Archives

Introduction to Lipids and Lipoproteins

ABSTRACT

 

Cholesterol and triglycerides are insoluble in water and therefore these lipids must be transported in association with proteins. Lipoproteins are complex particles with a central core containing cholesterol esters and triglycerides surrounded by free cholesterol, phospholipids, and apolipoproteins, which facilitate lipoprotein formation and function. Plasma lipoproteins can be divided into seven classes based on size, lipid composition, and apolipoproteins (chylomicrons, chylomicron remnants, VLDL, VLDL remnants (IDL), LDL, HDL, and Lp (a)).  Chylomicron remnants, VLDL, IDL, LDL, and Lp (a) are all pro-atherogenic while HDL is anti-atherogenic. Apolipoproteins have four major functions including 1) serving a structural role, 2) acting as ligands for lipoprotein receptors, 3) guiding the formation of lipoproteins, and 4) serving as activators or inhibitors of enzymes involved in the metabolism of lipoproteins. The exogenous lipoprotein pathway starts with the incorporation of dietary lipids into chylomicrons in the intestine. In the circulation, the triglycerides carried in chylomicrons are metabolized in muscle and adipose tissue by lipoprotein lipase releasing free fatty acids, which are subsequently metabolized by muscle and adipose tissue, and chylomicron remnants are formed. Chylomicron remnants are then taken up by the liver. The endogenous lipoprotein pathway begins in the liver with the formation of VLDL. The triglycerides carried in VLDL are metabolized in muscle and adipose tissue by lipoprotein lipase releasing free fatty acids and IDL are formed. The IDL are further metabolized to LDL, which are taken up by the LDL receptor in numerous tissues including the liver, the predominant site of uptake. Reverse cholesterol transport begins with the formation of nascent HDL by the liver and intestine. These small HDL particles can then acquire cholesterol and phospholipids that are effluxed from cells, a process mediated by ABCA1 resulting in the formation of mature HDL.  Mature HDL can acquire addition cholesterol from cells via ABCG1, SR-B1, or passive diffusion. The HDL then transports the cholesterol to the liver either directly by interacting with hepatic SR-B1 or indirectly by transferring the cholesterol to VLDL or LDL, a process facilitated by CETP. Cholesterol efflux from macrophages to HDL plays an important role in protecting from the development of atherosclerosis.

 

INTRODUCTION

 

Because lipids, such as cholesterol and triglycerides, are insoluble in water these lipids must be transported in association with proteins (lipoproteins) in the circulation. Large quantities of fatty acids from meals must be transported as triglycerides to avoid toxicity. These lipoproteins play a key role in the absorption and transport of dietary lipids by the small intestine, in the transport of lipids from the liver to peripheral tissues, and the transport of lipids from peripheral tissues to the liver and intestine (reverse cholesterol transport). A secondary function is to transport toxic foreign hydrophobic and amphipathic compounds, such as bacterial toxins, from areas of invasion and infection (1). For example, lipoproteins bind endotoxin (LPS) from gram negative bacteria and lipoteichoic acid from gram positive bacteria thereby reducing their toxic effects (1). In addition, apolipoprotein L1, associated with HDL particles, has lytic activity against the parasite Trypanosoma brucei brucei and lipoproteins can neutralize viruses (2,3). Thus, while this article will focus on the transport properties of lipoproteins the reader should recognize that lipoprotein may have other important roles.

 

STRUCTURE OF LIPOPROTEINS (4)

 

Lipoproteins are complex particles that have a central hydrophobic core of non-polar lipids, primarily cholesterol esters and triglycerides. This hydrophobic core is surrounded by a hydrophilic membrane consisting of phospholipids, free cholesterol, and apolipoproteins (Figure 1). Plasma lipoproteins are divided into seven classes based on size, lipid composition, and apolipoproteins (Table 1 and Figure 2).

 

Figure 1. Lipoprotein Structure (figure modified from Biochemistry 39: 9763, 2000)

 

 

Table 1. Lipoprotein Classes

Lipoprotein

Density (g/ml)

Size (nm)

Major Lipids

Major Apoproteins

Chylomicrons

<0.930

75-1200

Triglycerides

Apo B-48, Apo C, Apo E, Apo A-I, A-II, A-IV

Chylomicron Remnants

0.930- 1.006

30-80

Triglycerides Cholesterol

Apo B-48, Apo E

VLDL

0.930- 1.006

30-80

Triglycerides

Apo B-100, Apo E, Apo C

IDL

1.006- 1.019

25-35

Triglycerides Cholesterol

Apo B-100, Apo E, Apo C

LDL

1.019- 1.063

18- 25

Cholesterol

Apo B-100

HDL

1.063- 1.210

5- 12

Cholesterol Phospholipids

Apo A-I, Apo A-II, Apo C, Apo E

Lp (a)

1.055- 1.085

~30

Cholesterol

Apo B-100, Apo (a)

 

Figure 2: Classes of Lipoproteins (figure modified from Advances Protein Chemistry 45:303, 1994)

 

Chylomicrons (5)

 

These are large triglyceride rich particles made by the intestine, which are involved in the transport of dietary triglycerides and cholesterol to peripheral tissues and liver. These particles contain apolipoproteins A-I, A-II, A-IV, A-V, B-48, C-II, C-III, and E. Apo B-48 is the core structural protein and each chylomicron particle contains one Apo B-48 molecule. The size of chylomicrons varies depending on the amount of fat ingested. A high fat meal leads to the formation of large chylomicron particles due to the increased amount of triglyceride being transported whereas in the fasting state the chylomicron particles are small carrying decreased quantities of triglyceride. The quantity of cholesterol carried by chylomicrons also can vary depending upon dietary intake.

 

Chylomicron Remnants (5-7)

 

The removal of triglyceride from chylomicrons by lipoprotein lipase in peripheral tissues results in smaller particles called chylomicron remnants. Compared to chylomicrons these particles are enriched in cholesterol and are pro-atherogenic.

 

Very Low-Density Lipoproteins (VLDL)

 

These particles are produced by the liver and are triglyceride rich. They contain apolipoprotein B-100, C-I, C-II, C-III, and E. Apo B-100 is the core structural protein and each VLDL particle contains one Apo B-100 molecule. Similar to chylomicrons the size of the VLDL particles can vary depending on the quantity of triglyceride carried in the particle. When triglyceride production in the liver is increased, the secreted VLDL particles are large. However, VLDL particles are smaller than chylomicrons.

 

Intermediate-Density Lipoproteins (IDL; VLDL Remnants) (6,7)

 

The removal of triglycerides from VLDL by muscle and adipose tissue results in the formation of IDL particles which are enriched in cholesterol. These particles contain apolipoprotein B-100 and E. These IDL particles are pro-atherogenic.

 

Low-Density Lipoproteins (LDL)

 

These particles are derived from VLDL and IDL particles and they are even further enriched in cholesterol. LDL carries the majority of the cholesterol that is in the circulation. The predominant apolipoprotein is B-100 and each LDL particle contains one Apo B-100 molecule. LDL consists of a spectrum of particles varying in size and density. An abundance of small dense LDL particles is seen in association with hypertriglyceridemia, low HDL levels, obesity, type 2 diabetes (i.e. patients with the metabolic syndrome) and infectious and inflammatory states. These small dense LDL particles are considered to be more pro-atherogenic than large LDL particles for a number of reasons (8). Small dense LDL particles have a decreased affinity for the LDL receptor resulting in a prolonged retention time in the circulation. Additionally, they more easily enter the arterial wall and bind more avidly to intra-arterial proteoglycans, which traps them in the arterial wall. Finally, small dense LDL particles are more susceptible to oxidation, which could result in an enhanced uptake by macrophages. 

 

High-Density Lipoproteins (HDL) (9-11)

 

These particles play an important role in reverse cholesterol transport from peripheral tissues to the liver, which is one potential mechanism by which HDL may be anti-atherogenic. In addition, HDL particles have anti-oxidant, anti-inflammatory, anti-thrombotic, and anti-apoptotic properties, which may also contribute to their ability to inhibit atherosclerosis. HDL particles are enriched in cholesterol and phospholipids. Apolipoproteins A-I, A-II, A-IV, C-I, C-II, C-III, and E are associated with these particles. Apo A-I is the core structural protein and each HDL particle may contain multiple Apo A-I molecules. In addition, using mass spectrometry proteins involved in proteinase inhibition, complement activation, and the acute-phase response have been found associated with HDL particles (12). HDL particles are very heterogeneous and can be classified based on density, size, charge, or apolipoprotein composition (Table 2).

 

Table 2. Classification of HDL

Method of classification

Types of HDL

Density gradient ultracentrifugation

HDL2, HDL3, very high-density HDL

Nuclear magnetic resonance

large, medium, and small

Gradient gel electrophoresis

HDL 2a, 2b, 3a, 3b, 3c

2-dimensional gel electrophoresis

pre-beta 1 and 2, alpha 1, 2, 3, 4

Apolipoprotein composition

A-I particles, A-I: A-II particles, A-I: E particles

 

Lipoprotein (a) (Lp (a)) (13-16)

 

Lp (a) is an LDL particle that has apolipoprotein (a) attached to Apo B-100 via a disulfide bond. Lp (a) contain Apo (a) and Apo B-100 in a 1:1 molar ratio. The size of Lp(a) particles can vary greatly based on the size of apolipoprotein (a). This particle is pro-atherogenic.

 

APOLIPOPROTEINS (17,18)

 

Apolipoproteins have four major functions including 1) serving a structural role, 2) acting as ligands for lipoprotein receptors, 3) guiding the formation of lipoproteins, and 4) serving as activators or inhibitors of enzymes involved in the metabolism of lipoproteins (Table 3). Apolipoproteins thus play a crucial role in lipoprotein metabolism.

 

Apolipoprotein A-I (19)

 

Apo A-I is synthesized in the liver and intestine and is the major structural protein of HDL accounting for approximately 70% of HDL protein. It also plays a role in the interaction of HDL with ATP-binding cassette protein A1 (ABCA1), ABCG1, and class B, type I scavenger receptor (SR-B1). Apo A-I is an activator of lecithin: cholesterol acyltransferase (LCAT), an enzyme that converts free cholesterol into cholesteryl ester. High levels of Apo A-I are associated with a decreased risk of atherosclerosis.

 

Apolipoprotein A-II (20)

 

Apo A-II is synthesized in the liver and is the second most abundant protein on HDL accounting for approximately 20% of HDL protein. The role of Apo A-II in lipid metabolism is unclear. Apo A-II is a strong predictor of risk for CVD.

 

Apolipoprotein A-IV (21)

 

Apo A-IV is synthesized in the intestine during fat absorption. Apo A-IV is associated with chylomicrons and high-density lipoproteins, but is also found in the lipoprotein-free fraction.  Its precise role in lipoprotein metabolism remains to be determined but studies have suggested a role for Apo A-IV in regulating food intake.

 

Apolipoprotein A-V (22,23)

 

Apo A-V is synthesized in the liver and associates with triglyceride rich lipoproteins. It is an activator of LPL mediated lipolysis and thereby plays an important role in the metabolism of triglyceride rich lipoproteins.

 

Apolipoprotein B-48 (24)

 

Apo B-48 is synthesized in the intestine and is the major structural protein of chylomicrons and chylomicron remnants. There is a single molecule of apo B-48 per chylomicron particle. There is a single apolipoprotein B gene that is expressed in both the liver and intestine. The intestine expresses a protein that is approximately ½ the size of the liver due to mRNA editing. The apobec-1 editing complex is expressed in the intestine and edits a specific cytidine to an uracil in the apo B mRNA in the intestine creating a stop codon that results in the cessation of protein translation and a shorter Apo B (Apo B-48). The portion of Apo-B that is recognized by the LDL receptor is not contained in Apo-B48 and therefore Apo B-48 is not recognized by the LDL receptor.

 

Apolipoprotein B-100

 

Apo B-100 is synthesized in the liver and is the major structural component of VLDL, IDL, and LDL. There is a single molecule of Apo B-100 per VLDL, IDL, LDL and Lp(a) particle. Apo B-100 is a ligand for the LDL receptor and therefore plays an important role in the clearance of lipoprotein particles. Certain mutations in Apo B-100 result in decreased binding to the LDL receptor and familial hypercholesterolemia (25). High levels of Apo B-100 are associated with an increased risk of atherosclerosis.

 

Apolipoprotein C (26-29)

 

The C apolipoproteins are synthesized primarily in the liver and freely exchange between lipoprotein particles and therefore are found in association with chylomicrons, VLDL, and HDL.

 

Apo C-II is a co-factor for lipoprotein lipase (LPL) and thus stimulates triglyceride hydrolysis and the clearance of triglyceride rich lipoproteins (26,29). Loss of function mutations in Apo C-II result in marked hypertriglyceridemia due to a failure to metabolize triglyceride rich lipoproteins (30).

 

Apo C-III is an inhibitor of LPL (31). Additionally, Apo C-III inhibits the interaction of triglyceride rich lipoproteins with their receptors (31). Recent studies have shown that loss of function mutations in Apo C-III lead to decreases in serum triglyceride levels and a reduced risk of cardiovascular disease. Interestingly, inhibition of Apo C-III expression results in a decrease in serum triglyceride levels even in patients deficient in lipoprotein lipase indicating that the ability of Apo C-III to modulate serum triglyceride levels is not dependent solely on regulating lipoprotein lipase activity (32).

 

Apolipoprotein E (33)

 

Apolipoprotein E is synthesized in many tissues but the liver and intestine are the primary source of circulating Apo E. Apo E exchanges between lipoprotein particles and is associated with chylomicrons, chylomicron remnants, VLDL, IDL, and a subgroup of HDL particles. There are three common genetic variants of Apo E (Apo E2, E3, and E4). ApoE2 differs from the most common isoform, Apo E3, by a single amino acid substitution where cysteine substitutes for arginine at residue 158. Apo E4 differs from Apo E3 at residue 112, where arginine substitutes for cysteine. Apo E3 and E4 are ligands for the LDL receptor while Apo E2 is poorly recognized by the LDL receptor. Patients who are homozygous for Apo E2 can develop familial dysbetalipoproteinemia (30). Apo E4 is associated with an increased risk of Alzheimer’s disease and an increased risk of atherosclerosis.

 

Apolipoprotein (a) (14,16)

 

Apo (a) is synthesized in the liver. This protein is a homolog of plasminogen and its molecular weight varies from 300,000 to 800,000. It is attached to Apo B-100 via a disulfide bond. High levels of Apo (a) are associated with an increased risk of atherosclerosis. Apo (a) is an inhibitor of fibrinolysis and can also enhance the uptake of lipoproteins by macrophages, both of which could increase the risk of atherosclerosis. The physiologic function of Apo (a) is unknown. Interestingly this apolipoprotein is found in primates but not in other species.

 

Table 3. Apolipoproteins

Apolipoprotein

MW

Primary Source

Lipoprotein Association

Function

Apo A-I

28,000

Liver, Intestine

HDL, chylomicrons

Structural protein for HDL, Activates LCAT

Apo A-II

17,000

Liver

HDL, chylomicrons

Structural protein for HDL, Activates hepatic lipase

Apo A-IV

45,000

Intestine

HDL, chylomicrons

Unknown

Apo A-V

39,000

Liver

VLDL, chylomicrons, HDL               

Promotes LPL mediated TG lipolysis

Apo B-48

241,000

Intestine

Chylomicrons

Structural protein for chylomicrons

Apo B-100

512,000

Liver

VLDL, IDL, LDL, Lp (a)

Structural protein, Ligand for LDL receptor

Apo C-I

6,600

Liver

Chylomicrons, VLDL, HDL

Activates LCAT

Apo C-II

8,800

Liver

Chylomicrons, VLDL, HDL

Co-factor for LPL

Apo C-III

8,800

Liver

Chylomicrons, VLDL, HDL

Inhibits LPL and uptake of lipoproteins

Apo E

34,000

Liver

Chylomicron remnants, IDL, HDL

Ligand for LDL receptor

Apo (a)

250,000- 800,00

Liver

Lp (a)

Inhibits plasminogen activation

                                

LIPOPROTEIN RECEPTORS AND LIPID TRANSPORTERS

 

There are several receptors and transporters that play a crucial role in lipoprotein metabolism.

 

LDL Receptor (34)

 

The LDL receptor is present in the liver and most other tissues. It recognizes Apo B-100 and Apo E and hence mediates the uptake of LDL, chylomicron remnants, and IDL, which occurs via endocytosis (Figure 3). After internalization, the lipoprotein particle is degraded in lysosomes and the cholesterol is released. The delivery of cholesterol to the cell decreases the activity of HMGCoA reductase and other enzymes required for the biosynthesis of cholesterol, and the expression of LDL receptors. LDL receptors in the liver play a major role in determining plasma LDL levels (a low number of receptors is associated with high plasma LDL levels while a high number of hepatic LDL receptors is associated with low plasma LDL levels). The number of LDL receptors is regulated by the cholesterol content of the cell (35). When cellular cholesterol levels are decreased the transcription factor SREBP is transported from the endoplasmic reticulum to the Golgi where proteases cleave and activate SREBP, which then migrates to the nucleus and stimulates the expression of LDL receptors (Figure 4). Conversely, when cellular cholesterol levels are high SREBP remains in the endoplasmic reticulum in an inactive form and the expression of LDL receptors is low. As discussed later PCSK9 regulates the rate of degradation of LDL receptors.

 

Figure 3. LDL Receptor Pathway (figure modified from Annual Review of Biochemistry 46: 897, 1977)

Figure 4. SREBP Pathway (figure modified from Journal of Lipid Research 50: Supp S15, 2009)

 

 

LRP-1 is a member of the LDL receptor family. It is expressed in multiple tissues including the liver. LRP-1 recognizes Apo E and mediates the uptake of chylomicron remnants and IDL (VLDL remnants).

 

VLDL Receptor (37)

 

The VLDL receptor is a member of the LDL receptor family. The VLDLR is expressed in the heart, skeletal muscle, adipose tissue, endothelium, brain, macrophages, and other tissues. Interestingly it is not usually expressed in the liver but hepatic expression can be induced by endoplasmic reticulum stress and PPAR alpha activation. Apo E but not Apo B bind to the VLDL receptor thereby allowing for the uptake of triglyceride rich lipoprotein particles (VLDL and chylomicrons).

 

Class B Scavenger Receptor B1 (SR-B1) (38)

 

SR-B1 is expressed in the liver, adrenal glands, ovaries, testes, macrophages, and other cells. In the liver and steroid producing cells, it mediates the selective uptake of cholesterol esters from HDL particles. In macrophages and other cells, it facilitates the efflux of cholesterol from the cell to HDL particles.

 

ATP-Binding Cassette Transporter A1 (ABCA1) (39)

 

ABCA1 is expressed in many cells including hepatocytes, enterocytes, and macrophages. It mediates the transport of cholesterol and phospholipids from the cell to lipid poor HDL particles (pre-beta-HDL).

 

ATP-Binding Cassette Transporter G1 (ABCG1) (40)

 

ABCG1 is expressed in many different cell types and mediates the efflux of cholesterol from the cell to HDL particles.

 

ATP-Binding Cassette Transporter G5 and G8 (ABCG5/ABCG8) (41,42)

 

ABCG5 and ABCG8 are expressed in the liver and intestine and form a heterodimer. In the intestine, these transporters mediate the movement of plant sterols and cholesterol from inside the enterocyte into the intestinal lumen thereby decreasing the absorption of cholesterol and limiting the uptake of dietary plant sterols. In the liver, these transporters play a role in the movement of cholesterol and plant sterols into the bile facilitating the excretion of sterols.

 

Niemann-Pick C1-Like 1 (NPC1L1) (41)

 

NPC1L1 is expressed in the intestine and mediates the uptake of cholesterol and plant sterols from the intestinal lumen into the enterocyte. NPC1L1 is also expressed in the liver where it mediates the movement of cholesterol from hepatocytes into the bile.

 

ENZYMES AND TRANSFER PROTEINS INVOLVED IN LIPOPROTEIN METABOLISM

 

There are several enzymes and transfer proteins that play a key role in lipoprotein metabolism.

 

Lipoprotein Lipase (LPL) (43)

 

LPL is synthesized in muscle, heart, and adipose tissue, then secreted and attached to the endothelium of the adjacent blood capillaries. This enzyme hydrolyzes the triglycerides carried in chylomicrons and VLDL to fatty acids, which can be taken up by cells. The catabolism of triglycerides results in the conversion of chylomicrons into chylomicron remnants and VLDL into IDL (VLDL remnants). This enzyme requires Apo C-II as a cofactor. Apo A-V also plays a key role in the activation of this enzyme. In contrast Apo C-III and Apo A-II inhibit the activity of LPL. Insulin stimulates LPL expression and LPL activity is reduced in patients with poorly controlled diabetes, which can impair the metabolism of triglyceride rich lipoproteins leading to hypertriglyceridemia (44).

 

Hepatic Lipase (45)

 

Hepatic lipase is localized to the sinusoidal surface of liver cells. It mediates the hydrolysis of triglycerides and phospholipids in IDL and LDL leading to smaller particles (IDL is converted to LDL; LDL is converted from large LDL to small LDL). It also mediates the hydrolysis of triglycerides and phospholipids in HDL resulting in smaller HDL particles.

 

Endothelial Lipase (46)

 

Endothelial lipase plays a major role in hydrolyzing the phospholipids in HDL.

 

Lecithin: Cholesterol Acyltransferase (LCAT) (47)

 

LCAT is made in the liver. In the plasma, it catalyzes the synthesis of cholesterol esters in HDL by facilitating the transfer of a fatty acid from position 2 of lecithin to cholesterol. This allows for the transfer of the cholesterol from the surface of the HDL particle (free cholesterol) to the core of the HDL particle (cholesterol ester), which facilitates the continued uptake of free cholesterol by HDL particles by reducing the concentration of cholesterol on the surface of HDL.

 

Cholesteryl Ester Transfer Protein (CETP) (48,49)

 

This protein is synthesized in the liver and in the plasma mediates the transfer of cholesterol esters from HDL to VLDL, chylomicrons, and LDL and the transfer of triglycerides from VLDL and chylomicrons to HDL. Inhibition of CETP activity leads to an increase in HDL cholesterol and a decrease in LDL cholesterol.

 

Microsomal Triglyceride Transfer Protein (MTTP) (50)

 

MTTP is expressed primarily in the liver and small intestine and plays a crucial role in the synthesis of lipoproteins in these tissues. MTTP mediates the transfer of triglycerides to apolipoprotein B-100 in the liver to form VLDL and to apolipoprotein B-48 in the intestine to form chylomicrons.

