suppressing
                                                           glucagon secretion
                               GKAs
                               PF-04991532                 Liver-​selective GKA; GKA       II           PK/PD, safety established in         NCT01129258,
                                                           in liver increases hepatic                   T2DM                                 NCT01469065,
                                                           glucose uptake                                                                    NCT01336738,
                                                                                                                                             NCT01338870,
                                                                                                                                             NCT01369602
                               GK1-399 (formerly           Liver-​selective GKA            II           No published data                    NCT01474083,
                               TTP399)                                                                                                       NCT02405260,
                                                                                                                                             NCT01474083
                               DACRA, dual amylin–calcitonin receptor agonist; GKA, glucokinase activator; PD, pharmacodynamics; PK, pharmacokinetics;
                               SC, subcutaneous; T2DM, type 2 diabetes mellitus.

370 | June 2021 | volume 17                                                                                                            www.nature.com/nrendo

                                                                 0123456789();:
Reviews

                               The therapeutic potential of the incretin axis. Whether                incretin axis is not only being pursued for the treatment
                               by a new dual or triple agonist therapy or by established              of T2DM but also for the treatment of NAFLD and non-​
                               GLP1 receptor agonists, the utilization of the incre-                  alcoholic steatohepatitis51,52. Without question, the sub-
                               tin axis holds particular promise for the treatment of                 stantial weight loss observed with these novel therapies
                               T2DM and its common comorbidities. As mentioned                        is responsible for much of their metabolic benefits and
                               previously, the indirect, glucose-​dependent stimulation               probably extends to non-​metabolic benefits as well (for
                               of insulin secretion avoids the hypoglycaemia and dis-                 example, improvements in sleep apnoea and mobility).
                               rupted lipid metabolism that are seen with GKAs and                    Furthermore, expanded use of the incretin axis for ther-
                               GPCR40 agonists. By contrast, expanded use of the                      apeutic purposes has provided unique insights into the

                                                         Insulin                             Liver                              Muscle
                                                                                                                                              GH–ghrelin     Ghrelin
           PTP1B antisense                                                                                                                    co-agonist    analogue
           oligonucleotide                                                                  Glucagon              LYN kinase
               inhibitors                                Insulin           Glucagon           Glucose              activator
                                                         receptor          receptor
                                                                                                 GLUT2                                   GLUT4

                                                                                                                                      GH receptor
             PTP1B mRNA                                                        ATP       cAMP                     LYN kinase
                                                                                                                                                γ β
                                                                    PIP2
                               PTP1B            IRS1/2       PI3K                            ↓ G6Pase                                                  αq
          Mitochondrion                                                                                     Translocation
                                                                    PIP3                     ↓ FBPase

                                                 PKB               PDK1/2                            Pyruvate
             ATP:AMP                                                                                                                                  PLC
                                                                                                GLUT4
                             LKB                                                             facilitative
                                                                                             transporter                 Synaptic
              AMPK                                                                                                       vesicle
                                                                                              stimulator

               ACC
                                                                                                                                    Glucose
                                                                                                                                    uptake
       mTORC1                  MAPK cascade

                                        • Gluconeogenesis                    Transcription of genes             ↑ Glucose uptake,                  Ca2+ influx
                                        • Lipid synthesis                    involved in energy                 metabolism and
                                        • Lactate production                 storage and expenditure            glycolysis
                                                                                                                                      Endoplasmic
                                                                                                                                      reticulum