 

EXOGENOUS LIPOPROTEIN PATHWAY (CHYLOMICRONS)

 

Figure 5. Exogenous Lipoprotein Pathway

 

Fat Absorption (51-54)

 

The exogenous lipoprotein pathway starts in the intestine. Dietary triglycerides (approximately 100 grams per day) are hydrolyzed to free fatty acids and monoacylglycerol by intestinal lipases and emulsified with bile acids, cholesterol, plant sterols, and fat-soluble vitamins to form micelles. While the fatty acids in the intestine are overwhelmingly accounted for by dietary intake the cholesterol in the intestinal lumen is primarily derived from bile (approximately 800-1200mg of cholesterol from bile vs. 200-500mg from diet). Plant sterols account for approximately 25% of dietary sterol intake (approximately 100-150mg/day). The cholesterol, plant sterols, fatty acids, monoacylglycerol, and fat-soluble vitamins contained in the micelles are then transported into the intestinal cells. The uptake of cholesterol and plant sterols from the intestinal lumen into intestinal cells is facilitated by a sterol transporter, Niemann-Pick C1- like 1 protein (NPC1L1) (Figure 6). Ezetimibe, a drug which inhibits intestinal cholesterol and plant sterol uptake, binds to NPC1L1 and inhibits its activity. Once in the intestinal cell the cholesterol and plant sterols may be transported back into the intestinal lumen, a process mediated by ABCG5 and ABCG8, or converted to sterol esters by acyl-CoA cholesterol acyl transferase (ACAT), which attaches a fatty acid to the sterol. Compared to cholesterol, plant sterols are poor substrates for ACAT and therefore the formation of plant sterol esters does not occur as efficiently as the formation of cholesterol esters. In humans, <5% of dietary plant sterols are absorbed and the vast majority are transported out of the intestine cell, a process mediated by ABCG5 and ABCG8, which are very efficient at effluxing plant sterols from the intestinal cell into the intestinal lumen. Patients with sitosterolemia have mutations in either ABCG5 or ABCG8 and net absorption of dietary plant sterols is increased (20-30% absorbed vs. < 5% in normal subjects) (55). Thus, ABCG5 and ABCG8 along with ACAT serve as gate keepers and block the uptake of plant sterols and likely also play an important role in determining the efficiency of cholesterol absorption (humans typically absorb only approximately 50% of dietary cholesterol with a range of 25-75%).

 

Figure 6. Intestinal Cell and Sterol Metabolism. C= cholesterol, CE= cholesterol ester.

 

The pathway of absorption of free fatty acids is not well understood but it is likely that both passive diffusion and specific transporters play a role. The fatty acid transporter CD36 is strongly expressed in the proximal third of the intestine and is localized to the villi. While this transporter likely plays a role in fatty acid uptake by intestinal cells, this transporter is not essential as humans and mice deficient in this protein do not have fat malabsorption. However, in mice deficient in CD36 there is a shift in the absorption of lipid to the distal intestine, suggesting pathways that can compensate for the absence of CD36. Fatty acid transport protein 4 (FATP4) is also highly expressed in the intestine. However, mice deficient in FATP4 do not have abnormalities in fat absorption. It is likely that there are multiple pathways for the absorption of fatty acids into intestinal cells. The pathways by which monoacylglycerols are absorbed by intestinal cells remain to be defined.

 

Formation of Chylomicrons (51,54)

 

The absorbed fatty acids and monoacylglycerols are utilized to synthesize triglycerides. The key enzymes required for triglyceride synthesis are monoacylglycerol acyltransferase (MGAT) and diacylglycerol transferase (DGAT). MGAT catalyzes the addition of a fatty acid to monoacylglycerol while DGAT catalyzes the addition of a fatty acid to diacylglycerol resulting in triglyceride formation.  As noted above, the majority of the cholesterol absorbed by the intestine is esterified to cholesterol esters by ACAT. The triglycerides and cholesterol esters are packaged into chylomicrons in the endoplasmic reticulum. The size and composition of the chylomicrons formed in the intestine are dependent on the amount of fat ingested and absorbed by the intestine and the type of fat absorbed. Increased fat absorption results in larger chylomicrons. The formation of chylomicrons in the endoplasmic reticulum requires the synthesis of Apo B-48 by the intestinal cell (Figure 6).  Microsomal triglyceride transfer protein (MTTP) is required for the movement of lipid from the endoplasmic reticulum to the Apo B-48. The absence of MTTP results in the inability to form chylomicrons (Abetalipoproteinemia) (56). Lomitapide inhibits MTTP function and is used to treat patients with homozygous Familial Hypercholesterolemia (57).

 

Chylomicron Metabolism (26,31,43,58-62)

 

Chylomicrons are secreted into the lymph and delivered via the thoracic duct to the circulation. It should be noted that this results in the newly formed chylomicrons being delivered to the systemic circulation and not delivered directly to the liver via the portal circulation. This facilitates the delivery of the nutrients contained in the chylomicrons to muscle and adipose tissue. In muscle and adipose tissue lipoprotein lipase (LPL) is expressed at high levels. LPL is synthesized in muscle and adipocytes and transported to the luminal surface of capillaries. Lipase maturation factor 1 plays a key role in the stabilization and movement of LPL from muscle cells and adipocytes to the capillary endothelial cell surface. Glycosylphosphatidylinositol anchored high density lipoprotein binding protein 1 (GPIHBP1) binds LPL and transports it to the capillary lumen and anchors LPL to the capillary endothelium. Activation of LPL by Apo C-II, carried on the chylomicrons, leads to the hydrolysis of the triglycerides that are carried in the chylomicrons resulting in the formation of free fatty acids, which can be taken up by the adjacent muscle cells and adipocytes for either energy production or storage.  Fatty acid transport proteins (FATPs) and CD36 facilitate the uptake of fatty acids into adipocytes and muscle cells. Some of the free fatty acids released from chylomicrons bind to albumin and can be transported to other tissues. Apo A-V also plays an important role in activating LPL activity. Loss of function mutations in LPL, Apo C-II, GPIHPB1, lipase maturation factor 1, and Apo A-V can result in marked hypertriglyceridemia (familial chylomicronemia syndrome) (30). In addition, there are proteins that inhibit LPL activity. Apo C-III inhibits LPL activity and loss of function mutations in this gene are associated with increases in LPL activity and decreases in plasma triglyceride levels. Similarly, angiopoietin like protein 3 and 4, which target LPL for inactivation, regulate LPL activity. Loss of function mutations in these proteins also are associated with decreases in plasma triglyceride levels. Finally, the expression of LPL by muscle cells and adipocytes is regulated by hormones (particularly insulin), nutritional status, and inflammation.

 

The metabolism of the triglycerides carried in the chylomicrons results in a marked decrease in the size of these particles leading to the formation of chylomicron remnants, which are enriched in cholesterol esters and acquire Apo E. As these particles decrease in size phospholipids and apolipoproteins (Apo A and C) on the surface of the chylomicrons are transferred to other lipoproteins, mainly HDL. The transfer of Apo C-II from chylomicrons to HDL decreases the ability of LPL to further breakdown triglycerides. These chylomicron remnants are cleared from the circulation by the liver. The Apo E on the chylomicron remnants binds to the LDL receptor and other hepatic receptors such as LRP and syndecan-4 and the entire particle is taken up by the hepatocytes. Apo E is crucial for this process and mutations in Apo E (for example homozygosity for the Apo E2 isoform) can result in decreased chylomicron clearance and elevations in plasma cholesterol and triglyceride levels (familial dysbetalipoproteinemia) (30).

 

The exogenous lipoprotein pathway results in the efficient transfer of dietary fatty acids to muscle and adipose tissue for energy utilization and storage. The cholesterol is delivered to the liver where it can be utilized for the formation of VLDL, bile acids, or secreted back to the intestine via secretion into the bile. In normal individuals, this pathway can handle large amounts of fat (100 grams or more per day) without resulting in marked increases in plasma triglyceride levels. In fact, in a normal individual, a meal containing 75 grams of fat results in only a very modest increase in postprandial triglyceride levels. 

 

ENDOGENOUS LIPOPROTEIN PATHWAY (VLDL AND LDL)

 

Figure 7. Endogenous Lipoprotein Pathway

 

Formation of VLDL (50,63,64

 

In the liver triglycerides and cholesterol esters are transferred in the endoplasmic reticulum to newly synthesized Apo B-100. Similar to the intestine this transfer is mediated by MTTP. The availability of triglycerides is the primary determinant of the rate of VLDL synthesis. If the supply of triglyceride is limited the newly synthesized Apo B is rapidly degraded. Thus, in contrast to many proteins the rate of synthesis of the Apo B-100 is not the major determinant of the rate of secretion. Rather the amount of lipid available determines whether Apo B-100 is degraded or secreted. MTTP is required for the early addition of lipid to Apo B-100 particles but additional lipid is added via pathways that do not require MTTP. Additionally, the size of the VLDL particles is determined by the availability of triglycerides. When triglycerides are abundant the VLDL particles are large.

 

The quantity of fatty acids available for the synthesis of triglycerides is the main determinant of   triglyceride synthesis in the liver. The major sources of fatty acids are a) de novo fatty acid synthesis, b) the hepatic uptake of triglyceride rich lipoproteins, and c) the flux of fatty acids from adipose tissue to the liver. Diabetes, obesity, and the metabolic syndrome are common causes of an increase in hepatic triglyceride levels and the increased secretion of VLDL (44,65).

 

Loss of function mutations in either Apo B-100 or MTTP result in the failure to produce VLDL and marked decreases in plasma triglyceride and cholesterol levels (familial hypobetalipoproteinemia or abetalipoproteinemia) (56). The precise pathway by which the newly synthesized VLDL particles are secreted from the hepatocyte into the circulation is not resolved.

 

VLDL Metabolism (6,58)

 

VLDL particles are transported to peripheral tissues where the triglycerides are hydrolyzed by LPL and fatty acids are released. This process is very similar to that described above for chylomicrons and there is competition between the metabolism of chylomicrons and VLDL. High levels of chylomicrons can inhibit the clearance of VLDL. The removal of triglycerides from VLDL results in the formation of VLDL remnants (Intermediate density lipoproteins (IDL)). These IDL particles are relatively enriched in cholesterol esters and acquire Apo E from HDL particles. In a pathway analogous to the removal of chylomicron remnants these IDL particles can be removed from the circulation by the liver via binding of Apo E to LDL and LRP receptors. However, while the vast majority of chylomicron remnants are rapidly cleared from the circulation by the liver, only a fraction of IDL particles are cleared (approximately 50% but varies). The remaining triglycerides in the IDL particles are hydrolyzed by hepatic lipase leading to a further decrease in triglyceride content and the exchangeable apolipoproteins are transferred from the IDL particles to other lipoproteins leading to the formation of LDL. These LDL particles predominantly contain cholesterol esters and Apo B-100.  Thus, LDL is a product of VLDL metabolism.

 

LDL Metabolism (34,66-69)

 

The levels of plasma LDL are determined by the rate of LDL production and the rate of LDL clearance, both of which are regulated by the number of LDL receptors in the liver. The production rate of LDL from VLDL is partially determined by hepatic LDL receptor activity with a high LDL receptor activity resulting in a decrease in LDL production due to an increase in IDL uptake. Conversely, low LDL receptor activity results in an increase in LDL production formation due to a decrease in IDL uptake. With regards to LDL clearance, approximately 70% of circulating LDL is cleared via hepatocyte LDL receptor mediated endocytosis with the remainder taken up by extrahepatic tissues. An increase in the number of hepatic LDL receptors therefore increases LDL clearance leading to a decrease in plasma LDL levels. Conversely, a decrease in hepatic LDL receptors slows LDL clearance leading to an increase in plasma LDL levels. Thus, the level of hepatic LDL receptors plays a key role in regulating plasma LDL levels. Many of the drugs used to lower plasma LDL levels, such as the statins, ezetimibe, PCSK9 inhibitors, bile acid sequestrants and bempedoic acid lower plasma LDL levels by increasing the number of hepatic LDL receptors (57).

 

The levels of LDL receptors in the liver are mainly regulated by the cholesterol content of the hepatocyte. As cholesterol levels in the cell decrease, inactive sterol regulatory element binding proteins (SREBPs), which are transcription factors that mediate the expression of LDL receptors and key genes involved in cholesterol and fatty acid metabolism, are transported from the endoplasmic reticulum to the Golgi where proteases cleave the SREBPs into active transcription factors (Figure 4). These active SREBPs move to the nucleus where they stimulate the transcription of the LDL receptor and enzymes required for cholesterol synthesis, including HMG-CoA reductase, the rate limiting enzyme in cholesterol synthesis. If cholesterol levels in the cell are high, then the SREBPs remain in the endoplasmic reticulum in an inactive form and do not stimulate LDL receptor synthesis. In addition, cholesterol in the cell is oxidized and oxidized sterols activate LXR, a nuclear hormone receptor that is a transcription factor, which stimulates the transcription of E3 ubiquitin ligase that mediates the ubiquitination and degradation of the low-density lipoprotein receptor (Inducible degrader of the low-density lipoprotein receptor (IDOL)). Thus, the cell can sense the availability of cholesterol and regulate LDL receptor activity. If the cholesterol content of the cell is decreased LDL receptor activity is increased to allow for the increased uptake of cholesterol. Conversely, if the cholesterol content of the cell is increased LDL receptor activity is decreased and the uptake of LDL by the cell is diminished. Statins, ezetimibe, bile acid sequestrants, and bempedoic acid decrease hepatic cholesterol levels thereby increasing LDL receptor levels and decreasing plasma LDL levels (57). Finally, the LDL receptor is targeted for degradation by PCSK9, a secreted protein that binds to the LDL receptor and enhances LDL receptor degradation in the lysosomes. Loss of function mutations in PCSK9 and drugs that inhibit PCSK9 result in increased LDL receptor activity and decreased LDL levels while gain of function mutations in PCSK9 lead to decreased LDL receptor activity and elevations in LDL levels.

 

Thus, the endogenous lipoprotein pathway facilitates the movement of triglycerides synthesized in the liver to muscle and adipose tissue. Additionally, it also provides a pathway for the transport of cholesterol from the liver to peripheral tissues.

 

HDL METABOLISM AND REVERSE CHOLESTEROL TRANSPORT (38-40,47,48,70,71)

 

Figure 8. HDL Metabolism

 

HDL Formation

 

Several steps are required to generate mature HDL particles. The first step involves the synthesis of the main structural protein contained in HDL, Apo A-I. Apo A-I is synthesized predominantly by the liver and intestine. After Apo A-I is secreted, it acquires cholesterol and phospholipids that are effluxed from hepatocytes and enterocytes. The efflux of cholesterol and phospholipids to the newly synthesized lipid poor Apo A-I (pre-beta HDL) is facilitated by ABCA1. Patients with loss of function mutations in ABCA1 (Tangiers disease) fail to lipidate the newly secreted Apo A-I leading to the rapid catabolism of Apo A-I and very low HDL levels (72). Using mice with targeted knock-out of ABCA1 it has been shown that HDL cholesterol levels are reduced by 80% in mice lacking ABCA1 in the liver and 30% in mice lacking ABCA1 in the intestine. While initially cholesterol and phospholipids are obtained from the liver and intestine, HDL also acquires lipid from other tissues and from other lipoproteins. Muscle cells, adipocytes, and other tissues express ABCA1 and ABCG1 and are able to transfer cholesterol and phospholipids to Apo A-I particles. Additionally, as noted above, newly formed HDL can also obtain cholesterol and phospholipids from chylomicrons and VLDL during their lipolysis by LPL. This accounts for the observation that patients with high plasma triglyceride levels due to decreased clearance frequently have low HDL cholesterol levels. Additionally, phospholipid transfer protein (PLTP) facilitates the movement of phospholipids between lipoproteins; mice lacking PLTP have a marked reduction in HDL cholesterol and Apo A-I levels. Finally, the lipolysis of triglyceride rich lipoproteins also results in the transfer of apolipoproteins from these particles to HDL.

 

HDL Cholesterol Esterification

 

As noted earlier the cholesterol in the core of HDL is esterified (cholesterol esters). The cholesterol that is effluxed from cells to HDL is free cholesterol and is localized on the surface of HDL particles. In order to form mature large spherical HDL particles with a core of cholesterol esters the free cholesterol transferred from cells to the surface of HDL particles must be esterified. LCAT, an HDL associated enzyme catalyzes the transfer of a fatty acid from phospholipids to free cholesterol resulting in the formation of cholesterol esters. The cholesterol ester formed is then able to move from the surface of the HDL particle to the core allowing additional free cholesterol to be transferred from cells to HDL particles. Apo A-I is an activator of LCAT and facilitates this esterification process. LCAT activity is required for the formation of large HDL particles. LCAT deficiency in humans results in decreased HDL cholesterol and Apo A-I levels and a higher percentage of small HDL particles (72).

 

HDL Metabolism

 

Lipases and transfer proteins play an important role in determining the size and composition of HDL particles. The cholesterol ester carried in the core of HDL particles may be transferred to Apo B containing particles in exchange for triglyceride. This transfer is mediated by CETP and results in HDL enriched in triglycerides which may then be metabolized by lipases. Humans deficient in CETP activity have very high HDL cholesterol levels and large HDL particles (72). CETP also impacts LDL cholesterol levels and the absence of CETP results in a decrease in LDL cholesterol. Mice do not have CETP and have relatively high HDL cholesterol levels and low LDL cholesterol levels. Hepatic lipase hydrolyzes both triglycerides and phospholipids in HDL. The triglycerides that are transferred to HDL by CETP activity are catabolized by hepatic lipase resulting in the formation of small HDL particles and Apo A-I more easily disassociates from small HDL resulting in the release of Apo A-I and increased Apo A-I degradation. Genetic deficiency of hepatic lipase results in a modest elevation in HDL cholesterol levels and larger HDL particles (72). Hepatic lipase activity is increased in insulin resistant states and this is associated with reduced HDL cholesterol levels. Endothelial cell lipase is a phospholipase that hydrolyzes the phospholipids carried in HDL particles. In mice increased endothelial lipase activity results in decreased HDL cholesterol levels while decreased endothelial lipase activity increases HDL cholesterol levels.

 

The cholesterol carried on HDL is primarily delivered to the liver. The uptake of HDL cholesterol by the liver is mediated by SR-BI, which promotes the selective uptake of HDL cholesterol. The HDL particle binds to SR-BI and the cholesterol in HDL is transported into the liver without internalization of the HDL particle. A smaller cholesterol depleted HDL particle is formed, which is then released back into the circulation. In SR-BI deficient mice there is a marked increase in HDL cholesterol levels. Interestingly the risk of atherosclerosis is increased in these SR-BI deficient mice despite an increase in HDL cholesterol levels. Notably, while HDL cholesterol levels are increased in SR-B1 deficient mice the reverse cholesterol transport pathway is actually reduced. While in mice the physiological importance of the hepatic SR-BI pathway is clear, the role in humans is uncertain. In mice, the movement of cholesterol from peripheral tissues to the liver is dependent solely on SR-BI while in humans CETP can facilitate the transport of cholesterol from HDL to Apo B containing lipoproteins, which serves as an alternative pathway for the transport cholesterol to the liver. 

 

Apo A-I is metabolized independently of HDL cholesterol. Most of the Apo A-I is catabolized by the kidneys with the remainder catabolized by the liver. Lipid free or lipid poor Apo A-I is filtered by the kidneys and then taken up by the renal tubules. The size of the Apo A-I particle determines whether it can be filtered by the kidneys and hence the degree of lipidation of Apo A-I determines the rate of catabolism. Conditions or disease states (for example Tangiers disease, which is due to a mutation in ABCA1, or LCAT deficiency) that result in lipid poor HDL led to the accelerated catabolism of Apo A-I by the kidney. Apo A-I binds to cubilin, which in conjunction with megalin, a member of the LDL receptor gene family, leads to the uptake and degradation of filtered Apo A-I by renal tubular cells. While the liver is also involved in the catabolism of Apo A-I, the mechanisms are poorly understood. HDL particles may contain Apo E and it is therefore possible that Apo E containing HDL particles are taken up via the LDL receptor and other Apo E receptors in the liver and degraded.

 

Reverse Cholesterol Transport (73-78)

 

Peripheral cells accumulate cholesterol through the uptake of circulating lipoproteins and de novo cholesterol synthesis. Most cells do not have a mechanism for catabolizing cholesterol. Cells that synthesize steroid hormones can convert cholesterol to glucocorticoids, estrogen, testosterone, etc. Intestinal cells, sebocytes, and keratinocytes can secrete cholesterol into the intestinal lumen or onto the skin surface thereby eliminating cholesterol. However, in order for most cells to decrease their cholesterol content reverse cholesterol transport is required. From a clinical point of view, the ability of macrophages in the arterial wall to efficiently efflux cholesterol into the reverse cholesterol transport pathway may play an important role in the prevention of atherosclerosis.

 

As noted earlier ABCA1 plays an important role in the efflux of cholesterol to lipid poor pre-beta Apo A-I particles (Figure 9). ABCG1 plays an important role in the efflux of cholesterol from cells to mature HDL particles. In some studies, SR-B1 also plays a role in the efflux of cholesterol to mature HDL particles. Additionally, passive diffusion of cholesterol from the plasma membrane to HDL may also contribute to cholesterol efflux. The levels of both ABCA1 and ABCG1 are increased by LXR activation. LXR is a nuclear hormone transcription factor that is activated by oxysterols. As the cholesterol levels in a cell increase the formation of oxysterols increases leading to the activation of LXR resulting in an increase in ABCA1 and ABCG1 expression, which will result in the enhanced efflux of cholesterol from the cell to HDL.  Additionally, ABCA1 and ABCG1 mRNAs are targeted for degradation by miR-33, a microRNA that is embedded within the SREBP2 gene. An increase in cellular cholesterol decreases the expression of SREBP2 leading to a decrease in miR-33 resulting in enhanced LXR expression. Thus, the decrease in SREBP2 transcription will lead to a decrease in LDL receptor activity and a reduction in cholesterol uptake, while simultaneously, a decrease in miR-33 will lead to an increase in LXR activity stimulating the expression of ABCA1 and ABCG1 resulting in increased cholesterol efflux. Conversely a decrease in cellular cholesterol levels will increase SREBP2 expression resulting in an increase in LDL receptor activity and an increase in miR-33, which will result in a decrease in LXR activity, decreased expression of ABCA1 and ABCG1, and a reduction in cholesterol efflux. Together changes in cholesterol uptake mediated by the LDL receptor and cholesterol efflux mediated by ABCA1 and ABCG1 will maintain cellular cholesterol homeostasis.

 

Figure 9. Cholesterol Efflux from Macrophages (modified from J. Clinical Investigation 116: 3090, 2006)

 

Once cholesterol is transferred from cells to HDL there are two pathways for the cholesterol to be transported and taken up by the liver. As discussed earlier, HDL can interact with hepatic SR-BI receptors resulting in the selective uptake of cholesterol from HDL particles. Alternatively, CETP can transfer cholesterol from HDL particles to Apo B containing particles with the subsequent uptake of Apo B containing lipoproteins by the liver. After the delivery of cholesterol to the liver there are several pathways by which the cholesterol can be eliminated. Cholesterol can be converted to bile acids and secreted in the bile. Alternatively, cholesterol can be directly secreted into the bile. ABCG5 and ABCG8 promote the transport of cholesterol into the bile and the expression of these genes is enhanced by LXR activation. Thus, an increase in hepatic cholesterol levels leading to increased oxysterol production will activate LXR resulting in the increased expression of ABCG5 and ABCG8 facilitating the secretion of cholesterol in the bile.

 

Evidence suggests that reverse cholesterol transport plays an important role in protecting from the development of atherosclerosis. It should be noted that HDL cholesterol levels may not be indicative of the rate of reverse cholesterol transport. As described above reverse cholesterol transport involves several steps and the level of HDL cholesterol may not accurately reflect these steps. For example, studies have shown that the ability of HDL to promote cholesterol efflux from macrophages can vary. Thus, the same level of HDL cholesterol may not have equivalent abilities to mediate the initial step of reverse cholesterol transport.  