                     PPAR agonists

                                          PPARs RXR

                                                                                                                               Hepatocyte or muscle cell

Fig. 3 | Drugs that improve insulin sensitivity. Several new drugs are in             action of these drugs is not fully understood but they are thought to
development that improve insulin sensitivity through various mechanisms               increase endoplasmic reticulum calcium influx, thereby stimulating
of action. For example, the antisense oligonucleotide inhibitor specific for          physiological IGF1 production in the liver, which in turn increases insulin
protein tyrosine phosphatase 1B (PTP1B) mRNA disinhibits insulin signalling           action. Not shown on the figure are agents that target fibroblast growth
by reducing the translation of the negative regulator PTP1B. Furthermore,             factor 21 (FGF21) or inhibit diacylglycerol acetyl transferase 1 (DGAT1) and
novel peroxisome proliferator-​activated receptor (PPAR) agonists are being           microsomal triglyceride transfer protein (MTP). These drugs all act as
developed that stimulate the transcription of genes involved in energy                presumed insulin sensitizers through altering lipid metabolism. In addition,
storage and expenditure. In skeletal muscle, a selective allosteric activator         an agent targeting the type II B activin receptor is also thought to be an
of LYN kinase and a glucose transporter type 4 (GLUT4) facilitative                   insulin sensitizer through an unknown mechanism. Anti-​inflammatory
transporter stimulator both act to increase the translocation of GLUT4 to             agents, including a nuclear factor-​κB (NF-​κB) inhibitor and agent that
the plasma membrane. Finally, a ghrelin analogue and ghrelin–growth                   upregulates the nicotinic α7 receptor, are also in development. Adapted
hormone (GH) receptor agonist have been developed; the mechanism of                   from ref.106, Springer Nature Limited.

naTure RevIews | EnDoCRinology                                                                                                        volume 17 | June 2021 | 371

                                                                   0123456789();:
Reviews

 Table 4 | Drugs that improve insulin sensitivity
 Compound                        Mechanism of action                            Trial     Results                                            Ref. or clinical
                                                                                phase                                                        trial number
 Antisense oligonucleotide inhibitor for PTP1B mRNA
 ISIS 113715                     PTP1B has been shown to be a negative          III       92 people with T2DM randomized to ISIS             83

 (IONIS-​PTP-1BRx)               regulator of insulin and leptin signalling;              113715 200 mg SC Q week or placebo for
                                 the inhibition of PTP1B enhances insulin                 26 weeks; HbA1c –0.44% with ISIS 113715
                                 and leptin action
 FGF21 analogue
 LY2405319                       FGF21 has been shown to improve whole          I         47 people with T2DM randomized to                  66

                                 body insulin sensitivity, enhance insulin                LY2405319 at 3 mg, 10 mg or 20 mg QD for
                                 secretion while suppressing glucagon                     28 days; LY2405319 lowered LDL-​cholesterol,
                                 secretion, inhibit hepatic lipogenesis,                  triglycerides, fasting insulin and body weight
                                 and increase energy expenditure via the                  while increasing adiponectin but little effect
                                 activation of brown adipose tissue                       on plasma concentrations of glucose was
                                                                                          observed
 SRT2379                                                                        I         No published data                                  NCT01018628
 SAR425899                                                                      I         No published data                                  NCT03414736,
                                                                                                                                             NCT02973321,
                                                                                                                                             NCT02411825
 DGAT1 inhibitor
 PF-04620110                     Inhibits the acetylation of diacylglycerol     I         PK/PD established in T2DM                          NCT01298518
                                 to triacylglycerol; the ectopic deposition
                                 of triacylglycerol has been linked to
                                 insulin resistance
 Enterocytic MTP inhibitor
 KD026 (SLx-4090)                Inhibits the production of apo B               II        No published data                                  NCT02434744,
                                 containing lipoproteins in the gut;                                                                         NCT00871936,
                                 by-​products stimulate insulin signalling                                                                   NCT02434744
                                 independent of the insulin receptor
 Novel PPAR agonists
 CS038 (chiglitazar)             Dual PPARα/γ agonist; intranuclear             III       No published data                                  NCT02173457,
                                 transcription factors that lower plasma                                                                     NCT02121717
                                 levels of triglycerides and differentiate
                                 preadipocytes into mature adipocytes,
                                 respectively
 T0903131 (formerly              Partial PPARγ agonist; weak intranuclear       I         367 people with T2DM randomized to                 NCT00952445,
 INT-131; besylate)              transcription factor that differentiates                 T0903131 0.5 mg, 1 mg, 2 mg or 3 mg QD             NCT00631007
                                 pre-​adipocytes into mature adipocytes                   versus placebo for 24 weeks; dose-​dependent
                                                                                          HbA1c –0.3% to –1.0% in T0903131
 Other mechanisms
 DS-1150b                        GLUT4 facilitative transporter stimulator;     I         No published data                                  NCT02004678
                                 improves insulin signalling
 CAT-1004 (edasalonexent)        Anti-​inflammatory                             I         No published data                                  NCT01511900
 MLR-1023 (tolimidone)           Selective allosteric activator of LYN          I         No published data                                  NCT03279263,
                                 kinase; improves insulin signalling                                                                         NCT02317796
 TC-6987                         Nicotinic α7 receptor ligand;                  II        No published data                                  NCT01293669
                                 anti-​inflammatory
 AZP-531 (livoletide)            Ghrelin analogue; inhibits food intake         1         No published data                                  NCT02040012
                                 and induces weight loss
 TH-9507 (tesamorelin            Ghrelin–GH receptor agonist;                   2         53 people with T2DM randomized to TH-9507          NCT01264497
 or Egrifta)                     GH-​releasing hormone analogue;                          1 mg or 2 mg SC QD or placebo for 12 weeks;
                                 decreases visceral adipose tissue                        no difference in HbA1c was observed74
                                 accumulation
 S707106                         Unknown mechanism of action                    2         No published data                                  NCT01154348,
                                                                                                                                             NCT01240759
 Bimagrumab                      Type II-​B activin receptor modulator;         2         No published data                                  NCT03005288
                                 activates muscle growth                                                                                     (ref.80)
 DGAT1, diacylglycerol acetyl transferase 1; FGF21, fibroblast growth factor 21; GH, growth hormone; MTP, microsomal triglyceride transfer protein; PD,
 pharmacodynamics; PK, pharmacokinetics; PPAR, peroxisome proliferator-​activated receptor; PTP1B, protein tyrosine phosphatase 1B; QD, daily; Q week, dosed
 once per week; SC, subcutaneous; T2DM, type 2 diabetes mellitus.