 

LIPOPROTEIN (a) (14-16,79)

 

Figure 10. Lp (a)

 

Lp (a) consists of an LDL molecule and a unique apolipoprotein (a), which is attached to the Apo B-100 of the LDL via a single disulfide bound. Lp (a) contain Apo (a) and Apo B-100 in a 1:1 molar ratio. Like Apo B-100, apo (a) is also made by hepatocytes. Apo (a) contains multiple kringle motifs that are similar to the kringle repeats in plasminogen. The number of kringle repeats can vary and thus the molecular weight of apo (a) can range from 250,000 to 800,000.  The levels of Lp (a) in plasma can vary more than a 1000-fold ranging from undetectable to greater than 100mg/dl. Lp (a) levels largely reflect Lp (a) production rates, which are primarily genetically regulated and not greatly affected by environmental factors. Individuals with high molecular weight Apo (a) proteins tend to have lower levels of Lp (a) while individuals with low molecular weight Apo (a) tend to have higher levels. It is hypothesized that the liver is less efficient in secreting high molecular weight Apo (a). The mechanism of Lp (a) clearance is uncertain but does not appear to primarily involve LDL receptors. Therapies that accelerate LDL clearance and lower LDL levels do not lower Lp (a) levels (for example statin therapy). The kidney appears to play an important role in Lp (a) clearance as kidney disease is associated with delayed clearance and elevations in Lp (a) levels.

 

 Elevated plasma Lp(a) levels are associated with an increased risk of atherosclerosis. Apo (a) is an inhibitor of fibrinolysis and enhances the uptake of lipoproteins by macrophages, both of which could account for the increased the risk of atherosclerosis in individuals with elevated Apo (a) levels. Additionally, Lp (a) is the major lipoprotein carrier of oxidized phospholipids, which are inflammatory and could also increase the risk of atherosclerosis. The physiologic function of Apo (a) is unknown. Apo (a) is found in primates but not in other species.

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education.

 

REFERENCES

 

  1. Feingold KR, Grunfeld C. Lipids: a key player in the battle between the host and microorganisms. J Lipid Res 2012; 53:2487-2489
  2. Nielsen LB, Nielsen MJ, Moestrup SK. Lipid metabolism: an apolipoprotein-derived weapon combating trypanosoma infection. Curr Opin Lipidol 2006; 17:699-701
  3. Feingold KR. The bidirectional link between HDL and COVID-19 infections. J Lipid Res 2021; 62:100067
  4. Smith LC, Pownall HJ, Gotto AM, Jr. The plasma lipoproteins: structure and metabolism. Annu Rev Biochem 1978; 47:751-757
  5. Julve J, Martin-Campos JM, Escola-Gil JC, Blanco-Vaca F. Chylomicrons: Advances in biology, pathology, laboratory testing, and therapeutics. Clin Chim Acta 2016; 455:134-148
  6. Chait A, Ginsberg HN, Vaisar T, Heinecke JW, Goldberg IJ, Bornfeldt KE. Remnants of the Triglyceride-Rich Lipoproteins, Diabetes, and Cardiovascular Disease. Diabetes 2020; 69:508-516
  7. Krauss RM, King SM. Remnant lipoprotein particles and cardiovascular disease risk. Best Pract Res Clin Endocrinol Metab 2023; 37:101682
  8. Berneis KK, Krauss RM. Metabolic origins and clinical significance of LDL heterogeneity. J Lipid Res 2002; 43:1363-1379
  9. Asztalos BF, Niisuke K, Horvath KV. High-density lipoprotein: our elusive friend. Curr Opin Lipidol 2019; 30:314-319
  10. Thakkar H, Vincent V, Sen A, Singh A, Roy A. Changing Perspectives on HDL: From Simple Quantity Measurements to Functional Quality Assessment. J Lipids 2021; 2021:5585521
  11. Thomas SR, Zhang Y, Rye KA. The pleiotropic effects of high-density lipoproteins and apolipoprotein A-I. Best Pract Res Clin Endocrinol Metab 2023; 37:101689
  12. Vaisar T, Pennathur S, Green PS, Gharib SA, Hoofnagle AN, Cheung MC, Byun J, Vuletic S, Kassim S, Singh P, Chea H, Knopp RH, Brunzell J, Geary R, Chait A, Zhao XQ, Elkon K, Marcovina S, Ridker P, Oram JF, Heinecke JW. Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of HDL. J Clin Invest 2007; 117:746-756
  13. Kostner KM, Kostner GM. Lipoprotein (a): a historical appraisal. J Lipid Res 2017; 58:1-14
  14. Nordestgaard BG, Langsted A. Lipoprotein (a) as a cause of cardiovascular disease: insights from epidemiology, genetics, and biology. J Lipid Res 2016; 57:1953-1975
  15. Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016; 57:1339-1359
  16. Khovidhunkit W. Lipoprotein(a). In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  17. Mahley RW, Innerarity TL, Rall SC, Jr., Weisgraber KH. Plasma lipoproteins: apolipoprotein structure and function. J Lipid Res 1984; 25:1277-1294
  18. Breslow JL. Human apolipoprotein molecular biology and genetic variation. Annu Rev Biochem 1985; 54:699-727
  19. Frank PG, Marcel YL. Apolipoprotein A-I: structure-function relationships. J Lipid Res 2000; 41:853-872
  20. Chan DC, Ng TW, Watts GF. Apolipoprotein A-II: evaluating its significance in dyslipidaemia, insulin resistance, and atherosclerosis. Ann Med 2012; 44:313-324
  21. Wang F, Kohan AB, Lo CM, Liu M, Howles P, Tso P. Apolipoprotein A-IV: a protein intimately involved in metabolism. J Lipid Res 2015; 56:1403-1418
  22. Hubacek JA. Apolipoprotein A5 fifteen years anniversary: Lessons from genetic epidemiology. Gene 2016; 592:193-199
  23. Sharma V, Forte TM, Ryan RO. Influence of apolipoprotein A-V on the metabolic fate of triacylglycerol. Curr Opin Lipidol 2013; 24:153-159
  24. Anant S, Davidson NO. Molecular mechanisms of apolipoprotein B mRNA editing. Curr Opin Lipidol 2001; 12:159-165
  25. Levenson AE, de Ferranti SD. Familial Hypercholesterolemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  26. Wolska A, Dunbar RL, Freeman LA, Ueda M, Amar MJ, Sviridov DO, Remaley AT. Apolipoprotein C-II: New findings related to genetics, biochemistry, and role in triglyceride metabolism. Atherosclerosis 2017; 267:49-60
  27. Ramms B, Gordts P. Apolipoprotein C-III in triglyceride-rich lipoprotein metabolism. Curr Opin Lipidol 2018; 29:171-179
  28. D'Erasmo L, Di Costanzo A, Gallo A, Bruckert E, Arca M. ApoCIII: A multifaceted protein in cardiometabolic disease. Metabolism 2020; 113:154395
  29. Wolska A, Reimund M, Remaley AT. Apolipoprotein C-II: the re-emergence of a forgotten factor. Curr Opin Lipidol 2020; 31:147-153
  30. Patni N, Ahmad Z, Wilson DP. Genetics and Dyslipidemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  31. Taskinen MR, Boren J. Why Is Apolipoprotein CIII Emerging as a Novel Therapeutic Target to Reduce the Burden of Cardiovascular Disease? Curr Atheroscler Rep 2016; 18:59
  32. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, Yang Q, Hughes SG, Geary RS, Arca M, Stroes ESG, Bergeron J, Soran H, Civeira F, Hemphill L, Tsimikas S, Blom DJ, O'Dea L, Bruckert E. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N Engl J Med 2019; 381:531-542
  33. Mahley RW. Apolipoprotein E: from cardiovascular disease to neurodegenerative disorders. J Mol Med (Berl) 2016; 94:739-746
  34. Goldstein JL, Brown MS. The LDL receptor. Arterioscler Thromb Vasc Biol 2009; 29:431-438
  35. Goldstein JL, DeBose-Boyd RA, Brown MS. Protein sensors for membrane sterols. Cell 2006; 124:35-46
  36. van de Sluis B, Wijers M, Herz J. News on the molecular regulation and function of hepatic low-density lipoprotein receptor and LDLR-related protein 1. Curr Opin Lipidol 2017; 28:241-247
  37. Krauss RM, Lu JT, Higgins JJ, Clary CM, Tabibiazar R. VLDL receptor gene therapy for reducing atherogenic lipoproteins. Mol Metab 2023; 69:101685
  38. Trigatti BL. SR-B1 and PDZK1: partners in HDL regulation. Curr Opin Lipidol 2017; 28:201-208
  39. Wang S, Smith JD. ABCA1 and nascent HDL biogenesis. Biofactors 2014; 40:547-554
  40. Baldan A, Tarr P, Lee R, Edwards PA. ATP-binding cassette transporter G1 and lipid homeostasis. Curr Opin Lipidol 2006; 17:227-232
  41. Kidambi S, Patel SB. Cholesterol and non-cholesterol sterol transporters: ABCG5, ABCG8 and NPC1L1: a review. Xenobiotica 2008; 38:1119-1139
  42. Patel SB, Graf GA, Temel RE. ABCG5 and ABCG8: more than a defense against xenosterols. J Lipid Res 2018; 59:1103-1113
  43. Olivecrona G. Role of lipoprotein lipase in lipid metabolism. Curr Opin Lipidol 2016; 27:233-241
  44. Feingold KR. Dyslipidemia in Patients with Diabetes. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  45. Kobayashi J, Miyashita K, Nakajima K, Mabuchi H. Hepatic Lipase: a Comprehensive View of its Role on Plasma Lipid and Lipoprotein Metabolism. J Atheroscler Thromb 2015; 22:1001-1011
  46. Yasuda T, Ishida T, Rader DJ. Update on the role of endothelial lipase in high-density lipoprotein metabolism, reverse cholesterol transport, and atherosclerosis. Circ J 2010; 74:2263-2270
  47. Ossoli A, Simonelli S, Vitali C, Franceschini G, Calabresi L. Role of LCAT in Atherosclerosis. J Atheroscler Thromb 2016; 23:119-127
  48. Mabuchi H, Nohara A, Inazu A. Cholesteryl ester transfer protein (CETP) deficiency and CETP inhibitors. Mol Cells 2014; 37:777-784
  49. Shrestha S, Wu BJ, Guiney L, Barter PJ, Rye KA. Cholesteryl ester transfer protein and its inhibitors. J Lipid Res 2018; 59:772-783
  50. Hooper AJ, Burnett JR, Watts GF. Contemporary aspects of the biology and therapeutic regulation of the microsomal triglyceride transfer protein. Circ Res 2015; 116:193-205
  51. Abumrad NA, Davidson NO. Role of the gut in lipid homeostasis. Physiol Rev 2012; 92:1061-1085
  52. D'Aquila T, Hung YH, Carreiro A, Buhman KK. Recent discoveries on absorption of dietary fat: Presence, synthesis, and metabolism of cytoplasmic lipid droplets within enterocytes. Biochim Biophys Acta 2016; 1861:730-747
  53. Hussain MM. Intestinal lipid absorption and lipoprotein formation. Curr Opin Lipidol 2014; 25:200-206
  54. Kindel T, Lee DM, Tso P. The mechanism of the formation and secretion of chylomicrons. Atheroscler Suppl 2010; 11:11-16
  55. Liebeskind A, Peterson AL, Wilson D. Sitosterolemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  56. Shapiro MD, Feingold KR. Monogenic Disorders Causing Hypobetalipoproteinemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  57. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  58. Dallinga-Thie GM, Franssen R, Mooij HL, Visser ME, Hassing HC, Peelman F, Kastelein JJ, Peterfy M, Nieuwdorp M. The metabolism of triglyceride-rich lipoproteins revisited: new players, new insight. Atherosclerosis 2010; 211:1-8
  59. Dijk W, Kersten S. Regulation of lipid metabolism by angiopoietin-like proteins. Curr Opin Lipidol 2016; 27:249-256
  60. Fong LG, Young SG, Beigneux AP, Bensadoun A, Oberer M, Jiang H, Ploug M. GPIHBP1 and Plasma Triglyceride Metabolism. Trends Endocrinol Metab 2016; 27:455-469
  61. Peterfy M. Lipase maturation factor 1: a lipase chaperone involved in lipid metabolism. Biochim Biophys Acta 2012; 1821:790-794
  62. Young SG, Fong LG, Beigneux AP, Allan CM, He C, Jiang H, Nakajima K, Meiyappan M, Birrane G, Ploug M. GPIHBP1 and Lipoprotein Lipase, Partners in Plasma Triglyceride Metabolism. Cell Metab 2019; 30:51-65
  63. Tiwari S, Siddiqi SA. Intracellular trafficking and secretion of VLDL. Arterioscler Thromb Vasc Biol 2012; 32:1079-1086
  64. Choi SH, Ginsberg HN. Increased very low density lipoprotein (VLDL) secretion, hepatic steatosis, and insulin resistance. Trends Endocrinol Metab 2011; 22:353-363
  65. Feingold KR. Obesity and Dyslipidemia. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  66. Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 2015; 161:161-172
  67. Horton JD, Cohen JC, Hobbs HH. Molecular biology of PCSK9: its role in LDL metabolism. Trends Biochem Sci 2007; 32:71-77
  68. Zhang L, Reue K, Fong LG, Young SG, Tontonoz P. Feedback regulation of cholesterol uptake by the LXR-IDOL-LDLR axis. Arterioscler Thromb Vasc Biol 2012; 32:2541-2546
  69. Brown MS, Radhakrishnan A, Goldstein JL. Retrospective on Cholesterol Homeostasis: The Central Role of Scap. Annual review of biochemistry 2017;
  70. Rosenson RS, Brewer HB, Jr., Davidson WS, Fayad ZA, Fuster V, Goldstein J, Hellerstein M, Jiang XC, Phillips MC, Rader DJ, Remaley AT, Rothblat GH, Tall AR, Yvan-Charvet L. Cholesterol efflux and atheroprotection: advancing the concept of reverse cholesterol transport. Circulation 2012; 125:1905-1919
  71. Rye KA, Barter PJ. Cardioprotective functions of HDLs. J Lipid Res 2014; 55:168-179
  72. Shapiro MD, Feingold KR. Monogenic Disorders Altering HDL Levels. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, de Herder WW, Dhatariya K, Dungan K, Hofland J, Kalra S, Kaltsas G, Kapoor N, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, Levy M, McGee EA, McLachlan R, New M, Purnell J, Sahay R, Shah AS, Singer F, Sperling MA, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  73. Zhao Y, Van Berkel TJ, Van Eck M. Relative roles of various efflux pathways in net cholesterol efflux from macrophage foam cells in atherosclerotic lesions. Curr Opin Lipidol 2010; 21:441-453
  74. Lee-Rueckert M, Escola-Gil JC, Kovanen PT. HDL functionality in reverse cholesterol transport--Challenges in translating data emerging from mouse models to human disease. Biochim Biophys Acta 2016; 1861:566-583
  75. Tall AR. Cholesterol efflux pathways and other potential mechanisms involved in the athero-protective effect of high density lipoproteins. J Intern Med 2008; 263:256-273
  76. Siddiqi HK, Kiss D, Rader D. HDL-cholesterol and cardiovascular disease: rethinking our approach. Curr Opin Cardiol 2015; 30:536-542
  77. Moore KJ, Rayner KJ, Suarez Y, Fernandez-Hernando C. The role of microRNAs in cholesterol efflux and hepatic lipid metabolism. Annu Rev Nutr 2011; 31:49-63
  78. Ouimet M, Barrett TJ, Fisher EA. HDL and Reverse Cholesterol Transport. Circ Res 2019; 124:1505-1518
  79. Hoover-Plow J, Huang M. Lipoprotein(a) metabolism: potential sites for therapeutic targets. Metabolism 2013; 62:479-491

Monogenic Disorders Causing Hypobetalipoproteinemia

ABSTRACT

 

Monogenic mutations leading to hypobetalipoproteinemia are rare. The monogenic causes of hypobetalipoproteinemia include familial hypobetalipoproteinemia, abetalipoproteinemia, chylomicron retention disease, loss of function mutations in PCSK9, and loss of function mutations in angiopoietin-like protein 3 (ANGPTL3) (Familiar Combined Hypolipidemia). This chapter describes the etiology, pathogenesis, clinical and laboratory findings, and the treatment of these rare monogenic disorders.

 

INTRODUCTION

 

Monogenic mutations leading to hypobetalipoproteinemia are rare. The monogenic causes of hypobetalipoproteinemia include familial hypobetalipoproteinemia (FHBL), abetalipoproteinemia (ABL), chylomicron retention disease (CMRD), loss of function mutations in PCSK9, and loss of function mutations in angiopoietin-like protein 3 (ANGPTL3) (Familial Combined Hypolipidemia, FCH) (1). Increased understanding of the genetic and the molecular underpinnings of these disorders has allowed a focused prioritization of therapeutic targets for drug development. Table 1 summarizes genetic, lipid, and clinical features of the major hypobetalipoproteinemia syndromes and table 2 provides a new classification of these disorders. Of note the parental lipid profile is normal in abetalipoproteinemia and chylomicron retention disease.

 

It should be recognized that secondary, non-familial, forms of hypobetalipoproteinemia occur and include strict vegan diet, malnutrition, malabsorption, hyperthyroidism, malignancy, and chronic liver disease. In addition, hypobetalipoproteinemia can also be due to polymorphisms in multiple genes that together result in hypobetalipoproteinemia (polygenic etiology) (2-4). In a study of 111 patients with LDL-C levels below the fifth percentile 36% had monogenic hypobetalipoproteinemia, 34% had polygenic hypobetalipoproteinemia, and 30% had hypobetalipoproteinemia from an unknown cause (2). In a study of women with an LDL-C ≤1st percentile (≤50 mg/dL) 15.7% carried mutations causing monogenic hypocholesterolemia and 49.6% were genetically predisposed to a low LDL-C on the basis of an extremely low weighted polygenetic risk score (4). Of note individuals with monogenic hypobetalipoproteinemia are more likely to have liver steatosis than individuals without a monogenic disorder (2).

 

Table 1. Characteristics of the Hypobetalipoproteinemia Syndromes

 

Inheritance

Effected gene

Prevalence

Lipids

Clinical features

FHBL

ACD

Truncation mutations in Apo B

1:1000 – 1:3000

Apo B <5th percentile,

LDL-C 20- 50 mg/dL

Hepatic steatosis

Mild elevation of transaminases. Lower prevalence of ASCVD

ABL

 

FHBL

AR

 

AR

MTTP

 

Apo B

<1:1,000,000

Triglycerides < 30 mg/dL,

Cholesterol < 30 mg/dL),

LDL and Apo B undetectable

Hepatic steatosis

Malabsorption, steatorrhea, diarrhea, and failure to thrive.

Deficiency of fat-soluble vitamins.

PCSK9

ACD

Loss of function mutations in PCSK9

 

Heterozygous – mild to moderate reduction in LDL-C

Homozygous – LDL-C ~15 mg/dL

Normal health; significantly lower prevalence of ASCVD

FCH

ACD

Loss of function mutations in ANGPTL3

Very rare

Panhypolipidemia

Normal health; significantly lower prevalence of ASCVD

CMRD

AR

SAR1B

Very rare

LDL-C and HDL-C -decreased by 50%,

Triglycerides - normal

hypocholesterolemia associated with failure to thrive, diarrhea, steatorrhea, and abdominal distension

ACD- autosomal co-dominant; AR- autosomal recessive; FHBL- familial hypobetalipoproteinemia; ABL- abetalipoproteinemia; FCH- Familiar Combined Hypolipidemia; CMRD- chylomicron retention disease, MTTP- microsomal triglyceride transfer protein; ANGPTL3- angiopoietin-like protein 3; ASCVD- atherosclerotic cardiovascular disease.

 

Table 2. Classification of Disorders Causing Familial Hypocholesterolemia

New Name

Common Name

Gene Defect

Class I: Familial hypobetalipoproteinemia due to lipoprotein assembly and secretion defects

FHBL-SD1

Abetalipoproteinemia

Microsomal Triglyceride Transfer Protein

FHBL-SD2

Familial Hypobetalipoproteinemia

Apolipoprotein B

FHBL-SD3

Chylomicron retention disease

SAR1B

Class II: Familial hypobetalipoproteinemia due to enhanced lipoprotein catabolism

FHBL-EC1

Familial Combined Hypolipidemia

ANGPTL3

FHBL-EC2

 

PCSK9

Modified from (5).

 

FAMILIAL HYPOBETALIPOPROTEINEMIA  

 

Familial Hypobetalipoproteinemia (FHBL) is a relatively common autosomal semi-dominant disorder most commonly due to truncation mutations in the gene coding for Apo B (1,6-8). The prevalence of heterozygous FHBL is estimated to be 1 in 700 to 3000 (1). Variants that lead to truncated proteins that are 30% in length or shorter have more severe signs and symptoms than those with longer truncated proteins (6,7). The truncated forms of Apo B found in FHBL are generally non-functional (truncation decreases lipidation and secretion) and are catabolized quickly, resulting in markedly reduced levels in the plasma (Apo B <5th percentile and LDL-C typically between 20- 50 mg/dL) (7,8). Although there is one normal allele in heterozygous FHBL, plasma Apo B levels are approximately 25% of normal rather than the predicted 50% (8). These lower-than-expected levels result from a lower secretion rate of VLDL Apo B from the liver, decreased production of LDL Apo B, increased catabolism of VLDL, and extremely low secretion of the truncated Apo B (6-8). Given the reduced substrate (Apo B) for lipid (predominantly triglyceride) loading, fatty liver develops in these patients (6,9). Hepatic steatosis and mild elevation of liver enzymes are common in heterozygous FHBL (6,9). Interestingly, individuals with monogenic hypobetalipoproteinemia had a much greater prevalence of hepatic dysfunction than individuals with polygenic hypobetalipoproteinemia (2). In contrast to non-alcoholic fatty liver disease, FHBL is not associated with hepatic or peripheral insulin resistance (9). This observation, however, does not imply that hepatic steatosis associated with FHBL is benign. There are several reports of steatohepatitis, cirrhosis, and hepatocellular carcinoma in patients with FHBL and it is estimated that 5-10% of individuals with FHBL develop relatively more severe nonalcoholic steatohepatitis (6). Because of the risk of developing liver disease liver function tests should be checked every 1-2 years and a hepatic ultrasound in those with elevated liver transaminases (6). While hepatic fat accumulation is the rule, there is generally sufficient chylomicron production to handle dietary fat. However, oral fat intolerance and intestinal fat malabsorption have been reported (6). On the positive side the decrease in proatherogenic lipoproteins has been associated with a reduced risk of cardiovascular disease (10).

 

Given the association of FHBL and low LDL-C, Apo B has been an attractive target for drug development. Indeed, unraveling the genetic and molecular mechanisms of FHBL provided the motivation to pharmacologically antagonize Apo B synthesis for therapeutic gains. This culminated in the production of mipomersen, a synthetic single strand anti-sense oligonucleotide to Apo B (11,12). Essentially, anti-sense oligonucleotides contain approximately ~20 deoxyribonucleic acid (DNA) base pairs complementary to a unique messenger ribonucleic acid (mRNA) sequence. The hybridization of the anti-sense oligonucleotide to the mRNA of interest leads to its catabolism via RNase H1, with markedly reduced mRNA levels and ultimately reduced target protein levels. In this case, mipomersen binds to Apo B mRNA leading to reduced production of the protein, and mimicking (albeit to a lesser extent) FHBL. Mipomersen is the first anti-sense oligonucleotide approved by the United States Food and Drug Administration (FDA) and was commercialized in 2013 with a limited indication for adjunctive LDL-C lowering in patients with homozygous familial hypercholesterolemia (HoFH) (12). It is an injectable agent administered subcutaneously once a week. In the clinical trials, mipomersen was associated with a reduction of LDL-C by 21% in subjects with HoFH and 33% in subjects with heterozygous familial hypercholesterolemia (HeFH) (12). Interestingly, it was also found to lower Lp(a) by 21- 23% (12). While it is highly efficacious in LDL-C lowering, it has side effects, many of which can be predicted based on the experience with FHBL (e.g., hepatic steatosis, elevated liver enzymes) (12). It is also associated with injection site reactions in a considerable number of subjects (12). In May 2018 sales were discontinued due to safety concerns related to increased liver transaminases and fatty liver.