372 | June 2021 | volume 17                                                                                                          www.nature.com/nrendo

                                                                   0123456789();:
Reviews

                      complex interplay between diet, enteroendocrine cells            tissue-​selective but could be designed to be so. Proven
                      and the microbiome in human health and disease53.                safety with DACRAs would strengthen this hypothesis,
                      Collectively, the knowledge to be gained from and the            particularly if neither hypoglycaemia nor liver toxicity
                      therapeutic potential of the incretin axis is vast.              are observed. The development of partial and/or tissue-​
                                                                                       selective glucagon receptor antagonists might prove a
                      Drugs targeting metabolism in the liver                          worthy pursuit.
                      Plasma levels of glucose can be decreased using thera-
                      peutics that target glucose metabolism in the liver. These       Drugs that improve insulin sensitivity
                      agents can target the liver directly, either by modulating       Perhaps the greatest need for new therapies in T2DM
                      the action of glucagon in hepatocytes to reduce hepatic          falls in the area of insulin sensitization and the
                      glucose production or by activating glucokinase to               approaches of the pharmaceutical industry to meeting
                      increase glucose uptake by hepatocytes. Alternatively,           this need could not be more diverse. Unfortunately, most
                      these drugs can reduce glucagon secretion from islets,           of these approaches to date do not seem particularly
                      thereby indirectly reducing hepatic glucose production           promising for decreasing levels of glucose in patients
                      (Table 3; Fig. 2).                                               with T2DM (Table 4; Fig. 3).

                      Antagonizing glucagon action. Whether endogenous                 Targeting FGF21. Considerable interest has been
                      or exogenous, glucagon induces hepatic glycogenolysis            shown in FGF21, a hepatokine that activates the NAD-​
                      leading to a rapid increase in plasma concentrations of          dependent protein deacetylase sirtuin 1 (three drugs are
                      glucose. Thus, it is conceivable that blocking the actions       currently in phase I trials) (Table 4). FGF21 is thought
                      of glucagon might lead to improved glucose control.              to be an insulin sensitizer through its ability to mitigate
                      As a single agent, glucagon action in patients with T2DM         FFA-​induced insulin resistance60. Collectively, investiga-
                      must be antagonized so as to not exacerbate hypergly-            tions have revealed FGF21 as a stimulator of fatty acid
                      caemia. Indeed, this strategy is being approached in a           oxidation, particularly in the liver, that increases the
                      number of ways; for example, two drugs that are direct           production of ketone bodies and inhibits lipogenesis61,62.
                      glucagon receptor antagonists are currently in phase II          Despite reports of circulating levels of FGF21 being
                      trials54. Furthermore, RNA interference gene therapy has         higher in people with any combination of T2DM63,
                      been utilized for this purpose through the creation of           NAFLD64 or obesity than in individuals without these
                      an antisense oligonucleotide inhibitor for the glucagon          conditions, people treated with FGF21 analogues show
                      receptor (one drug is in a phase II trial)55. The use of         weight loss65. Nevertheless, a proof-​of-​concept study
                      antisense RNA antagonizes the action of glucagon by              in people with T2DM treated with FGF21 analogues
                      blocking the transcription of the gene that encodes the          observed a decrease in the plasma levels of lipids but
                      glucagon receptor.                                               not in plasma concentrations of glucose66.
                          Another strategy to indirectly target the hepatic
                      actions of glucagon involves the use of a DACRA (one             Inhibitors of DGAT1 and MTP. One inhibitor of DGAT1
                      drug is in a phase I trial and another is in a phase II trial)   is currently in phase I trials and one inhibitor of MTP
                      (Table 3). The rationale for DACRAs stems from using             is in phase II trials. The rationale for developing these
                      the action of calcitonin to stimulate the amylin recep-          agents lies in the assumption that altering lipid metabo-
                      tor on β-​cells leading to the co-​secretion of amylin and       lism will favourably affect glucose metabolism. However,
                      insulin. Amylin itself decreases blood concentrations            preclinical studies have yet to show that this scenario
                      of glucose by delaying gastric emptying56 as well as by          is the case67,68 and there might be reason to suspect
                      inhibiting post-​prandial glucagon secretion57. Although         that a glucose-​lowering ability by these drugs will not
                      each agent in this group has demonstrated promise in             be demonstrated after all. For example, the inhibition
                      decreasing levels of glucose in humans, their specific           of DGAT1 might indeed decrease the tissue accumu-
                      effect on suppressing hepatic glucose production, per se,        lation of triacylglycerol (aka triglyceride); however,
                      is presumptive and is yet to be clearly proven.                  it does so at the expense of an increased tissue accumu-
                                                                                       lation of diacylglycerol, which is a far more inflamma-
                      Safety issues. Unfortunately, an increase in hepatic             tory, insulin de-​sensitizing lipid than triacylglycerol69,70.
                      steatosis and blood biomarkers of liver injury (transam-         Moreover, MTP acts in the liver and intestine to pre-
                      inases) have been reported in clinical trials with the           vent the transfer of triglycerides to other apo-​B con-
                      antisense oligonucleotide inhibitor as well as after direct      taining lipoproteins, thereby reducing post-​prandial
                      antagonism of the glucagon receptor58,59. These safety           hypertriglyceridaemia71,72. Nevertheless, a link is lack-
                      concerns have led to the abandonment of a number of              ing between decreasing plasma levels of triglycerides and
                      compounds in development (for example, Bay 27-9955,              insulin sensitization and/or decreases in plasma levels of
                      MK-0893, MK-3577 and LY-24090215). Most note-                    glucose in humans73.
                      worthy is that the inhibition of hepatic glycogenolysis,
                      whether by glucagon antagonism, GKAs or GPCR40                   Targeting the GH receptor. Speculation exists as to
                      agonists, increases hepatic de novo lipogenesis. The             whether the adverse effects of GH on glucose metabo-
                      physiological nature of this response is predictable but         lism might be offset by its anabolic action as well as by its
                      could be avoidable, for example, by developing tissue-​          ability to stimulate the production of insulin-​like growth
                      selective drugs that avoid the liver. To date, gluca-            factor 1 in the liver. The gastric-​derived peptide, ghre-
                      gon receptor antagonists in development are not                  lin, is an endogenous ligand for GH-​releasing hormone

naTure RevIews | EnDoCRinology                                                                                        volume 17 | June 2021 | 373