 

Homozygous hypobetalipoproteinemia (HHBL) is extremely rare (6). These patients are homozygous or compound heterozygous for mutations in the Apo B gene. The clinical manifestations mimic ABL (see below) (6).

 

ABETALIPOPROTEINEMIA  

 

Abetalipoproteinemia (ABL) is a rare autosomal recessive disorder characterized by very low plasma concentrations of triglyceride and cholesterol (under 30 mg/dL) and undetectable levels of LDL and Apo B (1,7,13,14). The incidence of ABL is < 1 in 1,000,000. HDL-C levels are usually normal or modestly reduced. It is due to mutations in the gene that codes for microsomal triglyceride transfer protein (MTTP) (7,13-15). MTTP lipidates nascent Apo B in the endoplasmic reticulum to produce VLDL and chylomicrons in the liver and small intestine, respectively (15,16). Unlipidated Apo B is targeted for proteasomal degradation leading to the absence of Apo B containing lipoproteins in the plasma (and thus markedly reduced levels of LDL-C and triglycerides) (15,16). Similar to FHBL, VLDL production is inhibited (14). The reduced triglyceride export from the liver leads to hepatic steatosis, which rarely may progress to steatohepatitis, fibrosis, and cirrhosis (1,9,13). Additionally, lack of MTTP facilitated lipidation of chylomicrons in the small intestine results in lipid accumulation in enterocytes with associated malabsorption, steatorrhea, and diarrhea (1,7,13). The malabsorption and diarrhea lead to failure to thrive during infancy (1,7,13). A decrease in dietary fat can reduce the gastrointestinal symptoms. Acanthocytosis may encompass 50% of circulating red blood cells (red blood cells with spiked cell membranes, due to thorny projections) due to alterations in the lipid composition and fluidity of red cell membranes (1,13,14). An additional issue of importance related to ABL is deficiency of fat-soluble vitamins (1,13). Early diagnosis of ABL and homozygous hypobetalipoproteinemia is extremely important as vitamin E deficiency culminates in atypical retinitis pigmentosa, spinocerebellar degeneration with ataxia, vitamin K deficiency can lead to a significant bleeding diathesis, vitamin A deficiency can contribute to eye disorders, and vitamin D deficiency can lead to defects in bone formation (1,13). High dose supplementation with fat soluble vitamins early in life can prevent or delay these devastating complications (Table 3) (7,13). Additional treatment measures include a low-fat diet and supplementation with essential fatty acids (Table 3) (7,13).

 

 Table 3. Dietary Recommendations for Abetalipoproteinemia

Fat calories

Less than 10-15% (<15 g/day) of total daily caloric requirement. Increase as tolerated.

Essential fatty acids

Ensure 2-4% daily caloric intake of EFAs (alpha-linolenic acid/linoleic acid)

Medium chain triglycerides

May prevent or treat malnutrition

Vitamin E

100-300 IU/kg/day

Vitamin A

100-400 IU/kg/day

Vitamin D

800-1200 IU/day

Vitamin K

5-35 mg/week

Derived from (1)

 

Given the very low level of atherogenic lipoproteins and lipids associated with ABL, there was interest in inhibiting MTTP therapeutically. Lomitapide is an oral MTP inhibitor that has been developed over the course of many years (12,17). In early trials, it was tested at a relatively high dose and the side effect profile was prohibitive (nausea, flatulence, and diarrhea). The more recent clinical trial program tested lower doses with drug titration in subjects with Homozygote Familial Hypercholesterolemia (HoFH) (12,17). On an intention to treat basis, LDL-C was decreased by 40% and apolipoprotein B by 39% (12). In patients who were actually taking lomitapide, LDL-C levels were reduced by 50% (12). In addition to decreasing LDL-C levels, non-HDL-C levels were decreased by 50%, Lp(a) by 15%, and triglycerides by 45% (12). Lomitapide received the same limited indication as mipomersen for adjunctive treatment of patients with HoFH (12). Besides the gastrointestinal issues already alluded to, its side effect profile includes hepatic steatosis (12). Its long-term safety has not been established.

 

PROPROTEIN CONVERTASE SUBTILISIN/KEXIN TYPE 9 (PCSK9)

 

Proprotein convertase subtilisin/ kexin type 9 (PCSK9) belongs to the proprotein convertase class of serine proteases (18-20). After synthesis, PCSK9 undergoes autocatalytic cleavage. This step is required for secretion, most likely because the prodomain functions as a chaperone and facilitates folding (18,19). PCSK9 is associated with LDL particles and the LDL-receptor (LDLR) (20). In 2003, Abifadel reported the seminal work that mapped PCSK9 as the third locus for autosomal dominant hypercholesterolemia (Familial Hypercholesterolemia- FH) (21). This finding revealed a previously unknown actor involved in cholesterol homeostasis and served to launch a series of investigations into PCSK9 biology. As it turns out, PCSK9 functions as a central regulator of plasma LDL-C concentration (18-20). It binds to the LDLR and targets it for destruction in the lysosome (18-20). Overactivity of PCSK9 results in a decrease in LDLR and an increase in LDL-C levels while decreased activity of PCSK9 results in an increase in LDLR and a decrease in LDL-C.

 

Since the discovery of gain-of-function mutations in PCSK9 as a cause of FH, investigators have also uncovered loss of function mutations of PCSK9. Loss-of-function mutations in PCSK9 are associated with low LDL-C levels and markedly reduced ASCVD (18,19). In African Americans 2.6 percent had nonsense mutations in PCSK9 that resulted in a 28 percent reduction in LDL-C and an 88 percent reduction in the risk of coronary heart disease (22). The hypolipidemia is not associated with liver abnormalities or other disorders. Interestingly, rare individuals homozygous or compound heterozygotes for loss of function mutations in PCSK9 have been reported with extremely low levels of LDL-C (~15 mg/dL), normal health and reproductive capacity, and no evidence of neurologic or cognitive dysfunction (20,23,24). Collectively, these observations served as further motivation to pursue antagonism of PCSK9 as a therapeutic target. Antagonizing PCSK9 would prolong the lifespan of LDLR, leading to significant reductions in plasma LDL-C levels. Two fully human monoclonal antibodies (alirocumab and evolocumab) targeting PCSK9 became commercially available in 2015 and inclisiran, a small interfering RNA that inhibits translation of PCSK9 is also available. Other approaches to inhibit PCSK9 are under investigation.  

 

FAMILIAL COMBINED HYPOLIPIDEMIA   

 

Familial combined hypolipidemia (FCH) is due to loss of function mutations in the gene encoding angiopoietin-like protein 3 (ANGPTL3) (25,26). ANGPTL3 inhibits various lipases, such as lipoprotein lipase and endothelial lipase (25,26). Therefore, loss of function mutations in ANGPTL3 relinquishes this inhibition increasing the activity of lipases resulting in more efficient metabolism of VLDL and HDL particles (25,26). In addition, to increasing VLDL clearance the secretion of VLDL is also decreased due to a decrease in free fatty acid flux to the liver (25). LDL clearance is increased but the mechanism remains to be fully elucidated (25). Studies have suggested that ANGPTL3 inhibition lowers LDL-C by limiting LDL particle production due to ANGPTL3 inhibition and increased endothelial lipase activity reducing VLDL-lipid content and size, generating remnant particles that are efficiently removed from the circulation rather than being further metabolized to LDL (27).

 

Clinically, FCH manifests as panhypolipidemia (decreased triglycerides, LDL-C, HDL-C, apo B, and apo A-I) (25,26,28). Interestingly, heterozygotes for certain nonsense mutations in the first exon of ANGPTL3 have moderately reduced LDL-C and triglyceride levels while compound heterozygotes have significant reductions in HDL-C as well (25,26).  Homozygosity or compound heterozygosity for other loss-of-function mutations in exon 1 of ANGPTL3 have no detectable ANGPTL3 in plasma and striking reductions of atherogenic lipoproteins with HDL particles containing only apo A-I and preß-HDL. Individuals who are heterozygous for the loss of function mutations in ANGPTL3 have significantly reduced LDL-C and triglyceride levels and a reduced risk of atherosclerosis (25,26,28).

 

A pooled analysis of cases of familial combined hypolipidemia was published 2013 (29). One hundred fifteen individuals carrying 13 different mutations in the ANGPTL3 gene (14 homozygotes, 8 compound heterozygotes, and 93 heterozygotes) and 402 controls were evaluated. Homozygotes and compound heterozygotes (two mutant alleles) had no measurable ANGPTL3 protein. In heterozygotes, ANGPTL3 was reduced by 34-88%, according to genotype. All cases (homozygotes and heterozygotes) demonstrated significantly lower concentrations of all plasma lipoproteins (except for Lp(a)) as compared to controls. Familial combined hypolipidemia is not associated with any comorbidity. In fact, the prevalence of fatty liver was the same as controls. However, ANGPTL3 deficiency is associated with a reduced risk of cardiovascular disease (25,30).

 

Recently, evinacumab, a human monoclonal antibody against ANGPTL3, was approved for the treatment of Homozygous Familial Hypercholesterolemia (12). Evinacumab decreases LDL-C levels by mechanisms independent of LDL receptor activity (12).

 

CHYLOMICRON RETENTION DISEASE

 

Chylomicron retention disease (CMRD), known also as Anderson’s disease for the individual who first described the condition in 1961, is a rare inherited lipid malabsorption syndrome (31,32). It is due to mutations in the SAR1B gene which codes for the protein SAR1b, a small GTPase, involved in intracellular protein trafficking (31). Mutations in SAR1b result in the failure of pre-chylomicrons to move from the endoplasmic reticulum to the golgi (31). This disorder usually presents in young infants with diarrhea, steatorrhea, abdominal distention, and failure to thrive, which can improve with a low-fat diet (1,31,32). Patients with CMRD demonstrate a specific autosomal recessive hypocholesterolemia that differs from other familial hypocholesterolemias. CMRD is associated with a 50% reduction in both plasma LDL-C and HDL-C with normal fasting triglyceride levels (31,32). Mutations in SAR1B do not affect VLDL secretion by the liver. The decrease in HDL-C is postulated to be due to a decrease in Apo A-I secretion and cholesterol efflux by the small intestine (31). The mechanism accounting for the decrease in LDL-C is not clear. The usual increase in triglycerides and chylomicron levels following a fat meal is blocked (31). The duodenal mucosa is white on endoscopy and intestinal biopsy reveals cytosolic lipid droplets and lipoprotein-sized particles in enterocytes (31). As one would expect the absorption of fat-soluble vitamins (A, D, K, and E) and essential fatty acids is impaired (31,32). Neurological and eye manifestations are milder and occur at an older age compared to abetalipoproteinemia (1). Red blood cell acanthosis is rare (1). Heterozygotes with mutations in SAR1B are unaffected.

 

Treatment for individuals with CMRD is similar to that described above for individuals with ABL (32).

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education.

 

REFERENCES

 

  1. Bredefeld C, Hussain MM, Averna M, Black DD, Brin MF, Burnett JR, Charriere S, Cuerq C, Davidson NO, Deckelbaum RJ, Goldberg IJ, Granot E, Hegele RA, Ishibashi S, Karmally W, Levy E, Moulin P, Okazaki H, Poinsot P, Rader DJ, Takahashi M, Tarugi P, Traber MG, Di Filippo M, Peretti N. Guidance for the diagnosis and treatment of hypolipidemia disorders. J Clin Lipidol 2022; 16:797-812
  2. Rimbert A, Vanhoye X, Coulibaly D, Marrec M, Pichelin M, Charriere S, Peretti N, Valero R, Wargny M, Carrie A, Lindenbaum P, Deleuze JF, Genin E, Redon R, Rollat-Farnier PA, Goxe D, Degraef G, Marmontel O, Divry E, Bigot-Corbel E, Moulin P, Cariou B, Di Filippo M. Phenotypic Differences Between Polygenic and Monogenic Hypobetalipoproteinemia. Arterioscler Thromb Vasc Biol 2021; 41:e63-e71
  3. Blanco-Vaca F, Martin-Campos JM, Beteta-Vicente A, Canyelles M, Martinez S, Roig R, Farre N, Julve J, Tondo M. Molecular analysis of APOB, SAR1B, ANGPTL3, and MTTP in patients with primary hypocholesterolemia in a clinical laboratory setting: Evidence supporting polygenicity in mutation-negative patients. Atherosclerosis2019; 283:52-60
  4. Balder JW, Rimbert A, Zhang X, Viel M, Kanninga R, van Dijk F, Lansberg P, Sinke R, Kuivenhoven JA. Genetics, Lifestyle, and Low-Density Lipoprotein Cholesterol in Young and Apparently Healthy Women. Circulation 2018; 137:820-831
  5. Bredefeld C, Peretti N, Hussain MM, Medical Advisory P. New Classification and Management of Abetalipoproteinemia and Related Disorders. Gastroenterology 2021; 160:1912-1916
  6. Burnett JR, Hooper AJ, Hegele RA. APOB-Related Familial Hypobetalipoproteinemia. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A, eds. GeneReviews((R)). Seattle (WA) 2021.
  7. Hooper AJ, Burnett JR. Update on primary hypobetalipoproteinemia. Curr Atheroscler Rep 2014; 16:423
  8. Hooper AJ, van Bockxmeer FM, Burnett JR. Monogenic hypocholesterolaemic lipid disorders and apolipoprotein B metabolism. Crit Rev Clin Lab Sci 2005; 42:515-545
  9. Welty FK. Hypobetalipoproteinemia and abetalipoproteinemia: liver disease and cardiovascular disease. Curr Opin Lipidol 2020; 31:49-55
  10. Peloso GM, Nomura A, Khera AV, Chaffin M, Won HH, Ardissino D, Danesh J, Schunkert H, Wilson JG, Samani N, Erdmann J, McPherson R, Watkins H, Saleheen D, McCarthy S, Teslovich TM, Leader JB, Lester Kirchner H, Marrugat J, Nohara A, Kawashiri MA, Tada H, Dewey FE, Carey DJ, Baras A, Kathiresan S. Rare Protein-Truncating Variants in APOB, Lower Low-Density Lipoprotein Cholesterol, and Protection Against Coronary Heart Disease. Circ Genom Precis Med 2019; 12:e002376
  11. Crooke ST, Geary RS. Clinical pharmacological properties of mipomersen (Kynamro), a second generation antisense inhibitor of apolipoprotein B. Br J Clin Pharmacol 2013; 76:269-276
  12. Feingold KR. Cholesterol Lowering Drugs. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Grossman A, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  13. Burnett JR, Hooper AJ, Hegele RA. Abetalipoproteinemia. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A, eds. GeneReviews((R)). Seattle (WA)2018.
  14. Lee J, Hegele RA. Abetalipoproteinemia and homozygous hypobetalipoproteinemia: a framework for diagnosis and management. J Inherit Metab Dis 2014; 37:333-339
  15. Wetterau JR, Lin MC, Jamil H. Microsomal triglyceride transfer protein. Biochim Biophys Acta 1997; 1345:136-150
  16. Hussain MM, Shi J, Dreizen P. Microsomal triglyceride transfer protein and its role in apoB-lipoprotein assembly. J Lipid Res 2003; 44:22-32
  17. Cuchel M, Rader DJ. Microsomal transfer protein inhibition in humans. Curr Opin Lipidol 2013; 24:246-250
  18. Debose-Boyd RA, Horton JD. Opening up new fronts in the fight against cholesterol. Elife 2013; 2:e00663
  19. Seidah NG, Awan Z, Chretien M, Mbikay M. PCSK9: a key modulator of cardiovascular health. Circ Res 2014; 114:1022-1036
  20. Shapiro MD, Tavori H, Fazio S. PCSK9: From Basic Science Discoveries to Clinical Trials. Circ Res 2018; 122:1420-1438
  21. Abifadel M, Varret M, Rabes JP, Allard D, Ouguerram K, Devillers M, Cruaud C, Benjannet S, Wickham L, Erlich D, Derre A, Villeger L, Farnier M, Beucler I, Bruckert E, Chambaz J, Chanu B, Lecerf JM, Luc G, Moulin P, Weissenbach J, Prat A, Krempf M, Junien C, Seidah NG, Boileau C. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat Genet 2003; 34:154-156
  22. Cohen JC, Boerwinkle E, Mosley TH, Jr., Hobbs HH. Sequence variations in PCSK9, low LDL, and protection against coronary heart disease. N Engl J Med 2006; 354:1264-1272
  23. Zhao Z, Tuakli-Wosornu Y, Lagace TA, Kinch L, Grishin NV, Horton JD, Cohen JC, Hobbs HH. Molecular characterization of loss-of-function mutations in PCSK9 and identification of a compound heterozygote. Am J Hum Genet 2006; 79:514-523
  24. Cariou B, Ouguerram K, Zair Y, Guerois R, Langhi C, Kourimate S, Benoit I, Le May C, Gayet C, Belabbas K, Dufernez F, Chetiveaux M, Tarugi P, Krempf M, Benlian P, Costet P. PCSK9 dominant negative mutant results in increased LDL catabolic rate and familial hypobetalipoproteinemia. Arterioscler Thromb Vasc Biol 2009; 29:2191-2197
  25. Arca M, D'Erasmo L, Minicocci I. Familial combined hypolipidemia: angiopoietin-like protein-3 deficiency. Curr Opin Lipidol 2020; 31:41-48
  26. Kersten S. Angiopoietin-like 3 in lipoprotein metabolism. Nat Rev Endocrinol 2017; 13:731-739
  27. Adam RC, Mintah IJ, Alexa-Braun CA, Shihanian LM, Lee JS, Banerjee P, Hamon SC, Kim HI, Cohen JC, Hobbs HH, Van Hout C, Gromada J, Murphy AJ, Yancopoulos GD, Sleeman MW, Gusarova V. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res2020; 61:1271-1286
  28. Burnett JR, Hooper AJ, Hegele RA. Familial Combined Hypolipidemia. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Bean LJH, Gripp KW, Amemiya A, eds. GeneReviews((R)). Seattle (WA) 2023.
  29. Minicocci I, Santini S, Cantisani V, Stitziel N, Kathiresan S, Arroyo JA, Marti G, Pisciotta L, Noto D, Cefalu AB, Maranghi M, Labbadia G, Pigna G, Pannozzo F, Ceci F, Ciociola E, Bertolini S, Calandra S, Tarugi P, Averna M, Arca M. Clinical characteristics and plasma lipids in subjects with familial combined hypolipidemia: a pooled analysis. J Lipid Res 2013; 54:3481-3490
  30. Dewey FE, Gusarova V, Dunbar RL, O'Dushlaine C, Schurmann C, Gottesman O, McCarthy S, Van Hout CV, Bruse S, Dansky HM, Leader JB, Murray MF, Ritchie MD, Kirchner HL, Habegger L, Lopez A, Penn J, Zhao A, Shao W, Stahl N, Murphy AJ, Hamon S, Bouzelmat A, Zhang R, Shumel B, Pordy R, Gipe D, Herman GA, Sheu WHH, Lee IT, Liang KW, Guo X, Rotter JI, Chen YI, Kraus WE, Shah SH, Damrauer S, Small A, Rader DJ, Wulff AB, Nordestgaard BG, Tybjaerg-Hansen A, van den Hoek AM, Princen HMG, Ledbetter DH, Carey DJ, Overton JD, Reid JG, Sasiela WJ, Banerjee P, Shuldiner AR, Borecki IB, Teslovich TM, Yancopoulos GD, Mellis SJ, Gromada J, Baras A. Genetic and Pharmacologic Inactivation of ANGPTL3 and Cardiovascular Disease. N Engl J Med 2017; 377:211-221
  31. Levy E, Poinsot P, Spahis S. Chylomicron retention disease: genetics, biochemistry, and clinical spectrum. Curr Opin Lipidol 2019; 30:134-139
  32. Sissaoui S, Cochet M, Poinsot P, Bordat C, Collardeau-Frachon S, Lachaux A, Lacaille F, Peretti N. Lipids Responsible for Intestinal or Hepatic Disorder: When to Suspect a Familial Intestinal Hypocholesterolemia? J Pediatr Gastroenterol Nutr 2021; 73:4-8

 

Hypertriglyceridemia: Pathophysiology, Role of Genetics, Consequences, and Treatment

ABSTRACT

 

Hypertriglyceridemia (HTG) can result from a variety of causes. Mild to moderate HTG tracks along with the metabolic syndrome, obesity and diabetes. HTG can be the result of multiple small gene variants or secondary to several diseases and drugs. Severe HTG with plasma triglyceride (TG) levels >1000-1500 mg/dL typically results from: (1) rare variants in the lipoprotein lipase (LPL) complex, where it is termed the familial chylomicronemia syndrome (FCS), and (2) the co-existence of genetic and secondary forms of HTG, termed the multifactorial chylomicronemia syndrome (MFCS), which is a much more common cause of severe HTG.  Mild to moderate HTG is associated with an increased risk of premature cardiovascular disease (CVD), while severe HTG can lead to pancreatitis as well as an increased risk of premature CVD. Appropriate management of the patient with HTG requires knowledge of the likely cause of the HTG, to prevent itscomplications.

 

PHYSIOLOGY

 

A detailed overview of lipoprotein physiology is provided in the Endotext chapter on Lipoprotein Metabolism (1).  Here we will briefly review some aspects the metabolism of the triglyceride (TG)-rich lipoproteins, very low-density lipoproteins (VLDL) and chylomicrons (CM) of particular relevance to this chapter.

 

Secretion of TG-rich Lipoproteins Into Plasma

 

TGs are transported through plasma as VLDL), which transport TGs primarily made in the liver, and as CM, which transport dietary (exogenous) fat.  VLDL secretion by the liver is regulated in several ways.  Each VLDL particle has one ApoB100 molecule, making ApoB100 availability a key determinant of the number of VLDL particles, and hence, TG secretion by the liver.  In addition to one molecule of ApoB-100, each VLDL particle contains multiple copies of other apolipoproteins, together with varied amounts of TGs, cholesteryl esters, and phospholipids.  The extent of TG synthesis is in part determined by the flux of free fatty acids (FFA) to the liver.  The addition of TG to the developing VLDL particle in the endoplasmic reticulum is mediated by the enzyme microsomal triglyceride transfer protein (MTTP).  The pool of ApoB100 in the liver is not typically regulated by its level of synthesis, which is relatively constant, but by its level of degradation, which can occur in several proteolytic pathways (2). Insulin also plays a role in the regulation of VLDL secretion -  it decreases hepatic VLDL production by limiting fatty acid influx into the liver and decreases the stability of, and promotes the posttranslational degradation of ApoB100 (3).  Recent studies have shown that ApoC-III, an Apolipoprotein thought to primarily play a role in inhibiting TG removal (see below), also is involved in the assembly and secretion of VLDL (4).  VLDL particles (containing ApoB100) also increase in plasma in the postprandial state as well as CM that contain ApoB48 (5).

 

Consumption of dietary fat results in the formation of CM by enterocytes.  Fatty acids and monoacylglycerols that result from digestion of dietary TGs by acid and pancreatic lipases are transported into enterocytes by mechanisms that are not completely understood.  In the enterocyte, monoacylglycerol and fatty acids are resynthesized into TGs by the action of the enzymes acyl-coenzyme A: monoacylglycerol acyltransferase and acyl-coenzyme A: diacylglycerol acyltransferase 1 and 2 (DGAT 1 and 2).  The resulting TGs are packaged with ApoB48 to form CM, a process also mediated by MTTP (6).   CM then pass into the thoracic duct from where they enter plasma and acquire additional apolipoproteins.  Of particular relevance to their clearance from plasma is the acquisition of ApoC-II and ApoC-III. 