                                                       0123456789();:
Reviews

                              receptor. GH receptor agonism can be achieved either           insulin receptor substrate 1 in insulin-​responsive tissues,
                              directly using a GH–ghrelin receptor co-​agonist74 (one        which in return phosphorylates LYN kinase, suggesting
                              drug in phase II trials) or indirectly using a pure ghre-      a regulatory feedback loop between LYN kinase and
                              lin analogue75 (one drug in phase I trials) (Table 4). The     insulin receptor activation84. Interestingly, the insulin
                              potentiation of GH via ghrelin is unlikely to thrive in        secretagogue, glimepiride, harbours extra pancreatic
                              development as a treatment for T2DM owing to the ina-          action as an insulin sensitizer through the activation
                              bility of this mechanism to induce weight loss76,77 and        of LYN kinase; however, glimepiride is associated with
                              blunting of glucose-​stimulated insulin secretion78.           hypoglycaemia85. By contrast, hypoglycaemia has not
                                  The anabolic action of GH might be better achieved         been reported with the upregulation of insulin sig-
                              with a more direct approach such as type II-​B activin         nalling in peripheral tissues by other compounds that
                              receptor modulation (one drug in phase II trials).             specifically target LYN kinase currently in development.
                              Blockade of the type II-​B activin receptor inhibits the
                              actions of natural ligands that negatively regulate skeletal   PPAR agonists. The pipeline also revisits PPAR ago-
                              muscle growth, thereby leading to a preservation and           nists, striving to improve their efficacy and safety, either
                              even expansion of lean mass79. Bimagrumab, a monoclo-          through dual PPARα–PPARγ agonism86 (one drug in
                              nal antibody that inhibits the activin type II receptor, was   phase III trials) or through partial dual PPARα–PPARγ
                              shown in 2021 to increase lean mass, decrease fat mass         agonism (one drug in phase I trials) (Table 4). PPARα
                              and improve HbA1c in adults with T2DM80. Although the          agonists inhibit hepatic lipoprotein C-​III expression and
                              precise mechanism of action remains largely unknown,           subsequent VLDL production, making them useful as
                              it is clear that the anabolic action of bimagrumab is          single agents for decreasing plasma levels of triglyceride
                              distinct from GH and harbours favourable effects on            (for example, gemfibrozil and fenofibrate are approved
                              glucose metabolism.                                            for this indication). PPARγ agonists act as intranuclear
                                                                                             transcription factors that improve insulin sensitivity
                              Strategies that target inflammation. Insulin resistance is     by expanding subcutaneous adipose tissue depots and
                              widely regarded as a pro-​inflammatory state, although         inducing the differentiation of pre-​adipocytes into
                              it is unclear whether a unidirectional causative pathway       mature adipocytes (for example, rosiglitazone and pio-
                              exists or whether insulin resistance and inflammation          glitazone)87. Contemporary clinical trials have eased
                              are simply associated. Approaches targeting inflam-            historic concerns over the cardiovascular safety of
                              mation are attractive for their widespread benefits            some PPARγ single agonists88. However, thus far, none
                              on diabetes-​related complications but are too early in        of the dual PPARα–PPARγ agonists has followed suit
                              development to understand their efficacy and safety            with numerous development programmes halted owing
                              in T2DM. For example, the TINSAL trials (Targeting             to renal and cardiovascular concerns (for example,
                              Inflammation Using Salsalate in Type 2 Diabetes) exten-        aleglitazar89, muraglitazar90 and tesaglitazar91).
                              sively explored the role of the anti-​inflammatory agent           Altogether, pharmaceuticals in development for
                              salsalate as a treatment for T2DM. Results from these tri-     T2DM mainly directly or indirectly exploit one of the
                              als demonstrated HbA1c reduction but also raised safety        original ‘triumvirate’ of defects, that is, impaired insu-
                              concerns that precluded the introduction of salsalate          lin secretion, reduced insulin sensitivity and increased
                              to the market as an anti-​diabetic agent81,82. New com-        hepatic glucose production, described in T2DM many
                              pounds in the pipeline include a drug in phase I trials        years ago92. One agent in phase III clinical trials is
                              that inhibits NF-​κ B as well as a drug in phase II            imeglimin, a tetrahydrotriazine-​containing oxidative
                              trials that upregulates the nicotinic α7 receptor (Table 4).   phosphorylation blocker that might attend to the entire
                              The anti-​inflammatory mechanisms of action of these           triumvirate: stimulating insulin secretion, improving
                              compounds in development clearly differ from that of           insulin sensitivity and reducing hepatic glucose pro-
                              salsalate (inhibiting NF-​κB or activating the nicotinic α7    duction through the enhancement of mitochondrial
                              receptor ligand versus inhibiting cyclooxygenase) but are      bioenergetics93–96. However, the data remain highly
                              also likely to exert systemic effects.                         controversial.