 

Catabolism of the TG-rich Lipoproteins

 

TGs in both VLDL and CM are hydrolyzed by the lipoprotein lipase (LPL) complex.  LPL is synthesized by several tissues, including adipose tissue, skeletal muscle, and cardiac myocytes.  After secretion by adipocytes, the enzyme is transported by glycosylphosphatidylinositol-anchored high-density lipoprotein–binding protein 1 (GPIHBP1) to the luminal side of the capillary endothelium, where it becomes tethered to glycosaminoglycans (GAGs). This pool of LPL is referred to as “functional LPL”, since it is available to hydrolyze TGs in both VLDL and chylomicrons. LPL can be liberated from these GAG binding sites by heparin injection. Several other proteins, reviewed in (7), regulate LPL activity. These include ApoC-II, which activates LPL, and ApoC-III, which inhibits LPL in addition to its effect on VLDL secretion alluded to earlier. Both are produced by the liver and are present on TG-rich lipoproteins.  ApoC-III also inhibits the turnover of TG-rich lipoproteins through a hepatic clearance mechanism involving the LDL receptor/LDL receptor-related protein 1 (LDLR/LRP1) axis (8).  ApoE also is present on the TG-rich lipoproteins and plays an important role in the uptake and clearance of the remnants of the TG-rich lipoproteins that result from hydrolysis of TGs in these lipoproteins. Other activators of LPL include ApoA-IV (9), ApoA-V (10-12) and lipase maturation factor 1 (LMF1) (13, 14). In addition, several members of the angiopoietin-like (ANGPTL) protein family play a role in regulating LPL activity.  ANGPTL3 is produced by the liver and is an endocrine regulator by inhibiting LPL in peripheral tissues (7, 15, 16).  ANGPTL4 is produced in several tissues (7), where it inhibits LPL in a paracrine fashion (7, 17). Both ANPGTL3 and ANGPTL4 delay the clearance of the TG-rich lipoproteins (7).

 

The core TGs in VLDL and chylomicrons are hydrolyzed by ApoC-II activated LPL; FFA thus formed are taken up by adipocytes and re-incorporated into TGs for storage, or in skeletal and cardiac muscle, utilized for energy. Hydrolysis of chylomicron- and VLDL-TG results in TG-poor, cholesteryl ester and ApoE-enriched particles called chylomicron and VLDL remnants, respectively, which under physiological conditions are removed by the liver by binding to LDL receptors, LDL receptor related protein, and cell surface proteoglycans (12, 18). Hepatic TG lipase and ApoA-V also are involved in the remnant clearance process (10-12, 19, 20).

 

The clearance of TGs from plasma is saturable when plasma TGs exceed ~500-700 mg/dL (21).  When removal mechanisms are saturated, additional chylomicrons and VLDL entering plasma cannot readily be removed and hence accumulate in the plasma. As a result, plasma TGs can increase dramatically, resulting in very high levels and the accumulation of chylomicrons in plasma obtained after an overnight fast. 

 

NORMAL RANGE FOR PLASMA TRIGLYCERIDES AND DEFINITION OF HYPERTRIGLYCERIDEMIA

 

Plasma TG levels reflect the TG content of multiple lipoprotein particles, primarily chylomicrons and VLDL. Fasting TG levels less than 150 mg/dL has been generally accepted as “normal” (22, 23). A non-fasting TG of 175mg/dL represents ~75th percentile of the normal range, and levels of ~400mg/dL represent the 97th percentile (24). Plasma TGs are heavily skewed to the right in the general population with a tail towards highest levels and vary depending upon the population mix (25). The full range of TG extends from 30mg/dL to 10,000mg/dL (22). TG levels are different between sexes, being higher in males than in females, and increase with age and development of other coexisting conditions such as central adiposity, metabolic syndrome, and diabetes (24). TG levels also vary between geographic areas, among people of different ethnic backgrounds, with higher levels observed in certain populations such as Mexicans and South Asians. Lower TG levels have been observed in people of African descent and African-Americans but this may be changing due to adoption of urban lifestyles (26). Because of this skewed distribution, logarithmic transformation is required to establish statistical normal ranges of TG levels.  There is no current widely accepted definition of elevated non-fasting TG levels, but some groups have utilized 175mg/dL as a cut point (27, 28). Due to high variability of TG levels, precise definitions for non-fasting levels are difficult to establish. It is worthwhile noting that post-prandial TG levels rarely exceed 400mg/dL even after a high fat challenge.

 

Normal Range Based on Risk of Complications of Hypertriglyceridemia

 

The major complications of hypertriglyceridemia (HTG) are (1) acute pancreatitis and (2) increased risk of atherosclerotic cardiovascular disease (ASCVD). These two complications occur at different levels of TGs, the risk of pancreatitis occurring at much higher TG levels than the risk of premature ASCVD and are discussed in detail later in this chapter. 

 

Normal Range According to Guidelines

 

Despite concerns regarding establishment of an upper limit of normal for TGs, most guidelines define values for HTG, often without a strong biological rationale. Definitions for the diagnosis of HTG provided in several guidelines are shown in Table 1.   

 

Cut points for HTG were first defined by the National Cholesterol Education Program Adult Treatment Panel (NCEP-ATP). The terms mild, moderate and severe have been used based on degree of TG elevation (table 1). In general, mild to moderate HTG reflects TG levels under 500mg/dL. Severe hypertriglyceridemia (sHTG) has been arbitrarily defined by different national guidelines as either TG levels ≥500 mg/dL by the American Heart Association (AHA)/American College of Cardiology (ACC), Multispecialty Cholesterol and Canadian Cardiovascular Society Guidelines (29, 30) or TG levels ≥880 mg/dL according to the European Society of Cardiology guidelines (31). The Endocrine Society has used severe HTG for 1000 to 1999 mg/dL and very severe HTG for values >2000 mg/dL (23).  

 

Table 1. Definition of Hypertriglyceridemia According to Various Clinical Guidelines

Guideline

Classification

Triglyceride Levels

NCEP/ ATP III (32)

American Heart Association (33)

National Lipid Association (34)

Normal

Borderline-high TGs

High TGs

Very high TGs

<150 mg/dL (< 1.7 mmol/L)

150-199 mg/dL (1.7-2.3 mmol/L)

200-499 mg/dL (2.3-5.6 mmol/L)

≥500 mg/dL (≥5.6 mmol/L)

The Endocrine Society (35)

Normal

Mild HTG

Moderate HTG

Severe HTG

Very severe HTG

<150 mg/dL (< 1.7 mmol/L)

150-199 mg/dL (1.7-2.3 mmol/L)

200-999 mg/dL (2.3-11.2 mmol/L)

1000-1999 mg/dL (11.2-22.4 mmol/L)

≥2000 mg/dL (≥22.4 mmol/L)

European Society of Cardiology/European Atherosclerosis Society (36)

Normal

Mild-moderate HTG

Severe HTG

<1.7 mmol/L (<150mg/dL)

>1.7-< 10mmol/L (150-880 mg/dL)

> 10 mmol/L (> 880mg/dL)

Hegele (22)

Normal

Mild to moderate

Severe

<2.0 mmol/L (<175 mg/dL)

2.0-10 mmol/L (175- 885 mg/dL)

>10 mmol/dL (>885 mg/dL)

 

In summary, establishing a precise definition of what constitutes abnormal TG values is fraught with difficulty.  An acceptable level for the prevention of pancreatitis is likely to be quite different from that at which CVD risk might be increased. The impact of HTG on CVD risk needs to be evaluated in the context of the family history of premature CVD, associated abnormalities of lipids and lipoproteins, and other CVD risk factors, particularly those associated with the metabolic syndrome (see below).

 

CAUSES AND CLASSIFICATION OF HYPERTRIGLYCERIDEMIA

 

In general, HTG has been classified as primary, when a genetic or familial basis is suspected, or secondary, where other conditions that raise TG levels can be identified. However, this classification is likely overly simplistic. It has become clear in the past decade that the spectrum of plasma TG levels, ranging from mild elevation to very severe HTG, is modulated by a multitude of genes working in concert with non-genetic secondary and environmental contributors. Thus, in the vast majority of individuals, mutations in multiple genes with interaction from non-genetic factors result in altered TG-rich lipoprotein synthesis and catabolism and subsequent HTG.

 

Historical Perspective

 

Phenotypic heterogeneity among patients with HTG has been historically defined by qualitative and quantitative differences in plasma lipoproteins. In the pre-genomic era, the Fredrickson classification of hyperlipoproteinemia was based on electrophoretic patterns of lipoprotein fractions (37). The phenotypes are distinguished based on the specific class or classes of accumulated TG-rich lipoprotein particles, including chylomicrons, VLDL and VLDL-remnants. This classification included 6 phenotypes, five of which included HTG in their definition (except for Frederickson type 2 A hyperlipoproteinemia, which equates with genetic primary hypercholesterolemia). It has now become apparent that except for type 1 hyperlipoproteinemia (FCS), the HTG phenotypes, particularly Frederickson type 4 and type 5 hyperlipoproteinemia, are due to the accumulation of polygenic traits predisposing to HTG. However, this classification system is dated, has neither improved clinical or scientific insight, and therefore does not find wide use at this time (22).

 

In 1973, Goldstein and colleagues characterized a variable pattern of lipid abnormalities in families of survivors of myocardial infarction that they termed familial combined hyperlipidemia (FCHL) (38).  At the same time, this phenotype of mixed or combined hyperlipidemia was observed in another cohort, where it was called multiple-type familial hyperlipoproteinemia (39).  Affected family members can present with hypercholesterolemia alone, HTG alone, or with elevations in both TGs and LDL. This pattern was estimated to have a population prevalence of 1-2% (40), making it the most common inherited form of dyslipidemia.

 

In the aforementioned study, a pattern of isolated HTG, historically called familial HTG (FHTG) also was described (38). This condition was characterized by increased TG synthesis, with secretion of normal numbers of large TG-enriched VLDL particles (41), elevated VLDL levels, but normal levels of LDL and HDL cholesterol (42).  FHTG did not appear to be associated with an increased risk of premature CVD in an early study (43), but baseline TG levels predicted subsequent CVD mortality after 20 years of follow up among relatives in families classified as having FHTG (44, 45).  

 

FCHL and FHTG were initially believed to be monogenic disorders (38). However, more recent genetic characterization of individuals with familial forms of HTG indicates that these are not disorders associated with variation within a single gene, but rather polymorphisms in multiple genes associated with HTG, as detailed below. Therefore, classification of FCHL and FHTG is potentially misleading. Nevertheless, it is important to note that FCHL as originally described is associated with a very high prevalence of premature CVD (43, 44, 46).   

 

Genetic Forms of Hypertriglyceridemia

 

It is now evident that clinically relevant abnormalities of plasma TG levels appear to require a polygenic foundation of common or rare genetic variants (22).  Common small-effect gene variants confer a background predisposition that interact with rare large-effect heterozygous variants in genes that govern synthesis or catabolism of TG-rich lipoproteins, or nongenetic secondary factors, leading to the expression of a more severe TG phenotype (47). Recently, the most prevalent genetic feature underlying severe HTG was shown to be the polygenic accumulation of common (rather than rare) variants—more specifically, the accumulation of TG-raising alleles across multiple SNP loci (48).

 

Thus, mild to moderate hypertriglyceridemic states are complex, genetically heterogeneous disorders. Mild-to-moderate HTG is typically polygenic and results from the cumulative burden of common and rare variants in more than 30 genes, as quantified by genetic risk scores. All genetic forms can be exacerbated by non-genetic factors. Because they are a consequence of interaction between multiple susceptibility genes and lifestyle factors, individuals with moderate HTG should be considered as a single group without distinction, irrespective of concomitant lipoprotein disturbances (22). Because of the complexity of these disorders, routine genetic testing is not recommended.

 

Pathogenesis of Genetic Forms of Hypertriglyceridemia

           

Genetic forms of HTG without other lipoprotein disturbances (i.e., pure HTG) are characterized by increased TG synthesis, where normal numbers of large TG-enriched VLDL particles are secreted (41, 49-51). Reduced TG clearance also has been observed in some individuals (50-52).  Affected people have elevated VLDL levels, but normal levels of LDL, and are generally asymptomatic unless clinical CVD or severe HTG develops. 

 

A variety of metabolic defects that differ among families are associated with the combined hyperlipidemia phenotype. The characteristic lipoprotein abnormalities are increased ApoB levels and increased number of small dense LDL particles (42), a phenotype similar to that seen in the metabolic syndrome and type 2 diabetes (53). These primary defects occur due to 1) hepatic overproduction of VLDL particles (41) due to increased ApoB synthesis in the setting of disordered adipose metabolism (54, 55), insulin resistance (41, 56-58), and liver fat accumulation, and, 2) impaired clearance of ApoB containing particles (59, 60). Increased VLDL secretion results in an elevated plasma ApoB and HTG (56).  Long residence time of VLDL particles favors the formation of small dense LDL (59). An abundance of small dense LDL particles traditionally is associated with the presence of HTG; however, these LDL characteristics remain even after correction of the HTG by treatment with fibrates (61, 62).

 

In addition to Apo B abnormalities, other lipoprotein disturbances include abnormal expression of ApoA-II, ApoC-III, and PCSK9.  VLDL-TG levels in combined hyperlipidemia are modulated by ApoA-II and ApoC-III (63).  Plasma PCSK9 levels are higher in these patients, and levels correlate with TG and Apo B levels (64).

 

Visceral adiposity appears to be an important determinant of insulin resistance, which occurs commonly in subjects with both isolated HTG (65) and combined hyperlipidemia (65-69). Other abnormalities that have been reported in clinical FCHL include impaired lipolysis due to decreased cyclic AMP dependent signaling (54, 69), abnormal adipocyte TG turnover (70), fatty liver (71), increased arterial stiffness (72), and increased carotid intimal-medial thickness (73). 

 

In all of the phenotypes described above, severe HTG can occur when secondary causes of HTG such as untreated diabetes, marked weight gain, or use of TG-raising drugs are present concurrently, leading to the Multifactorial Chylomicronemia Syndrome (MFCS), described later (74).

 

Secondary Forms of Hypertriglyceridemia

 

These are described in greater detail in the chapters on Secondary Disorders of Lipid and Lipoprotein Metabolism (75-78).  However, in the section where we describe MFCS we will briefly touch on some aspects of secondary forms of HTG, since they assume importance in the pathogenesis of the severe HTG seen in the MFCS, where they often co-exist in individuals with genetic forms of HTG. In our experience, the commonest secondary forms of HTG that interact with genetic forms of HTG are type 2 diabetes (usually as part of the metabolic syndrome), obesity, recent weight gain, excessive alcohol consumption, the use of drugs that can raise TGs, and chronic kidney disease (CKD)(74, 79, 80). (Table 3)

           

Severe Hypertriglyceridemia and the Chylomicronemia Syndrome

 

In the late 1960s Fredrickson, Levy and Lees (37) classified HTG into types dependent on the pattern of lipoproteins on paper electrophoresis and the presence or absence of chylomicrons in fasting plasma. They recognized that acute pancreatitis and eruptive xanthomata occurred in the presence of chylomicronemia that accumulate in what they termed Type I and Type V hyperlipoproteinemia. Chylomicrons are present in the post-prandial state, and usually are present in fasting plasma when TG levels exceed 800 mg/dL, but absent in fasting plasma below that value (81). The term chylomicronemia syndrome was first used to describe a constellation of clinical findings such as abdominal pain, acute pancreatitis, eruptive xanthoma and lipemia retinalis that occurred in association with very high TG levels (82). Two groups of conditions can lead to severe HTG and clinical manifestations of the chylomicronemia syndrome; (1) familial chylomicronemia syndrome (FCS) due to variants in the LPL complex, and (2) multifactorial chylomicronemia syndrome (MFCS), in which genetic predisposition and secondary forms of HTG co-exist.

 

FAMILIAL CHYLOMICRONEMIA SYNDROME (FCS)

 

FCS is a monogenic disorder due to variants in one or more genes of the LPL complex that affect chylomicron catabolism. FCS incidence is very rare, with an estimated prevalence ranging from 1 in 20,000 to 300,000 (83).    

 

Genetics: Biallelic loss of function variants in five canonical genes lead to impaired hydrolysis of TG-rich lipoproteins, with subsequent increases in chylomicron particle numbers and markedly increased TG concentrations. The most common gene affected in FCS is LPL itself, in which patients are homozygous or compound heterozygous for two defective LPL alleles. Over 180 variants that result in LPL deficiency have been described with some clustered mutations due to founder effects (84-87). Loss of function variants account for over 90% of cases (83). Many are missense variants, some in catalytically important sites and some in regions that predispose to instability of the homodimeric structure of LPL required for enzyme activity (88). However, many common LPL gene variants have been described that have no clinical phenotype (89). Variants in the APOC2 gene, encoding ApoC-II, an activator of LPL, is another cause of FCS.  Variants have been described in several families (90, 91). In FCS thus far there is no known gene variant that affects synthesis or production of TG-rich lipoproteins.

 

FCS can also occur from biallelic loss of function variants in other components of the LPL complex, namely APOA5, LMF1, and GPIHBP1 genes (Table 2), each of which plays an important role in determining LPL function (92). The lipoprotein phenotype in these mimics that seen in classical LPL deficiency. Loss of function variants in GPIHBP1, which directs transendothelial LPL transport and helps anchor chylomicrons to the endothelial surface near LPL, thereby providing a platform for lipolysis, has been described in several families (83).  Autoantibodies to GPIHBP1 also can lead to chylomicronemia (93). A small number of individuals with homozygous variants in Apo A-V, which stabilizes  the lipoprotein–enzyme complex thereby enhancing lipolysis (10), have been described (94). Variants in LMF1, an endoplasmic reticulum chaperone protein required for post-translational activation of LPL, have also been identified in a few individuals (95).

 

Clinical presentation: FCS usually manifests in childhood or early adolescence with nausea, vomiting, failure to thrive and recurrent abdominal pain in infancy and childhood. Occasionally it can present in adulthood (87) but this is often due to delayed diagnosis with median age at diagnosis being due to unfamiliarity in most healthcare providers (96). Adults may report “brain fog” or transient confusion.

 

Classical clinical findings include eruptive xanthomas (often seen on buttocks, back, extensor surfaces of upper limbs), lipemia retinalis, and hepatosplenomegaly. Less common symptoms of FCS can include intestinal bleeding, anemia, and neurological features such as irritability and seizures. Patients present with TG levels ≥1000 mg/dL and often much higher, due to abnormal accumulation of chylomicrons, which can be detected by the appearance of lipemic/milky plasma.  Despite prolonged overnight fasting, plasma TG levels are >1000mg/dL due to the presence of chylomicrons in the circulation as a result of impaired clearance. The most serious concern, however, is the development of acute pancreatitis, which can lead to systemic inflammatory response syndrome, multi-organ failure, and death.

 

Table 2. Genetic Disorders Resulting in Familial Chylomicronemia Syndromes (FCS)

Disorder

Inheritance

Incidence

Lipid Phenotype

Underlying Defect

Clinical Features

LPL deficiency

Autosomal Recessive

1 in 1,000,000

Marked HTG/ chylomicronemia in infancy or childhood

Very low or absent LPL activity; circulating inhibitor of LPL

Hepato-splenomegaly; severe chylomicronemia

Apo C-II deficiency

Autosomal Recessive

Rare

Marked HTG/ chylomicronemia in infancy or childhood

Absent Apo C-II

Hepato-splenomegaly; severe chylomicronemia

Apo A-V mutation

Autosomal Recessive

Rare

Marked HTG/ chylomicronemia in adulthood

Defective or absent Apo A-V

Chylomicronemia

GPIHBP1 mutation

Autosomal Recessive

Rare

Marked HTG/ chylomicronemia in adulthood

Defective or absent GPIHBP1

Chylomicronemia

LFM1 mutation

Autosomal Recessive

Rare

Marked HTG/ chylomicronemia in adulthood

Defective or absent LFM1

Chylomicronemia

 Adapted from Ref (83)

 

MULTIFACTORIAL CHYLOMICRONEMIA SYNDROME (MFCS)

 

In contrast to FCS, MFCS is more common and complex. The prevalence of MFCS is much higher than FCS and estimated to be ~1:600-1000 (84).   

 

Genetics: MFCS has a genetic basis, but unlike FCS (where recessive or biallelic variants in the affected genes are causative), the genetic alteration does not always result in the phenotypic expression of the trait but only increases the possibility of the risk of developing the condition. Other factors, including non-genomic effects (epigenetics, methylation), gene-gene, or gene-environment interactions can also contribute. MFCS develops due to two main types of genetic factors that increase the odds that a patient will develop very high TG levels. First, heterozygous rare large-effect variants in one of the five canonical TG metabolism genes (LPL, Apo C-II, Apo A5, LMF-1 and GPIHBP-1) can contribute to TG elevations. These variants have variable penetrance, i.e.- clinical presentation can vary from normal to severe hypertriglyceridemia.

 

Secondly, the presence of a high burden of common small-effect TG-raising SNPs; cumulatively, these common SNP alleles increase susceptibility for developing hypertriglyceridemia. These SNPs may have an indirect impact of the metabolism of TG-rich

Lipoproteins. There is incomplete understanding of how an excess burden of SNPs contributes to TG levels, but their prevalence in patients with severe hypertriglyceridemia has been consistently demonstrated.

 

Several polygenic risk scores (PRS) for TG levels have been published (97). A recent study found that 32.0% of patients had a high polygenic score of TG-raising alleles across 16 loci compared to only 9.5% of normolipidemic controls (25). When the PRS is high, there is a significantly increased risk of developing HTG but this is not diagnostic or definitive.

 

Secondary Causes Contributing to Severe Hypertriglyceridemia in MFCS: The most common secondary cause in the past was undiagnosed or untreated diabetes (74), although earlier detection of diabetes may be making the association of marked hyperglycemia of untreated diabetes with very severe HTG less common. In addition, individuals with the metabolic syndrome and obesity have mild to moderate HTG which can become severe HTG; weight regain following successful weight loss can lead to marked HTG (23, 84). These patients almost always have relatives with genetic forms of HTG, whose TG levels are considerably lower than the index patient with severe HTG, in whom secondary forms of HTG also are present (74).  MFCS can result from the addition of specific drugs in patients with a genetic predisposition (23). These drugs include beta-adrenergic blocking agents (selective and non-selective) and/or diuretics (thiazides and loop-diuretics such as furosemide) used for hypertension, retinoid therapy for acne, oral estrogen therapy for menopause or birth control, selective estrogen receptor modulators (particularly raloxifene) for osteoporosis or breast cancer, protease inhibitors for HIV/AIDS, atypical anti-psychotic drugs, alcohol, and possibly sertraline (84).  Rarer causes of very severe HTG include autoimmune disease (sometimes with LPL- or GPIHBP1- specific antibodies), asparaginase therapy for acute lymphoblastic leukemia (98), (99) and bexarotene, a RXR agonist used in the treatment of cutaneous T cell lymphoma (100).  

 

Table 3. Secondary Causes That Can Contribute to Severe HTG

Conditions

Hypothyroidism

Suboptimally managed or new onset diabetes

Obesity

Sudden weight gain, weight regain after weight loss

Chronic kidney disease

Nephrotic syndrome

Pregnancy

Acute hepatitis

Sepsis

Inflammatory disorders

Cushing syndrome

Autoimmune chylomicronemia

            Systemic lupus erythematosis

            Anti-LPL antibodies

GPIHBP-1 antibodies

Rare Genetic Causes

Glycogen storage disorders

Lipodystrophies

            Congenital- generalized or partial

            Acquired- HIV, autoimmune

Drugs

Alcohol ingestion

Beta blockers

Thiazide diuretics

Oral estrogens

Selective estrogen reuptake modulators - tamoxifen, raloxifene, clomiphene

Androgens

Glucocorticoids

Atypical anti-psychotics

Sertraline

Bile acid resins

Sirolimus, tacrolimus

Cyclosporine

RXR agonists -bexarotene, isotretinoin, acetretin

HIV Protease inhibitors

L- asparaginase

Alpha-interferon

Propofol

Lipid emulsions

 

Following correction of treatable secondary forms of HTG in the MFCS, TG levels usually decrease to the moderately elevated levels seen in their affected relatives (101, 102).  

 

OTHER CONDITIONS RESULTING IN HYPERTRIGLYCERIDEMIA

 

Familial Dysbetalipoproteinemia (FDB or Remnant Removal Disease)

 

Familial dysbetalipoproteinemia, also referred to as remnant removal disease or type III hyperlipoproteinemia, is a rare autosomal recessive disorder that can present with elevated TG levels. This disorder is characterized by the accumulation of remnant lipoproteins.

 

PATHOGENESIS AND GENETICS

 

Remnant removal disease requires homozygosity for the ApoE2 genotype or a rare heterozygosity for a variant in the ApoE gene, which results in pathologic accumulation of remnant lipoproteins in the circulation due to impaired hepatic uptake of ApoE-containing lipoproteins (103). ApoE is a glycoprotein synthesized in the liver, brain and tissue macrophages and present on chylomicrons, VLDL and HDL. Apo E through interaction with the LDLR and heparan sulphate proteoglycans promotes the hepatic clearance of remnants of chylomicrons and VLDL (104); it also facilitates cholesterol efflux from macrophages to HDL (105). In humans, there are 3 common isoforms of ApoE , ApoE2, ApoE3, and ApoE4 (106).  Each differs in isoelectric point by one charge unit, ApoE4 being the most basic isoform and ApoE2 the most acidic.  ApoE3 (Cys112Arg158) is the commonest isoform.  ApoE2 (Arg158Cys) and ApoE4 (Cys112Arg) differ from ApoE3 by single amino acid substitutions at positions 158 and 112, respectively (107).  In the majority of cases (90%), remnant removal disease is associated with the E2/E2 genotype and results from impaired binding to the Apo E receptor. It is  an autosomal recessive disorder with the prevalence of ApoE2 homozygosity in Caucasian populations estimated to be about 1% (108).  Rarer ApoE variants such as ApoE3-Leiden (109) and ApoE2 (Lys1463Gln) that also can cause remnant accumulation are dominantly inherited (110) and account for 10% of cases (111, 112).  Rare APOE variants in the population, other than the APOE2 and APOE4 alleles, play an important role in the development of isolated hypercholesterolemia (113) and mixed hyperlipidemia, with and without familial dysbetalipoproteinemia (114). Thus it is becoming apparent that two different DBL phenotypes may exist- a genetic Apo E dysfunction and a multifactorial form (115). Modern prevalence of FDB is estimated at 1-2% (116).

 

In the absence of additional genetic, hormonal, or environmental factors, remnants do not accumulate to a degree sufficient to cause hyperlipidemia in ApoE2 homozygotes; in fact, lipid levels are commonly low. Remnant accumulation results when the E2/2 genotype is accompanied by a second genetic or acquired defect that causes overproduction of VLDL such as obesity or diabetes (117) (111, 118) , a decrease in remnant clearance, or a reduction in LDL receptor activity (e.g., hypothyroidism (119)). Thus, full phenotypic expression requires the presence of other environmental or genetic factors (120). In these circumstances, the reduced uptake of remnant lipoproteins by the liver results in reduced conversion of VLDL and intermediate density lipoproteins to LDL, with subsequent accumulation of remnant lipoproteins (121, 122), hence the term remnant removal disease.

 

DIAGNOSIS

 

Patients with remnant removal disease have roughly equivalent elevations in plasma cholesterol and TGs. The disease rarely manifests before adulthood, and in some individuals never manifests clinically. It is more common in men than in women, where expression seldom occurs before menopause, since estrogen has a protective effect in women who are ApoE2 homozygotes (108). Palmar xanthomas (Figure 1), orange lipid deposits in the palmar or plantar creases, are pathognomonic of remnant removal disease but are not always present (123). Tuberoeruptive xanthomas can be found at pressure sites on the elbows, knees and buttocks. The presence of remnant removal disease should be suspected when total cholesterol and TG levels range from 300 to 1000 mg/dL and are roughly equal in magnitude. Special diagnostic tests such as beta-quantification or lipoprotein electrophoresis are often required and are time consuming and not widely available. VLDL particles are cholesterol- enriched, which can be determined by isolation of VLDL by ultracentrifugation and by the demonstration of beta migrating VLDL on lipoprotein electrophoresis.  A VLDL-cholesterol/plasma TG ratio of <0.30 is usually observed (124).  A low ApoB/total cholesterol ratio of <0.33 also can be helpful in making the diagnosis (125). Simplified criteria for the diagnosis of DBL using a 3-step process has been proposed (126). The diagnosis of remnant removal disease should be confirmed by demonstrating the presence of the E2/E2 genotype. If the genotype result is not E2/E2, an autosomal dominant variant of APOE should be suspected. There is a high prevalence of premature coronary artery disease (127-129) and peripheral arterial disease (130-132).  Occasionally severe HTG and an increased risk of pancreatitis can develop in the presence of a concomitant secondary form of HTG or TG-raising drugs.

Figure 1. Palmar Xanthomas: Orange-yellow discoloration confined to the palmar creases.

 

Familial Partial Lipodystrophy (FPLD) Syndromes

 

A distinct entity that results in moderate and severe hypertriglyceridemia include partial lipodystrophy syndromes. Inherited lipodystrophies are a heterogeneous group of disorders considered to be rare, that manifest as complete or partial loss of white adipose tissue with accompanying severe metabolic dysregulation(133) and are reviewed elsewhere in the Endotext chapter on Lipodystrophies (134).  Loss of fat can be either localized to small discrete areas, in some cases partial with loss from extremities, or generalized with fat loss from nearly the entire body. Inherited lipodystrophies, while rare, can be autosomal dominant or recessive.  Some forms manifest at birth, while others become evident later in life.

 

Partial or generalized lipodystrophic disorders frequently are associated with significant metabolic derangements associated with severe insulin resistance, including HTG. The extent of fat loss sometimes determines the severity of metabolic complications (135).  HTG is a common accompaniment of many lipodystrophies, often in conjunction with low HDL-C levels.    The pathophysiology of hypertriglyceridemia in these subjects is possibly related to the reduced ability to deposit free fatty acids in adipose tissue due to its maldevelopment, and accelerated lipolysis with increased hepatic VLDL synthesis and delayed clearance (135).

 

Genetics: Several genes have been implicated in the manifestation of various forms including LMNA, PPARG, LIPE, CIDEC (136). In the Dunnigan variety, the most commonly identified genetic variant of FPLD, the commonest variants are in the LMNA gene and less frequently PPARG (133).  No specific genetic defect has been identified in Köbberling’s FPLD, although recent evidence suggests a heavy polygenic burden in these individuals (137, 138).

 

Diagnosis: Congenital generalized lipodystrophy (CGL) is a rare autosomal recessive disorder in which near total absence of subcutaneous adipose tissue is evident from birth.  HTG and hepatic steatosis are evident at a young age and are often difficult to control. Severe HTG, often associated with eruptive xanthoma and recurrent pancreatitis, can occur in patients with CGL. The prevalence of HTG in case series of CGL patients is over 70% (135, 139).  Plasma TGs are normal or slightly increased during early childhood, with severe HTG manifesting at puberty along with onset of diabetes mellitus. 

 

Familial partial lipodystrophies (FPLD) are complex metabolic disorders that are often not recognized clinically (140).  Partial lipodystrophies are characterized by partial loss of adipose tissue and significant metabolic derangements.  The Dunnigan variety of FPLD (FPLD type 2) is a rare autosomal dominant disorder in which fat loss mostly involves the extremities and the trunk.  Onset of fat loss in the buttocks and extremities occurs at puberty or late adolescence, with gain of fat to the face and neck. Acanthosis nigricans, calf muscle hypertrophy, and phlebomegaly (prominent veins) due to lack of subcutaneous fat, can be observed. Significant metabolic dysfunction including diabetes, which is often very insulin resistant, resistant hypertension, and HTG often severe and difficult to treat, can occur.  Myopathy, cardiomyopathy, and/or conduction system abnormalities can occur (141).  ASCVD risk also is increased (142, 143).

 

Some lipodystrophies, where fat loss appears to be proportionate to loss of total and lean body mass, do not result in dyslipidemia. Elevated TG levels have been reported in patients with atypical progeroid syndrome due to LMNA mutations (144, 145).  Of the acquired lipodystrophies, the HIV-associated form usually is characterized by more moderate HTG.  HIV-associated lipodystrophy occurs in patients receiving protease inhibitor containing highly active anti-retroviral therapy regimens (146).  Fat loss occurs in the face, buttocks, and extremities.

 

CONSEQUENCES OF HYPERTRIGLYCERIDEMIA

 

Atherosclerotic Cardiovascular Disease

 

EPIDEMIOLOGY

 

HTG has long been known to be a risk factor for ASCVD (33, 147-150), which has been confirmed in meta-analyses (45).  However, HTG also is frequently associated with low levels of HDL-cholesterol and an accumulation of remnants of the TG-rich lipoproteins, both known risk factors for ASCVD.  When adjusted for both HDL-C and non-HDL-C, which contains both remnants of the TG-rich lipoproteins and LDL, the association of TGs with ASCVD risk remained significant, although somewhat attenuated (151).  Postprandial TGs are elevated throughout the day in subjects with HTG, and postprandial TG-rich lipoproteins and their remnants also have been hypothesized to be important in the pathogenesis of atherosclerosis (150). It is therefore of interest that non-fasting TGs have been associated with ASCVD risk (150, 152, 153), despite non-fasting TGs being quite variable. However, unlike the situation with elevated LDL-C levels, the magnitude of the TG elevation does not appear to correlate with the extent of ASCVD risk. In particular, very severe HTG per se does not always appear to confer increased ASCVD risk, possibly because the chylomicrons that accumulate are too large to enter the arterial intima (154, 155). 

 

TRIGLYCERIDES IN THE PATHOGENESIS OF ASCVD

 

Although chylomicrons may be too large to enter the arterial intima, ApoE-and cholesterol-enriched remnants of the TG-rich lipoproteins can enter with ease (153) where they can bind to vascular proteoglycans, similar to LDL (156, 157).  Modification of these retained lipoproteins by either oxidative damage or enzyme digestion of some of the lipid components can liberate toxic by-products, which have been hypothesized to play a role in atherogenesis by facilitating local injury, generation of adhesion molecule, and cytokine expression and inflammation (157).  Remnants of the TG-rich lipoproteins also can be taken up by macrophages leading to the formation of foam cells, an important component of atherosclerotic plaques.  HTG also is associated with a preponderance of small, dense LDL, particles, reduced levels of HDL-C, and in the metabolic syndrome, with abnormalities of HDL composition (see earlier). Small, dense LDL can traverse the endothelial barrier more easily than large, buoyant LDL particles (158), are retained more avidly than large, buoyant LDL (159), and also are more readily oxidized (160, 161), all of which may facilitate atherogenesis. HDL particles in some hypertriglyceridemic states, e.g., in association with the metabolic syndrome, might be dysfunctional with respect to their cholesterol efflux, anti-inflammatory, and anti-oxidant properties. Moreover, a hypercoagulable state has been reported in association with both HTG and the metabolic syndrome (162). Thus, HTG might accelerate atherosclerosis by several mechanisms, all of which could increase CVD risk.

 

GENETIC EVIDENCE OF HYPERTRIGLYCERIDEMIA AND ATHEROSCLEROSIS

 

Recent human genetic studies have provided important insight into the contribution of TGs to ASCVD. Several genetic approaches, including candidate gene sequencing, GWAS of common DNA sequence variants, and genetic analysis of TG phenotypes have unraveled new proteins and gene variants involved in plasma TG regulation (163). Some genetic variants that influence TG levels appear to be associated with increased CVD risk even after adjusting for their effects on other lipid traits (164).  GWAS have identified common noncoding variants of the LPL gene locus associated with TG and CVD risk (165, 166).  A common gain-of-function mutation in the LPL gene, S447X (10% allele frequency), is associated with reduced TG levels and reduced risk of CVD (167) and an LPL variant associated with reduced TG and ApoB levels was associated with reduced CVD similar to LDL-C lowering variants, suggesting that the clinical benefit of lowering triglyceride and LDL-C levels may be proportional to the absolute change in ApoB (168).  Conversely, several loss-of-function LPL variants linked with elevated TG levels are associated with increased CVD risk (169). Variants in the TRIB1 locus have been associated with LDL, HDL-C and TG levels (166), hepatic steatosis (170) and coronary artery disease (171). Mutations that disrupt APOC3 gene function and reduce plasma ApoC-III concentration are associated with lower TG levels and decreased risk of clinical CVD (172, 173).  In contrast, carriers of rare mutations in APOA5, encoding ApoA-V, an activator of LPL, are associated with elevated TGs and with increased risk of myocardial infarction (174, 175).  Loss of function variants in ANGPTL4 that had lower TG levels also were associated with reduced CVD risk (176, 177).  Thus, exciting new human genetics findings have causally implicated TG and TG-rich lipoproteins in the development of CVD risk. In particular, the LPL pathway and its reciprocal regulators ApoC-III and ApoA-V appear to have an important influence on atherosclerotic CVD risk. However, despite this mountain of evidence demonstrating a causal relationship of TG with atherosclerosis, the possible involvement of a correlated trait, usually low HDL-C levels, or other unmeasured traits, cannot be ruled out.

 

CLINICAL TRIAL EVIDENCE OF TRIGLYCERIDE LOWERING AND ASCVD

 

In the pre-statin era, use of gemfibrozil monotherapy demonstrated cardiovascular benefit in men with coronary heart disease. However, since the advent of statins, drugs that specifically lower plasma triglyceride levels have not clearly been shown to have a benefit with cardiovascular risk reduction in clinical trials when added to background statin therapy. Reasons for this are unclear. In genetic studies to get a comparable reduction in Apo B and coronary heart disease (CHD) risk in clinical trials, a TG reduction of ~70mg/dL is required compared to a decrease in LDL-C of only 14mg/dL. Additionally, lipid alterations due to genetics are lifelong and result in much bigger reductions in CHD than a 5-year drug study. Based on genetic studies, the magnitude of TG reduction required to demonstrate cardiovascular benefit is quite large.

 

CARDIOVASCULAR DISEASE IN THE CHYLOMICRONEMIA SYNDROME

 

As described earlier, chylomicrons have been considered to be too large to penetrate the vascular endothelium and play a role in atherogenesis (152), although  remnants of the TG-rich lipoproteins may be atherogenic (152, 178-181). The incidence of CVD is low in individuals with FCS (182), although premature atherosclerosis has been documented in well characterized subjects with this disorder (183).  However, CVD risk clearly is increased in many patients with MFCS, although the exact frequency remains unclear. The frequency of CVD outcomes does not appear to relate to the magnitude of the TG elevations (184). It is not surprising that CVD is increased in MFCS considering the association between TGs and CVD that has been documented in many studies (reviewed in (150, 185, 186)).  Many subjects develop severe HTG due to the co-existence of polygenic mutations that result in mild to moderate HTG (22) with secondary causes of HTG.  Residual HTG due to these genetic disorders persists even after severely elevated TG levels have been reduced by treatment of the secondary forms of HTG and treatment of the HTG per se.  Moreover, many patients with the MFCS have other CVD risk factors such as diabetes, reduced levels of HDL-C, and hypertension, the latter resulting in use of diuretics and beta-blockers, which play a role in raising their TGs to levels at which chylomicrons accumulate due to saturation of clearance mechanisms. Therefore, strategies to prevent CVD need to be undertaken once the TGs have been lowered to a level where pancreatitis is unlikely to recur. 

 

Pancreatitis

 

Severe hypertriglyceridemia is the third most common cause of acute pancreatitis after alcohol and gallstones. The chylomicronemia syndrome describes a constellation of findings that occur with severe elevations of plasma TG levels. There is some lack of consensus as to what constitutes severe HTG, values >1000-1500 mg/dL are generally classified as severe, although some groups consider values in the 500-1000 mg/dL range also severe hypertriglyceridemia (187). 

 

Individuals with both FCS and MFCS often present with hypertriglyceridemia induced acute pancreatitis, which can be recurrent if triglyceride levels remain elevated persistently. Women with genetic HTG can develop severe HTG and pancreatitis during pregnancy particularly during the third trimester (188).

 

The pancreatitis that occurs with severe HTG can be recurrent.  In a prospective study of patients admitted with acute pancreatitis, the distribution of plasma TGs was bimodal when measured at the peak of the pain (101, 102).  TG levels <880 mg/dL were associated with gall bladder disease and chronic alcoholism, while those above 2000 mg/dL were associated with the simultaneous presence of familial and secondary forms of HTG.  It has been suggested that individuals become prone to the development of TG-induced pancreatitis at TG values between 1500-2000 mg/dL (189).  TG-induced pancreatitis has been reported with TG levels lower than 500 mg/dL(190, 191),  although in our experience this usually occurs when patients with severe HTG stopped eating some time prior to the blood draw. The frequency of severe HTG leading to acute pancreatitis varies widely from about 6-20% of subjects, possibly related to the type of patient presenting to different type of medical centers (192, 193).  Pancreatitis often is recurrent if HTG is not appreciated to be the cause and if TG levels are not adequately controlled (87). With long term multiple episodes of acute, recurrent pancreatitis, exocrine pancreatic insufficiency or insulin deficient secondary diabetes may occur. A meta-analysis of observational studies suggests that TG-induced pancreatitis has worse outcomes than pancreatitis from other causes, with an approximate doubling of renal and respiratory failure, a nearly 4-fold increase of shock and a near doubling of mortality (194). Pancreatitis due to very severe HTG also may occur during infusion of lipid emulsions for parenteral feeding (195) or with use of the anesthetic agent propofol, which is infused in a 10% fat emulsion (196). 

 

MECHANISM OF SEVERE HYPERTRIGLYCERIDEMIC PANCREATITIS

 

The mechanism by which very severe HTG leads to pancreatitis remains speculative. Suggested mechanisms include the local liberation of FFA from TGs and lysophosphatidylcholine from phosphatidycholine when pancreatic lipase encounters very high levels of TG-rich lipoproteins in the pancreatic capillaries (197). High local concentrations of FFA overwhelm the binding capacity of albumin with resultant aggregation into micellar structures with detergent properties.  Both FFA and lysophosphatidylcholine have been shown to cause chemical pancreatitis when infused into pancreatic arteries in animal models (198-200). This leads to local liberation of more lipases from the damaged pancreatic acini, resulting in a vicious cycle (198, 201).  It also has been hypothesized that increased plasma viscosity due to the presence of increased numbers of chylomicrons in the pancreatic microcirculation contributes to the development of pancreatitis (202). There also is recent evidence of gene associations in TG-induced pancreatitis; in a Chinese cohort with HTG, a CFTR variant and TNF alpha promoter polymorphism were found to be independent risk factors for developing pancreatitis (203), while another study found an increased frequency of ApoE4 (204).

 

DIAGNOSIS OF SEVERE HYPERTRIGLYCERIDEMIC INDUCED PANCREATITIS

 

The diagnosis of HTG-associated pancreatitis can be made by the presence of severely elevated TG levels in a patient with acute pancreatitis. Falsely low serum amylase levels can be encountered due to assay interference by the TG-rich lipoproteins (205). Pseudohyponatremia due to the presence of large numbers of TG-rich lipoproteins in plasma can be seen with very high TG levels. Interference with liver transaminase assays may also occur, giving spuriously high values making it difficult to exclude alcoholic liver disease (205).

 

With chronic chylomicronemia, patients may develop eruptive xanthomata (Figure 2). These xanthomas represent an inflammatory response to the deposition of chylomicron-associated lipids in tissues and are yellow-red papules that usually appear on the buttocks, back and extensor surfaces of the upper limbs. Histologically, these lesions contain lipid laden foamy macrophages (206).  

 

Figure 2. Eruptive Xanthomas. The commonest site is on the buttocks. The lesions are papular with an erythematous base. They often are itchy.

 

Lipemia retinalis, where the retinal vessels take on a whitish hue with pallor of the optic fundus and retina can be observed with very high TG levels (Figure 3).  There is no associated visual impairment.  

Figure 3. Lipemia retinalis. Note the pale color of the retinal vessels.

 

Acute recent memory loss and mental fogginess (82) can also occasionally be seen, but has not been extensively studied. Symptoms such as fatigue, blurred vision, dysesthesias, and transient ischemic attacks have been suggested to be related to hyperviscosity resulting from high TG levels (207, 208).  Hepatosplenomegaly is frequently present in FCS due to macrophage infiltration in response to the chylomicron accumulation. Fatty liver is a common finding on imaging in both FCS and MFCS.

 

MANAGEMENT OF SEVERE HYPERTRIGLYCERIDEMIA

 

Management of HTG by lifestyle and pharmacological means is discussed in detail in the Endotext chapters on The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels and Triglyceride Lowering Drugs (209, 210).  However, in this section we will make a few points specifically relevant to this chapter. 

 

Before initiating lifelong therapy for hypertriglyceridemia, evaluation for and treatment of reversible secondary disorders that can elevate plasma triglyceride levels is crucial. This includes appropriate management of diabetes and hypothyroidism and substituting drugs that can elevate triglyceride levels with lipid-neutral agents. Management of hypertension should include calcium channel blockers, angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and alpha-adrenergic blockers rather than beta-adrenergic blocking agents and diuretics.

 

Cardiovascular Disease Prevention

 

ASCVD risk in HTG is modulated by the presence of several other factors, including other lipoprotein abnormalities, other CVD risk factors, and family history of CVD, with some families with HTG appearing to have a greater risk of CVD than others (44). The role of TG lowering by pharmacological means remains controversial, but there is consensus that the presence of HTG imparts residual risk after LDL has been adequately lowered with statins.

 

Statins: The best clinical trial data currently available for the prevention of ASCVD in patients with HTG demonstrate that statins are likely to confer the most benefit, even though their primary mode of action is not to reduce plasma TGs, nor are they very effective in so doing (211).  In patients with elevated TG levels statins will result in a significant decrease in TG levels. Based on the results of the IMPROVE-IT trial (212), the addition of ezetimibe may be of additional benefit.