                              Revisiting existing strategies. Remaining novel com-           Future directions
                              pounds in development for insulin sensitization for the        In reviewing the 7,484 trials registered in ClinicalTrials.gov
                              purpose of decreasing blood concentrations of glucose          that were generated using the key word ‘type 2 diabe-
                              in T2DM reminisce on existing themes. One approach             tes’, 43 novel therapies were identified. Through unique
                              seeks to upregulate insulin signalling in peripheral tis-      mechanisms of action that directly stimulate insulin
                              sues by inhibiting dephosphorylation of the insulin            secretion in the β-​cell, utilize the incretin axis, suppress
                              receptor using an antisense oligonucleotide inhibitor for      hepatic glucose production and/or improve insulin sen-
                              PTP1B mRNA (one drug in phase III trials)83. A second          sitivity, several recurring themes dominate the potential
                              approach seeks to completely bypass glucose dependency         of these novel therapies to move into clinical care. First,
                              by stimulating the translocation of the insulin-​dependent     clinical trials continue to reveal important safety signals,
                              glucose facilitative transporter (GLUT4) directly (one         most notably liver toxicity. Given the exquisite ability of
                              drug in phase I trials) (Table 4). A third strategy aims at    the human body to shift substrate utilization and storage,
                              the indirect potentiation of insulin action through the        it is clearly important to recognize that the mechanism
                              allosteric activation of LYN kinase (one drug in phase I       by which one decreases plasma concentrations of glu-
                              trials) (Fig. 3). Activated LYN kinase can phosphorylate       cose in T2DM is probably as important as the absolute

374 | June 2021 | volume 17                                                                                                     www.nature.com/nrendo