 

Fibrates: Fibrates such as gemfibrozil and fenofibrate, are PPAR-α agonists, and very effective in lowering plasma TG levels (by up to 50%).  Several studies have failed to demonstrate a benefit of fibrates on ASCVD events, either alone or in combination with statins.  However, participants in these studies were not confined to individuals with HTG.  Nonetheless, post-hoc analysis showed that subgroups of subjects who had mild HTG >200mg/dL and LDL-C <34mg/dL had a significant reduction of ASCVD events (213-216).  In addition, the Action to Control Cardiovascular Risk in Diabetes (ACCORD)-LIPID trial, which was confined to subjects with diabetes, showed a similar outcome in the subgroups with HTG, although the trial was negative for all subjects (214).  Recently a novel selective peroxisome proliferator-activated receptor α modulator, pemafibrate, that possesses unique PPARα activity and selectivity (217), was evaluated in individuals with HTG and diabetes in the Pemafibrate to Reduce Cardiovascular OutcoMes by Reducing Triglycerides IN patiENts With diabetes (PROMINENT) trial. Patients with type 2 diabetes, triglyceride level 200 to 499 mg/dL and HDL-C of </=40 mg/dL were assigned to pemafibrate or placebo. Unfortunately, pemafibrate failed to demonstrate cardiovascular benefit in patients with type 2 diabetes and mild to moderate HTG despite significantly lowering triglyceride levels (218).  Thus, addition of a fibrate to a statin for cardiovascular risk reduction cannot be recommended at this time.

 

Omega-3 fish oil: Omega-3 (n-3) fatty acids are polyunsaturated fatty acids that lower TGs. The two main n-3 fatty acids are eicosapentanoic acid (EPA) and docosahexanoic acid (DHA) which can lower VLDL secretion and are agonists of PPARa. However, their role in ASCVD prevention also has been controversial as several RCTs of various dosages of n-3 mixtures failed to demonstrate CV benefit in mild to moderate HTG (219). The Reduction of Cardiovascular Events with Icosapent Ethyl–Intervention Trial (REDUCE-IT), evaluated the addition of high dose icosapent ethyl (highly purified eicosapentanoic acid or EPA) compared to placebo in high-risk patients with mild to moderate HTG on statin therapy, demonstrated a surprising 25% lower CV risk in subjects. Notably this effect was independent of baseline TG levels and TG reduction.  (220).  Subsequently the STRENGTH (Statin Residual Risk Reduction With Epanova in High Cardiovascular Risk Patients with Hypertriglyceridemia) trial of n–3 fatty acid mixtures (EPA + DHA) failed to demonstrate cardiovascular benefit and was terminated early for futility (221). Similarly, use of an EPA/DHA mixture (1.8 g/d) in patients with a recent myocardial infarction in the OMega-3 fatty acids in Elderly with Myocardial Infarction (OMEMI) trial (222) also failed to meet its primary endpoint. Tissue EPA levels may be a contributor to the positive results in the REDUCE-IT study, as there is evidence that EPA inhibits inflammation, causes membrane stabilization, and decreases plaque volume. The REDUCE-IT trial has generated controversy due to use of mineral oil in the control group which resulted in an increase in both LDL-C and C-reactive protein (223, 224). Nonetheless, several current guidelines suggest addition of icosapent ethyl in addition to a statin for residual hypertriglyceridemia in high-risk individuals (those with known ASCVD or diabetes with additional risk factors).

.

Niacin: Niacin effectively lowers triglycerides and LDL-C while raising HDL-C. Niacin inhibits lipolysis in adipocytes, therefore decreasing the available fatty acids for VLDL synthesis. However, it has fallen out of favor for ASCVD risk reduction. Two RCTs, Atherothrombosis Intervention in Metabolic Syndrome with Low HDL/High Triglycerides: Impact on Global Health Outcomes (AIM-HIGH) and Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE) demonstrated no benefit to the addition of niacin to statins for decreasing cardiovascular risk. However, it is important to note that neither of these trials were confined to subjects with high TG levels. Therefore, due to the lack of efficacy and potential for increasing insulin resistance, niacin is not recommended for treatment of HTG.

 

Newer Therapies for HTG

 

Apo C-III Inhibitors: ApoC-III is an endogenous inhibitor of LPL, by displacing ApoC-II an activator of LPL. This leads to inhibition of lipolysis and elevations in TG levels. Apo C-III can also increase TGs by LPL independent mechanisms. Currently, ApoC-III inhibitors include volanesorsen (a second-generation antisense oligonucleotide (ASO)), olezarsen (a third generation ASO), and ARO-APOC3 (a small interfering RNA), all of which are directed at APOC3 gene (225). Volanesorsen has been studied in individuals with FCS and MFCS and has been found to decrease TG levels by up to 70% from baseline and potentially prevent pancreatitis. However, thrombocytopenia and injection site reactions are common. This drug is therefore not approved for use by the FDA due to concerns of bleeding but is approved for use by the European Medicines Agency.  

Olezarsen is a third generation ASO for which early studies have been completed.

 

ANGPTL-3 inhibitors: ANGPTL3 is a key regulator of lipoprotein metabolism and is able to repress LPL and endothelial lipase activity, with resultant increase in TGs and TG rich lipoprotein levels (226). Homozygous loss of function variants in ANGPTL3 result in combined hypolipidemia with low TG and LDL-C levels. ANGPTL3 inhibition results in decreased LDL-C, and TGs. ANGPTL3 inhibitors include evinacumab, a monoclonal antibody, vupanorsen, an antisense oligonucleotide (ASO), and ARO-ANG3, a small-interfering ribonucleic acid (siRNA). Evinacumab has been studied in patients with sHTG. A phase 2 study of evinacumab in patients with sHTG, demonstrated significant reductions in TGs only in patients with MFCS with and without LPL mutations, but not in patients with FCS, suggesting that ANGPTL3 inhibition is dependent on presence of some LPL activity (227). In the cohort with polygenic sHTG and MCS, evinacumab 15 mg/kg IV every 4 weeks resulted in an 81.7% reduction in TGs. It should be noted that evinacumab is currently only approved for homozygous familial hypercholesterolemia.

 

Management of Severe HTG-Induced Pancreatitis

 

Because of the low frequency of severe HTG in the general population, and because only some patients with severe HTG develop pancreatitis, large random controlled clinical trials are difficult to perform and unlikely to be undertaken in the foreseeable future. Therefore, therapeutic decisions need to be based on less stringent criteria than might otherwise be desirable.  However, keeping TG levels <500 mg/dL should prevent the onset of TG-induced pancreatitis (187, 228, 229). 

 

ACUTE MANAGEMENT

 

The clinical presentation of HTG-induced pancreatitis is similar to that from other causes of acute pancreatitis and can be preceded by episodic nausea, epigastric pain radiating through to the back, and increasing heart-burn. Individuals with recurrent acute pancreatitis may present without severe elevations in pancreatic enzymes (230). The immediate goal is to lower TGs in hospitalized patients.  Management is similar to the management of non-TG induced pancreatitis, which includes cessation of all oral intake for pancreatic rest, fluid resuscitation, pain management, and management of metabolic abnormalities. TGs fall rapidly with discontinuation of oral intake, often to under 1000mg/dL with cessation of oral intake. With clinical improvement, oral diet advancement should be done slowly and cautiously in the hospital as this can result in rebound TG elevations. Supportive care as needed should be instituted for organ failure. Lipid emulsions for parenteral feeding should be avoided since their use will further delay clearance and exacerbate the HTG. If long term nutrition is required for very ill individuals who cannot eat, total parental nutrition without lipids should be utilized.   

 

Heparin: Heparin will liberate LPL into plasma from its endothelial binding sites and hence rapidly lowers TGs (231). However, it also can cause rebound HTG due to rapid degradation of released LPL (232) and increases the risk of hemorrhagic pancreatitis. Therefore, the use of heparin is not recommended (233). 

 

Insulin: The rationale for the use of an IV insulin infusion of regular insulin (in conjunction with IV glucose administration as needed) is that it can activate LPL and enhance clearance of TG-rich lipoproteins (234). Intravenous insulin can be beneficial in individuals with diabetes needing glycemic control. Its use in TG--induced pancreatitis without diabetes has been reported in several case reports (235-239), and has become widespread but it is unclear whether similar changes would have occurred simply by restricting oral intake without the use of insulin.  Regular insulin at 0.1-0.2 units/kg/hour with a separate iv dextrose infusion to prevent hypoglycemia in individuals without diabetes is often used. IV insulin can be stopped when TG drops to below 1000mg/dL. However, TGs will increase when the individual consumes an oral diet; therefore, caution should be exercised with slow advancement of the diet. In a study of chylomicronemia with uncontrolled diabetes, insulin infusion lowered TGs more rapidly than plasmapheresis (240).

 

Plasmapheresis: The use of plasmapheresis to acutely lower TGs is controversial. Plasmapheresis is extracorporeal therapy where plasma is removed and replaced; plasma is separated from the blood and discarded removing chylomicrons. Substitute fluid is replaced to maintain blood volume. The procedure is highly effective in rapidly decreasing TG levels by 85% after 1 session. However, the effect is not persistent, without evidence of long-term efficacy or mortality and morbidity benefit demonstrated. Although recommended by some (241, 242), the current evidence for the benefit of use of plasmapheresis is limited to small uncontrolled anecdotal series (243) from which no firm conclusion can be made regarding its use in acute TG-induced pancreatitis (244). A recent retrospective analysis demonstrated no benefit on length of hospital stay or mortality when therapeutic plasma exchange was added to medical management of severely elevated plasma TGs (245). TG levels fall rapidly with cessation of oral intake and use of non-lipid-containing intravenous fluids.  Plasmapheresis requires a specialized center, needs central venous access, and transient anticoagulation; it only temporarily improves TG levels without addressing the underlying cause (83). Risks include line sepsis, deep vein thrombosis, and bleeding. Therefore, we do not recommend its routine use in this situation unless clinical circumstances necessitate plasmapheresis such as severe acute necrotizing pancreatitis (246), shock, or pregnancy (247). 

 

LONG-TERM MANAGEMENT TO PREVENT PANCREATITIS

 

After TG lowering in the setting of acute pancreatitis, it is essential to determine both the primary and secondary causes of the severe HTG that precipitated the acute pancreatitis.   Continued management of any secondary form of HTG, as well as lifestyle and drug therapy to maintain low TG levels is required to prevent recurrent pancreatitis. If fasting plasma TG levels remain above 1000 mg/dL after treating or removing the precipitating causes of the severe HTG, life-long therapy with fibrates or n-3 fatty acids, as described earlier, might be considered for these patients. Limited evidence suggests that orlistat, a gastrointestinal lipase inhibitor that decreases absorption of ingested fat, thereby reducing intestinal chylomicron synthesis, may be of benefit in reducing TG levels when used in conjunction with fibrate therapy (248, 249). TG and glucose control can be particularly challenging in individuals with familial partial lipodystrophy.

 

Management of Specific Syndromes that Accompany Severe HTG

 

FAMILIAL CHYLOMICRONEMIA SYNDROME (FCS)

 

Treatment of FCS includes management of an acute crisis (pancreatitis) and long-term management of HTG. Management of acute HTG-induced pancreatitis is described in the previous section. Long-term management of individuals with FCS involves patient education and maintaining a very low-fat diet. Consumption of even small amounts of fat can lead to severe HTG in FCS due to the absence of functional LPL. Infants with FCS presenting with abdominal pain or failure to thrive require discontinuation of breast feeding with replacement by very low-fat formula feeding to decrease TG levels and symptoms. In children and adults with FCS, dietary fat calories should be severely restricted to control the severe HTG and abdominal pain. This translates to about 5% to 10% of total daily calories from fat, which is a major burden for these patients (250).  Medium-chain TGs, which are taken up directly by the liver after absorption and do not enter plasma as chylomicrons via the thoracic duct, are a potential alternate fat source for these patients. n-3 fatty acids can aggravate the severe HTG of FCS and therefore are contraindicated in these individuals (251, 252).  Fibrates are not efficacious in FCS (253).  There are limited studies showing that orlistat might be beneficial in patients with FCS (254, 255).  Alcohol, oral contraceptives, and other TG-elevating drugs (see Table 3) can exacerbate severe HTG and precipitate acute pancreatitis in FCS.  Successful pregnancies in patients with FCS have become more common of late (256, 257).

 

Alipogene tiparvovec, an adeno-associated virus LPL gene therapy that was developed and resulted in significant improvement in postprandial chylomicron metabolism in patients with FCS has been abandoned and no longer available (258), Antisense oligonucleotide inhibitors of ApoC-III (volanesorsen is approved in Europe but no in the US) and of ANGPLT3 are in development for FCS (96).

 

MULTIFACTORIAL CHYLOMICRONEMIA SYNDROME (MFCS)

 

Management of acute HTG-induced pancreatitis is described in the previous section.

 

Long term management: To prevent acute HTG-induced pancreatitis in MFCS, the goal is to maintain TG levels below the threshold for pancreatitis, preferably <500 mg/dL. This requires instituting lifestyle adjustments, reversal of any secondary causes of HTG, such as treatment of suboptimally managed or undiagnosed diabetes, treating hypertension with lipid neutral agents such as ACE inhibitors, ARBs, calcium channel inhibitors, or alpha blockers rather than beta-adrenergic blockers and diuretics, and discontinuing other TG-raising drugs (table 3) where possible.  Alcohol intake should be limited or eliminated, since even small amounts of alcohol can substantially raise TG levels in individuals with baseline HTG.  Attention should be paid to avoid rebound weight gain that commonly occurs after successful weight loss. Oral estrogens should be substituted by transdermal or vaginal preparations, which raise plasma TGs to a lesser extent than oral estrogens (259, 260). Residual HTG should be treated with fibrates (229), which together with management of the secondary disorder or disorders, can reduce TG levels to below the threshold for developing pancreatitis. Other agents that can be used to lower TGs alone or in combination with fibrates, include n-3 fatty acids, and high-dose statins. We do not recommend using niacin due to risk of worsening insulin resistance and lack of clinical trial data for benefit. Lifestyle measures and weight loss are important, but patients should be educated on risks of rapid weight regain after successful weight loss can be associated with rebound severe HTG. Bariatric surgery also has been used to reduce severe HTG in refractory HTG (261). Inhibition of ApoC-III or ANGPTL3, which have been shown to lower TGs in patients with severe HTG (262), may have a role to play in the future the management of severe HTG in patients with MFCS. 

 

REFERENCES

 