                                                                0123456789();:
Reviews

                                     decrease achieved. For example, drugs in development                          patient characteristics, risk of complications and expo-
                                     that block hepatic glucose output decrease plasma lev-                        sure to glucose-​lowering medications97. Several genetic
                                     els of glucose at the expense of hepatic steatosis. Future                    variants and genetic risk scores were differentially
                                     compounds could avoid these issues if designed to be                          associated with the clusters; however, they were not
                                     tissue selective. Second, dual and triple agonist thera-                      examined for their independent association with com-
                                     pies harbour the potential to minimize adverse effects                        plications or drug exposure. Of note, genetic variabil-
                                     as smaller doses can be used and the different agonists                       ity has been shown to alter the responses of patients to
                                     can also exert synergistic effects with each other. This                      sulfonylureas99, metformin100 and thiazolidinediones101,
                                     advantage is best exemplified by compounds in develop-                        suggesting that the same might be true for the newer and
                                     ment that utilize the incretin axis. Incretin-​induced insu-                  emerging medical therapies. Most importantly, future
                                     lin secretion is indirect and glucose dependent, thereby                      studies could unveil genetic predictors of patients with
                                     avoiding unnecessary hypoglycaemia while invoking the                         an improved response to a glucose-​lowering medication
                                     gut microbiome and its metabolites to diversify the drug                      as well as of patients who are likely to have complications
                                     response53. Most certainly, therapies that facilitate weight                  prevented by such medications. Fortunately, comprehen-
                                     loss will treat much more than T2DM.                                          sive modelling approaches are under way102 to bring this
                                                                                                                   collective science from the bench to the bedside.
                                     Precision mechanisms
                                     As the treatments for T2DM continue to diversify, our                         Conclusions
                                     ability to deliver precision health care is becoming real-                    Novel therapies currently in development for the treat-
                                     ized; drugs with different mechanisms of action can be                        ment of T2DM directly stimulate insulin secretion in
                                     combined to produce the best possible treatment out-                          the β-​cell, utilize the incretin axis, suppress hepatic glu-
                                     comes in individual patients. Undoubtedly, guidelines                         cose production and/or improve insulin sensitivity via
                                     will continue to integrate emerging data that focus on                        unique mechanisms of action. In our judgement, com-
                                     reaching individualized glycaemic targets while optimiz-                      pounds with the most promise — by virtue of validated
                                     ing safety, non-​glycaemic benefits, and the prevention                       mechanisms of action and advancement in clinical trials
                                     of microvascular and macrovascular complications for                          by meeting preliminary efficacy and safety end points
                                     individual patients at risk of specific complications20.                      — include dual-​acting GLP1–GIP receptor agonists,
                                     To achieve these aims, considerable work is under way to                      dual-​acting GLP1–glucagon receptor agonists, and
                                     predict which patients are most likely to benefit from the                    perhaps triple-​acting GLP1–GIP–glucagon receptor ago-
                                     various non-​glycaemic effects and prevention of specific                     nists and oxyntomodulin. Each is fully expected to attain
                                     complications.                                                                individualized glycaemic targets while optimizing safety,
                                         Revelations have come from both clinical biomarkers                       off-​target benefits and the prevention of complications,
                                     and phenotyping97, as well as from pharmacogenomics98.                        thereby fuelling hope that they might also treat a host of
                                     For example, one data-​driven cluster analysis of nearly                      obesity-​related complications beyond T2DM.
                                     9,000 people with new-​onset T2DM identified five rep-
                                     licable phenotypic clusters with substantially different                      Published online 4 May 2021

1.   National Diabetes Data Group. Classification and           10. Nissen, S. E. & Wolski, K. Effect of rosiglitazone               the American Diabetes Association (ADA) and the
     diagnosis of diabetes mellitus and other categories of         on the risk of myocardial infarction and death                   European Association for the Study of Diabetes
     glucose intolerance. Diabetes 28, 1039–1057 (1979).            from cardiovascular causes. N. Engl. J. Med. 356,                (EASD). Diabetes Care 41, 2669–2701 (2018).
2.   World Health Organization. Report of the Expert                2457–2471 (2007).                                          21.   Rosenstock, J. et al. Efficacy and safety of ITCA 650,
     Committee on Diabetes. WHO https://apps.who.int/           11. Gerstein, H. C. et al. Dulaglutide and cardiovascular            a novel drug-​device GLP-1 receptor agonist, in type 2
     iris/handle/10665/41399 (1980).                                outcomes in type 2 diabetes (REWIND): a double-​                 diabetes uncontrolled with oral antidiabetes drugs:
3.   The Diabetes Control and Complications Trial                   blind, randomised placebo-​controlled trial. Lancet              the FREEDOM-1 trial. Diabetes Care 41, 333–340
     Research Group et al. The effect of intensive treatment        394, 121–130 (2019).                                             (2018).
     of diabetes on the development and progression of          12. Hernandez, A. F. et al. Albiglutide and cardiovascular     22.   American Diabetes Association. Pharmacologic
     long-​term complications in insulin-​dependent diabetes        outcomes in patients with type 2 diabetes and                    approaches to glycemic treatment: standards of
     mellitus. N. Engl. J. Med. 329, 977–986 (1993).                cardiovascular disease (Harmony Outcomes):                       medical care in diabetes-2019. Diabetes Care 42,
4.   UK Prospective Diabetes Study (UKPDS) Group.                   a double-​blind, randomised placebo-​controlled trial.           S90–S102 (2019).
     Intensive blood-​glucose control with sulphonylureas           Lancet 392, 1519–1529 (2018).                              23.   Mahajan, A. et al. Fine-​mapping type 2 diabetes
     or insulin compared with conventional treatment            13. Marso, S. P. et al. Semaglutide and cardiovascular               loci to single-​variant resolution using high-​density
     and risk of complications in patients with type 2              outcomes in patients with type 2 diabetes. N. Engl.              imputation and islet-​specific epigenome maps.
     diabetes (UKPDS 33). Lancet 352, 837–853 (1998).               J. Med. 375, 1834–1844 (2016).                                   Nat. Genet. 50, 1505–1513 (2018).
5.   Action to Control Cardiovascular Risk in Diabetes          14. Marso, S. P. et al. Liraglutide and cardiovascular         24.   Zelent, D. et al. Glucokinase and glucose homeostasis:
     Study Group et al. Effects of intensive glucose lowering       outcomes in type 2 diabetes. N. Engl. J. Med. 375,               proven concepts and new ideas. Biochem. Soc. Trans.
     in type 2 diabetes. N. Engl. J. Med. 358, 2545–2559            311–322 (2016).                                                  33, 306–310 (2005).
     (2008).                                                    15. Neal, B. et al. Canagliflozin and cardiovascular and       25.   Matschinsky, F. M. Assessing the potential of
6.   Duckworth, W. et al. Glucose control and vascular              renal events in type 2 diabetes. N. Engl. J. Med. 377,           glucokinase activators in diabetes therapy. Nat. Rev.
     complications in veterans with type 2 diabetes.                644–657 (2017).                                                  Drug Discov. 8, 399–416 (2009).
     N. Engl. J. Med. 360, 129–139 (2009).                      16. Wiviott, S. D. et al. Dapagliflozin and cardiovascular     26.   Matschinsky, F. M. & Wilson, D. F. The central role of
7.   The ADVANCE Collaborative Group et al. Intensive               outcomes in type 2 diabetes. N. Engl. J. Med. 380,               glucokinase in glucose homeostasis: a perspective
     blood glucose control and vascular outcomes in                 347–357 (2019).                                                  50 years after demonstrating the presence of the enzyme
     patients with type 2 diabetes. N. Engl. J. Med. 358,       17. Zinman, B. et al. Empagliflozin, cardiovascular                  in islets of Langerhans. Front. Physiol. 10, 148 (2019).
     2560–2572 (2008).                                              outcomes, and mortality in type 2 diabetes. N. Engl.       27.   Bonadonna, R. C. et al. Piragliatin (RO4389620),
8.   American Diabetes Association. Glycemic targets:               J. Med. 373, 2117–2128 (2015).                                   a novel glucokinase activator, lowers plasma glucose
     standards of medical care in diabetes-2020.                18. McMurray, J. J. V. et al. Dapagliflozin in patients with         both in the postabsorptive state and after a
     Diabetes Care 43 (Suppl. 1), S66–S76 (2020).                   heart failure and reduced ejection fraction. N. Engl.            glucose challenge in patients with type 2 diabetes
9.   Garber, A. J. et al. Consensus statement by the                J. Med. 381, 1995–2008 (2019).                                   mellitus: a mechanistic study. J. Clin. Endocrinol.
     American Association of Clinical Endocrinologists          19. Packer, M. et al. Cardiovascular and renal outcomes              Metab. 95, 5028–5036 (2010).
     and American College of Endocrinology on the                   with empagliflozin in heart failure. N. Engl. J. Med.      28.   Katz, L. et al. AMG 151 (ARRY-403), a novel
     comprehensive type 2 diabetes management                       383, 1413–1424 (2020).                                           glucokinase activator, decreases fasting and
     algorithm - 2019 executive summary. Endocr. Pract.         20. Davies, M. J. et al. Management of hyperglycemia                 postprandial glycaemia in patients with type 2
     25, 69–100 (2019).                                             in type 2 diabetes, 2018. A consensus report by                  diabetes. Diabetes Obes. Metab. 18, 191–195 (2016).

naTure RevIews | EnDoCRinology                                                                                                                         volume 17 | June 2021 | 375

                                                                            0123456789();:
You can also read