  1. Feingold, K.R., Introduction to Lipids and Lipoproteins, in Endotext, K.R. Feingold, et al., Editors. 2021: South Dartmouth (MA).
  2. Fisher, E.A., The degradation of Apolipoprotein B100: multiple opportunities to regulate VLDL triglyceride production by different proteolytic pathways. Biochim Biophys Acta, 2012. 1821(5): p. 778-81.
  3. Sundaram, M. and Z. Yao, Recent progress in understanding protein and lipid factors affecting hepatic VLDL assembly and secretion. Nutr Metab (Lond), 2010. 7: p. 35.
  4. Yao, Z., Human Apolipoprotein C-III - a new intrahepatic protein factor promoting assembly and secretion of very low density lipoproteins. Cardiovasc Hematol Disord Drug Targets, 2012. 12(2): p. 133-40.
  5. Schneeman, B.O., et al., Relationships between the responses of triglyceride-rich lipoproteins in blood plasma containing Apolipoproteins B-48 and B-100 to a fat-containing meal in normolipidemic humans. Proc Natl Acad Sci U S A, 1993. 90(5): p. 2069-73.
  6. Kindel, T., D.M. Lee, and P. Tso, The mechanism of the formation and secretion of chylomicrons. Atheroscler Suppl, 2010. 11(1): p. 11-6.
  7. Kersten, S., Physiological regulation of lipoprotein lipase. Biochim Biophys Acta, 2014. 1841(7): p. 919-33.
  8. Gordts, P.L., et al., ApoC-III inhibits clearance of triglyceride-rich lipoproteins through LDL family receptors. J Clin Invest, 2016. 126(8): p. 2855-66.
  9. Goldberg, I.J., et al., Lipoprotein ApoC-II activation of lipoprotein lipase. Modulation by Apolipoprotein A-IV. J Biol Chem, 1990. 265(8): p. 4266-72.
  10. Nilsson, S.K., et al., Apolipoprotein A-V; a potent triglyceride reducer. Atherosclerosis, 2011. 219(1): p. 15-21.
  11. Priore Oliva, C., et al., Inherited Apolipoprotein A-V deficiency in severe hypertriglyceridemia. Arterioscler Thromb Vasc Biol, 2005. 25(2): p. 411-7.
  12. Gonzales, J.C., et al., Apolipoproteins E and AV mediate lipoprotein clearance by hepatic proteoglycans. J Clin Invest, 2013. 123(6): p. 2742-51.
  13. Kroupa, O., et al., Linking nutritional regulation of Angptl4, Gpihbp1, and Lmf1 to lipoprotein lipase activity in rodent adipose tissue. BMC Physiol, 2012. 12: p. 13.
  14. Lamiquiz-Moneo, I., et al., (Identification of variants in LMF1 gene associated with primary hypertriglyceridemia). Clin Investig Arterioscler, 2015.
  15. Inukai, K., et al., ANGPTL3 is increased in both insulin-deficient and -resistant diabetic states. Biochem Biophys Res Commun, 2004. 317(4): p. 1075-9.
  16. Shimamura, M., et al., Leptin and insulin down-regulate angiopoietin-like protein 3, a plasma triglyceride-increasing factor. Biochem Biophys Res Commun, 2004. 322(3): p. 1080-5.
  17. Koster, A., et al., Transgenic angiopoietin-like (angptl)4 overexpression and targeted disruption of angptl4 and angptl3: regulation of triglyceride metabolism. Endocrinology, 2005. 146(11): p. 4943-50.
  18. Foley, E.M., et al., Hepatic remnant lipoprotein clearance by heparan sulfate proteoglycans and low-density lipoprotein receptors depend on dietary conditions in mice. Arterioscler Thromb Vasc Biol, 2013. 33(9): p. 2065-74.
  19. Crawford, S.E. and J. Borensztajn, Plasma clearance and liver uptake of chylomicron remnants generated by hepatic lipase lipolysis: evidence for a lactoferrin-sensitive and Apolipoprotein E-independent pathway. J Lipid Res, 1999. 40(5): p. 797-805.
  20. Dichek, H.L., et al., Hepatic lipase overexpression lowers remnant and LDL levels by a noncatalytic mechanism in LDL receptor-deficient mice. J Lipid Res, 2001. 42(2): p. 201-10.
  21. Brunzell, J.D., et al., Evidence for a common saturable triglyceride removal mechanism for chylomicrons and very low density lipoproteins in man. Journal of Clinical Investigation, 1973. 52: p. 1578-1585.
  22. Hegele, R.A., et al., The polygenic nature of hypertriglyceridaemia: implications for definition, diagnosis, and management. Lancet Diabetes Endocrinol, 2014. 2(8): p. 655-66.
  23. Berglund, L., et al., Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2012. 97(9): p. 2969-89.
  24. Subramanian, S., Approach to the Patient With Moderate Hypertriglyceridemia. J Clin Endocrinol Metab, 2022. 107(6): p. 1686-1697.
  25. Dron, J.S. and R.A. Hegele, Genetics of Hypertriglyceridemia. Front Endocrinol (Lausanne), 2020. 11: p. 455.
  26. Noubiap, J.J., et al., Prevalence of dyslipidaemia among adults in Africa: a systematic review and meta-analysis. Lancet Glob Health, 2018. 6(9): p. e998-e1007.
  27. Laufs, U., et al., Clinical review on triglycerides. Eur Heart J, 2020. 41(1): p. 99-109c.
  28. Pedersen, S.B., A. Langsted, and B.G. Nordestgaard, Nonfasting Mild-to-Moderate Hypertriglyceridemia and Risk of Acute Pancreatitis. JAMA Intern Med, 2016. 176(12): p. 1834-1842.
  29. Grundy, S.M., et al., 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation, 2019. 139(25): p. e1082-e1143.
  30. Pearson, G.J., et al., 2021 Canadian Cardiovascular Society Guidelines for the Management of Dyslipidemia for the Prevention of Cardiovascular Disease in Adults. Can J Cardiol, 2021. 37(8): p. 1129-1150.
  31. Mach, F., et al., 2019 ESC/EAS Guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J, 2020. 41(1): p. 111-188.
  32. Expert Panel on Detection, E. and A. Treatment of High Blood Cholesterol in, Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III). JAMA, 2001. 285(19): p. 2486-97.
  33. Miller, M., et al., Triglycerides and cardiovascular disease: a scientific statement from the American Heart Association. Circulation, 2011. 123(20): p. 2292-333.
  34. Jacobson, T.A., et al., National lipid association recommendations for patient-centered management of dyslipidemia: part 1--full report. J Clin Lipidol, 2015. 9(2): p. 129-69.
  35. Berglund, L., et al., Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab, 2012. 97(9): p. 2969-89.
  36. Authors/Task Force, M., et al., 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias: The Task Force for the Management of Dyslipidaemias of the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS) Developed with the special contribution of the European Assocciation for Cardiovascular Prevention & Rehabilitation (EACPR). Atherosclerosis, 2016. 253: p. 281-344.
  37. Fredrickson, D., R. Levy, and R. Lees, Fat transport and lipoproteins - an integrated approach to mechanisms and disorders. N Engl J Med, 1967. 276: p. 32,94,148,215,273.
  38. Goldstein, J.L., et al., Hyperlipidemia in coronary heart disease. II. Genetic analysis of lipid levels in 176 families and delineation of a new inherited disorder, combined hyperlipidemia. J Clin Invest, 1973. 52(7): p. 1544-68.
  39. Nikkila, E.A. and A. Aro, Family study of serum lipids and lipoproteins in coronary heart-disease. Lancet, 1973. 1(7810): p. 954-9.
  40. Brunzell, J.D., Clinical practice. Hypertriglyceridemia. N Engl J Med, 2007. 357(10): p. 1009-17.
  41. Chait, A., J.J. Albers, and J.D. Brunzell, Very low density lipoprotein overproduction in genetic forms of hypertriglyceridemia. European Journal of Clinical Investigation, 1980. 10: p. 17-22.
  42. Brunzell, J.D., et al., Plasma lipoproteins in familial combined hyperlipidemia and monogenic familial hypertriglyceridemia. Journal of Lipid Research, 1983. 24: p. 147-155.
  43. Brunzell, J.D., et al., Myocardial infarction in the familial forms of hypertriglyceridemia. Metabolism: Clinical and Experimental, 1976. 25: p. 313-320.
  44. Austin, M.A., et al., Cardiovascular disease mortality in familial forms of hypertriglyceridemia: A 20-year prospective study. Circulation, 2000. 101: p. 2777-2782.
  45. Hokanson, J.E. and M.A. Austin, Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta- analysis of population-based prospective studies. J Cardiovasc Risk, 1996. 3(2): p. 213-9.
  46. McNeely, M., et al., Lipoprotein and Apolipoprotein abnormalities in familial combined hyperlipidemia: a 20-year prospective study. Atherosclerosis, 2001. 159: p. 417-481.
  47. Lewis, G.F., C. Xiao, and R.A. Hegele, Hypertriglyceridemia in the genomic era: a new paradigm. Endocr Rev, 2015. 36(1): p. 131-47.
  48. Dron, J.S., et al., Severe hypertriglyceridemia is primarily polygenic. J Clin Lipidol, 2019. 13(1): p. 80-88.
  49. Eaton, R.P., R.C. Allen, and D.S. Schade, Overproduction of a kinetic subclass of VLDL-ApoB, and direct catabolism of VLDL-ApoB in human endogenous hypertriglyceridemia: an analytical model solution of tracer data. J Lipid Res, 1983. 24(10): p. 1291-303.
  50. Grundy, S.M., et al., Transport of very low density lipoprotein triglycerides in varying degrees of obesity and hypertriglyceridemia. Journal of Clinical Investigation, 1979. 63: p. 1274-1283.
  51. Beil, U., et al., Triglyceride and cholesterol metabolism in primary hypertriglyceridemia. Arteriosclerosis, 1982. 2(1): p. 44-57.
  52. Sigurdsson, G., A. Nicoll, and B. Lewis, Metabolism of very low density lipoproteins in hyperlipidaemia: studies of Apolipoprotein B kinetics in man. Eur J Clin Invest, 1976. 6(2): p. 167-77.
  53. Ayyobi, A.F. and J.D. Brunzell, Lipoprotein distribution in the metabolic syndrome, type 2 diabetes mellitus, and familial combined hyperlipidemia. Am J Cardiol, 2003. 92(4A): p. 27J-33J.
  54. Reynisdottir, S., et al., Impaired activation of adipocyte lipolysis in familial combined hyperlipidemia. J Clin Invest, 1995. 95(5): p. 2161-9.
  55. Reynisdottir, S., et al., Adipose tissue lipoprotein lipase and hormone-sensitive lipase. Contrasting findings in familial combined hyperlipidemia and insulin resistance syndrome. Arterioscler Thromb Vasc Biol, 1997. 17(10): p. 2287-92.
  56. Venkatesan, S., et al., Stable isotopes show a direct relation between VLDL ApoB overproduction and serum triglyceride levels and indicate a metabolically and biochemically coherent basis for familial combined hyperlipidemia. Arteriosclerosis and Thrombosis, 1993. 13: p. 1110-1118.
  57. Aitman, T., et al., Defects of insulin action on fatty acid and carbohydrate metabolism in familial combined hyperlipidemia. Arterioscler Thromb Vasc Biol, 1997. 17: p. 748-754.
  58. Janus, E.D., et al., Kinetic bases of the primary hyperlipidemias:Studies of Apolipoprotein B turnover in genetically defined subjects. European Journal of Clinical Investigation, 1980. 10: p. 161-172.
  59. Berneis, K.K. and R.M. Krauss, Metabolic origins and clinical significance of LDL heterogeneity. J Lipid Res, 2002. 43(9): p. 1363-79.
  60. Cabezas, M.C., et al., Impaired chylomicron remnant clearance in familial combined hyperlipidemia. Arterioscler Thromb, 1993. 13(6): p. 804-14.
  61. Hokanson, J.E., et al., Plasma triglyceride and LDL heterogeneity in familial combined hyperlipidemia. Arteriosclerosis and Thrombosis, 1993. 13: p. 427-434.
  62. Hokanson, J.E., et al., LDL physical and chemical properties in familial combined hyperlipidemia. Arteriosclerosis, Thrombosis, and Vascular Biology, 1995. 15: p. 452-459.
  63. Cruz-Bautista, I., et al., Determinants of VLDL composition and Apo B-containing particles in familial combined hyperlipidemia. Clin Chim Acta, 2015. 438: p. 160-5.
  64. Brouwers, M.C. and M.M. van Greevenbroek, Lipid metabolism: the significance of plasma proprotein convertase subtilisin kexin type 9 in the elucidation of complex lipid disorders. Curr Opin Lipidol, 2011. 22(4): p. 317-8.
  65. Hopkins, P.N., et al., Coronary artery disease risk in familial combined hyperlipidemia and familial hypertriglyceridemia: a case-control comparison from the National Heart, Lung, and Blood Institute Family Heart Study. Circulation, 2003. 108(5): p. 519-23.
  66. Purnell, J.Q., et al., Relationship of insulin sensitivity and ApoB levels to intra-abdominal fat in subjects with familial combined hyperlipidemia. Arterioscler Thromb Vasc Biol, 2001. 21(4): p. 567-72.
  67. Ascaso, J., et al., Insulin resistance in patients wtih familial combined hyperlipidemia and coronary artery disease. Am J Cardiol, 1997. 80: p. 1481-1487.
  68. Castro Cabezas, M., et al., Impaired fatty acid metabolism in familial combined hyperlipidemia: a mechanism associating hepatic Apolipoprotein B overproduction and insulin resistance. J Clin Invest, 1993. 92: p. 160-168.
  69. van der Kallen, C., et al., Evidence of insulin resistant lipid metabolism in adipose tissue in familial combined hyperlipidemia, but not type 2 diabetes mellitus. Atherosclerosis, 2002. 164: p. 337-346.
  70. Arner, P., et al., Dynamics of human adipose lipid turnover in health and metabolic disease. Nature, 2011. 478(7367): p. 110-3.
  71. Brouwers, M.C., et al., Fatty liver is an integral feature of familial combined hyperlipidaemia: relationship with fat distribution and plasma lipids. Clin Sci (Lond), 2007. 112(2): p. 123-30.
  72. Brouwers, M.C., et al., Increased arterial stiffness in familial combined hyperlipidemia. J Hypertens, 2009. 27(5): p. 1009-16.
  73. Keulen, E.T., et al., Increased intima-media thickness in familial combined hyperlipidemia associated with Apolipoprotein B. Arterioscler Thromb Vasc Biol, 2002. 22(2): p. 283-8.
  74. Chait, A. and J.D. Brunzell, Severe hypertriglyceridemia:Role of familial and acquired disorders. Metabolism: Clinical and Experimental, 1983. 32: p. 209-214.
  75. Herink, M. and M.K. Ito, Medication Induced Changes in Lipid and Lipoproteins, in Endotext, K.R. Feingold, et al., Editors. 2018: South Dartmouth (MA).
  76. Feingold, K.R., The Effect of Inflammation and Infection on Lipids and Lipoproteins, in Endotext, K.R., Feingold, et al., Editors. 2022: South Dartmouth (MA).
  77. Feingold, K.R., Obesity and Dyslipidemia, in Endotext, K.R. Feingold, et al., Editors. 2023: South Dartmouth (MA).
  78. Feingold, K.R., The Effect of Endocrine Disorders on Lipids and Lipoproteins, in Endotext, K.R. Feingold, et al., Editors. 2023: South Dartmouth (MA).
  79. Chait, A., Secondary hyperlipidemia. Journal of Clinical Pathology, 1973. 26(suppl 5): p. 68-71.
  80. Chait, A. and J.D. Brunzell, Acquired hyperlipidemia (secondary dyslipoproteinemia). Endocrinology and Metabolism Clinics of North America, 1990. 19: p. 259-278.
  81. Brunzell, J.D. and E.L. Bierman, Chylomicronemia syndrome.Interaction of genetic and acquired hypertriglyceridemia. Medical Clinics of North America, 1982. 66: p. 455-468.
  82. Chait, A., H.T. Robertson, and J.D. Brunzell, Chylomicronemia syndrome in diabetes mellitus. Diabetes Care, 1981. 4: p. 343-348.
  83. Brahm, A.J. and R.A. Hegele, Chylomicronaemia-current diagnosis and future therapies. Nat Rev Endocrinol, 2015.
  84. Brunzell, J. and S. Deeb, Familial lipoprotein lipase deficiency, Apo CII deficiency, and hepatic lipase deficiency, in The Metabolic and Molecular Basis of Inherited Disease, C. Scriver, et al., Editors. 2001, McGraw-Hill Book Co.: New York. p. 2789-2816.
  85. Rahalkar, A.R., et al., Novel LPL mutations associated with lipoprotein lipase deficiency: two case reports and a literature review. Can J Physiol Pharmacol, 2009. 87(3): p. 151-60.
  86. Martin-Campos, J.M., et al., Molecular analysis of chylomicronemia in a clinical laboratory setting: diagnosis of 13 cases of lipoprotein lipase deficiency. Clin Chim Acta, 2014. 429: p. 61-8.
  87. Blom, D.J., et al., Characterizing familial chylomicronemia syndrome: Baseline data of the APPROACH study. J Clin Lipidol, 2018. 12(5): p. 1234-1243 e5.
  88. Peterson, J., et al., Structural and functional consequences of missense mutations in exon 5 of the lipoprotein lipase gene. J Lipid Res, 2002. 43(3): p. 398-406.
  89. Nickerson, D.A., et al., DNA sequence diversity in a 9.7-kb region of the human lipoprotein lipase gene. Nat Genet, 1998. 19(3): p. 233-40.
  90. Breckenridge, W.C., et al., Hypertriglyceridemia associated with deficiency of Apolipoprotein C-II. New England Journal of Medicine, 1978. 298: p. 1265.
  91. Rabacchi, C., et al., Spectrum of mutations of the LPL gene identified in Italy in patients with severe hypertriglyceridemia. Atherosclerosis, 2015. 241(1): p. 79-86.
  92. Surendran, R.P., et al., Mutations in LPL, APOC2, APOA5, GPIHBP1 and LMF1 in patients with severe hypertriglyceridaemia. J Intern Med, 2012. 272(2): p. 185-96.
  93. Kristensen, K.K., et al., A disordered acidic domain in GPIHBP1 harboring a sulfated tyrosine regulates lipoprotein lipase. Proc Natl Acad Sci U S A, 2018. 115(26): p. E6020-E6029.
  94. Calandra, S., et al., APOA5 and triglyceride metabolism, lesson from human APOA5 deficiency. Curr Opin Lipidol, 2006. 17(2): p. 122-7.
  95. Peterfy, M., Lipase maturation factor 1: a lipase chaperone involved in lipid metabolism. Biochim Biophys Acta, 2012. 1821(5): p. 790-4.
  96. Baass, A., et al., Familial chylomicronemia syndrome: an under-recognized cause of severe hypertriglyceridaemia. J Intern Med, 2020. 287(4): p. 340-348.
  97. Dron, J.S. and R.A. Hegele, The evolution of genetic-based risk scores for lipids and cardiovascular disease. Curr Opin Lipidol, 2019. 30(2): p. 71-81.
  98. Parsons, S.K., et al., Asparaginase-associated lipid abnormalities in children with acute lymphoblastic leukemia. Blood, 1997. 89(6): p. 1886-95.
  99. Tozuka, M., et al., Characterization of hypertriglyceridemia induced by L-asparaginase therapy for acute lymphoblastic leukemia and malignant lymphoma. Ann Clin Lab Sci, 1997. 27(5): p. 351-7.
  100. Yadav, D. and C.S. Pitchumoni, Issues in hyperlipidemic pancreatitis. J Clin Gastroenterol, 2003. 36(1): p. 54-62.
  101. Brunzell, J.D. and H.G. Schrott, The interaction of familial and secondary causes of hypertriglyceridemia:Role in pancreatitis. Transactions of the Association of American Physicians, 1973. 86: p. 245-254.
  102. Brunzell, J.D. and H.G. Schrott, The interaction of familial and secondary causes of hypertriglyceridemia: role in pancreatitis. J Clin Lipidol, 2012. 6(5): p. 409-12.
  103. Johansen, C.T., et al., An increased burden of common and rare lipid-associated risk alleles contributes to the phenotypic spectrum of hypertriglyceridemia. Arterioscler Thromb Vasc Biol, 2011. 31(8): p. 1916-26.
  104. Schneider, W.J., et al., Familial dysbetalipoproteinemia. Abnormal binding of mutant Apoprotein E to low density lipoprotein receptors of human fibroblasts and membranes from liver and adrenal of rats, rabbits, and cows. J Clin Invest, 1981. 68(4): p. 1075-85.
  105. Vedhachalam, C., et al., The C-terminal lipid-binding domain of Apolipoprotein E is a highly efficient mediator of ABCA1-dependent cholesterol efflux that promotes the assembly of high-density lipoproteins. Biochemistry, 2007. 46(10): p. 2583-93.
  106. Siest, G., et al., Apolipoprotein E: an important gene and protein to follow in laboratory medicine. Clin Chem, 1995. 41(8 Pt 1): p. 1068-86.
  107. Weisgraber, K.H., S.C. Rall, Jr., and R.W. Mahley, Human E Apoprotein heterogeneity. Cysteine-arginine interchanges in the amino acid sequence of the Apo-E isoforms. J Biol Chem, 1981. 256(17): p. 9077-83.
  108. Smelt, A.H. and F. de Beer, Apolipoprotein E and familial dysbetalipoproteinemia: clinical, biochemical, and genetic aspects. Semin Vasc Med, 2004. 4(3): p. 249-57.
  109. Dong, L.M., et al., The carboxyl terminus in Apolipoprotein E2 and the seven amino acid repeat in Apolipoprotein E-Leiden: role in receptor-binding activity. J Lipid Res, 1998. 39(6): p. 1173-80.
  110. Mahley, R.W., Y. Huang, and S.C. Rall, Jr., Pathogenesis of type III hyperlipoproteinemia (dysbetalipoproteinemia). Questions, quandaries, and paradoxes. J Lipid Res, 1999. 40(11): p. 1933-49.
  111. Koopal, C., A.D. Marais, and F.L. Visseren, Familial dysbetalipoproteinemia: an underdiagnosed lipid disorder. Curr Opin Endocrinol Diabetes Obes, 2017. 24(2): p. 133-139.
  112. Koopal, C., et al., Autosomal dominant familial dysbetalipoproteinemia: A pathophysiological framework and practical approach to diagnosis and therapy. J Clin Lipidol, 2017. 11(1): p. 12-23 e1.
  113. Cenarro, A., et al., The p.Leu167del Mutation in APOE Gene Causes Autosomal Dominant Hypercholesterolemia by Down-regulation of LDL Receptor Expression in Hepatocytes. J Clin Endocrinol Metab, 2016. 101(5): p. 2113-21.
  114. Bea, A.M., et al., Contribution of APOE Genetic Variants to Dyslipidemia. Arterioscler Thromb Vasc Biol, 2023. 43(6): p. 1066-1077.
  115. Paquette, M., S. Bernard, and A. Baass, Diagnosis of remnant hyperlipidaemia. Curr Opin Lipidol, 2022. 33(4): p. 227-230.
  116. Pallazola, V.A., et al., Modern prevalence of dysbetalipoproteinemia (Fredrickson-Levy-Lees type III hyperlipoproteinemia). Arch Med Sci, 2020. 16(5): p. 993-1003.
  117. Mahley, R.W. and S.C. Rall, Jr., Type III hyperlipoproteinemia (dysbetalipoproteinemia):The role of Apolipoprotein E in normal and abnormal lipoprotein metabolism, in The Metabolic Basis of Inherited Disease, C.R. Scriver, et al., Editors. 1989, McGraw-Hill: New York. p. 1195.
  118. Brummer, D., et al., Expression of type III hyperlipoproteinemia in patients homozygous for Apolipoprotein E-2 is modulated by lipoprotein lipase and postprandial hyperinsulinemia. J Mol Med (Berl), 1998. 76(5): p. 355-64.
  119. Feussner, G. and R. Ziegler, Expression of type III hyperlipoproteinaemia in a subject with secondary hypothyroidism bearing the Apolipoprotein E2/2 phenotype. J Intern Med, 1991. 230(2): p. 183-6.
  120. Breslow, J.L., et al., Studies of familial type III hyperlipoproteinemia using as a genetic marker the ApoE phenotype E2/2. J Lipid Res, 1982. 23(8): p. 1224-35.
  121. Chait, A., et al., Type-III Hyperlipoproteinaemia ("remnant removal disease"). Insight into the pathogenetic mechanism. Lancet, 1977. 1(8023): p. 1176-8.
  122. Chait, A., et al., Impaired very low density lipoprotein and triglyceride removal in broad beta disease: comparison with endogenous hypertriglyceridemia. Metabolism, 1978. 27(9): p. 1055-66.
  123. Rothschild, M., et al., Pathognomonic Palmar Crease Xanthomas of Apolipoprotein E2 Homozygosity-Familial Dysbetalipoproteinemia. JAMA Dermatol, 2016. 152(11): p. 1275-1276.
  124. Albers, J.J., G.R. Warnick, and W.R. Hazzard, Type III hyperlipoproteinemia: a comparative study of current diagnostic techniques. Clin Chim Acta, 1977. 75(2): p. 193-204.
  125. Blom, D.J., F.H. O'Neill, and A.D. Marais, Screening for dysbetalipoproteinemia by plasma cholesterol and Apolipoprotein B concentrations. Clin Chem, 2005. 51(5): p. 904-7.
  126. Paquette, M., et al., A simplified diagnosis algorithm for dysbetalipoproteinemia. J Clin Lipidol, 2020. 14(4): p. 431-437.
  127. Morganroth, J., R.I. Levy, and D.S. Fredrickson, The biochemical, clinical, and genetic features of type III hyperlipoproteinemia. Ann Intern Med, 1975. 82(2): p. 158-74.
  128. Havel, R.J. and J.P. Kane, Primary dysbetalipoproteinemia:Predominancy of a specific Apoprotein species in triglyceride-rich lipoproteins. Proceedings of the National Academy of Sciences of the USA, 1973. 70: p. 2015.
  129. Koopal, C., et al., Vascular risk factors, vascular disease, lipids and lipid targets in patients with familial dysbetalipoproteinemia: a European cross-sectional study. Atherosclerosis, 2015. 240(1): p. 90-7.
  130. Mahley, R. and S. Rall, Type III Hyperlipoproteinemia (Dysbetalipoproteinemia): The Role of Apolipoprotein E in Normal and Abnormal Lipoprotein Metabolism, in The Metabolic & Molecular Bases of Inherited Disease, C. Scriver, et al., Editors. 2001, McGraw-Hill: New York. p. 2835-2862.
  131. Koopal, C., et al., The relation between Apolipoprotein E (APOE) genotype and peripheral artery disease in patients at high risk for cardiovascular disease. Atherosclerosis, 2016. 246: p. 187-92.
  132. Paquette, M., S. Bernard, and A. Baass, Dysbetalipoproteinemia Is Associated With Increased Risk of Coronary and Peripheral Vascular Disease. J Clin Endocrinol Metab, 2022. 108(1): p. 184-190.
  133. Garg, A., Clinical review#: Lipodystrophies: genetic and acquired body fat disorders. J Clin Endocrinol Metab, 2011. 96(11): p. 3313-25.
  134. Akinci, B., M. Sahinoz, and E. Oral, Lipodystrophy Syndromes: Presentation and Treatment, in Endotext, K.R. Feingold, et al., Editors. 2000: South Dartmouth (MA).
  135. Simha, V. and A. Garg, Inherited lipodystrophies and hypertriglyceridemia. Curr Opin Lipidol, 2009. 20(4): p. 300-8.
  136. Lightbourne, M. and R.J. Brown, Genetics of Lipodystrophy. Endocrinol Metab Clin North Am, 2017. 46(2): p. 539-554.
  137. Lotta, L.A., et al., Integrative genomic analysis implicates limited peripheral adipose storage capacity in the pathogenesis of human insulin resistance. Nat Genet, 2017. 49(1): p. 17-26.
  138. Guillin-Amarelle, C., et al., Type 1 familial partial lipodystrophy: understanding the Kobberling syndrome. Endocrine, 2016. 54(2): p. 411-421.
  139. Agarwal, A.K. and A. Garg, A novel heterozygous mutation in peroxisome proliferator-activated receptor-gamma gene in a patient with familial partial lipodystrophy. J Clin Endocrinol Metab, 2002. 87(1): p. 408-11.
  140. Ajluni, N., et al., Spectrum of disease associated with partial lipodystrophy: lessons from a trial cohort. Clin Endocrinol (Oxf), 2017. 86(5): p. 698-707.
  141. Subramanyam, L., V. Simha, and A. Garg, Overlapping syndrome with familial partial lipodystrophy, Dunnigan variety and cardiomyopathy due to amino-terminal heterozygous missense lamin A/C mutations. Clin Genet, 2010. 78(1): p. 66-73.
  142. Hussain, I. and A. Garg, Lipodystrophy Syndromes. Endocrinol Metab Clin North Am, 2016. 45(4): p. 783-797.
  143. Hussain, I., N. Patni, and A. Garg, Lipodystrophies, dyslipidaemias and atherosclerotic cardiovascular disease. Pathology, 2019. 51(2): p. 202-212.
  144. Jacob, K.N., et al., Phenotypic heterogeneity in body fat distribution in patients with atypical Werner's syndrome due to heterozygous Arg133Leu lamin A/C mutation. J Clin Endocrinol Metab, 2005. 90(12): p. 6699-706.
  145. Garg, A., et al., Atypical progeroid syndrome due to heterozygous missense LMNA mutations. J Clin Endocrinol Metab, 2009. 94(12): p. 4971-83.
  146. Calvo, M. and E. Martinez, Update on metabolic issues in HIV patients. Curr Opin HIV AIDS, 2014. 9(4): p. 332-9.
  147. Castelli, W.P., The triglyceride issue: a view from Framingham. Am Heart J, 1986. 112(2): p. 432-7.
  148. Harchaoui, K.E., et al., Triglycerides and cardiovascular risk. Curr Cardiol Rev, 2009. 5(3): p. 216-22.
  149. Langsted, A., et al., Nonfasting cholesterol and triglycerides and association with risk of myocardial infarction and total mortality: the Copenhagen City Heart Study with 31 years of follow-up. J Intern Med, 2011. 270(1): p. 65-75.
  150. Nordestgaard, B.G. and A. Varbo, Triglycerides and cardiovascular disease. Lancet, 2014. 384(9943): p. 626-35.
  151. Hokanson, J.E. and M.A. Austin, Plasma triglyceride level is a risk factor for cardiovascular disease independent of high-density lipoprotein cholesterol level: a meta-analysis of population-based prospective studies. J Cardiovasc Risk, 1996. 3(2): p. 213-9.
  152. Zilversmit, D.B., Atherogenesis:A postprandial phenomenon. Circulation, 1979. 60: p. 473-485.
  153. Zilversmit, D.B., Atherogenic nature of triglycerides, postprandial lipidemia, and triglyceride-rich remnant lipoproteins. Clin Chem, 1995. 41(1): p. 153-8.
  154. Nordestgaard, B.G. and D.B. Zilversmit, Large lipoproteins are excluded from the arterial wall in diabetic cholesterol-fed rabbits. J Lipid Res, 1988. 29(11): p. 1491-500.
  155. Nordestgaard, B.G., S. Stender, and K. Kjeldsen, Reduced atherogenesis in cholesterol-fed diabetic rabbits. Giant lipoproteins do not enter the arterial wall. Arteriosclerosis, 1988. 8(4): p. 421-8.
  156. Williams, K.J. and I. Tabas, The response-to-retention hypothesis of early atherogenesis. Arteriosclerosis, Thrombosis, and Vascular Biology, 1995. 15: p. 551-561.
  157. Williams, K.J. and I. Tabas, The response-to-retention hypothesis of atherogenesis reinforced. Curr Opin Lipidol, 1998. 9(5): p. 471-4.
  158. Krauss, R.M., Dense low density lipoproteins and coronary artery disease. Am J Cardiol, 1995. 75(6): p. 53B-57B.
  159. Olin-Lewis, K., et al., ApoC-III content of ApoB-containing lipoproteins is associated with binding to the vascular proteoglycan biglycan. J Lipid Res, 2002. 43(11): p. 1969-77.
  160. Chait, A., et al., Susceptibility of small, dense low density lipoproteins to oxidative modification in subjects with the atherogenic lipoprotein phenotype, pattern B. American Journal of Medicine, 1993. 94: p. 350-356.
  161. Tribble, D.L., et al., Oxidative susceptibility of LDL density subfractions is related to their ubiquinol-10 and à-tocopherol content. Proceedings of the National Academy of Sciences of the USA, 1994. 91: p. 1183-1187.
  162. Cornier, M.A., et al., The metabolic syndrome. Endocr Rev, 2008. 29(7): p. 777-822.
  163. Rosenson, R.S., et al., Genetics and causality of triglyceride-rich lipoproteins in atherosclerotic cardiovascular disease. J Am Coll Cardiol, 2014. 64(23): p. 2525-40.
  164. Do, R., et al., Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet, 2013. 45(11): p. 1345-52.
  165. Waterworth, D.M., et al., Genetic variants influencing circulating lipid levels and risk of coronary artery disease. Arterioscler Thromb Vasc Biol, 2010. 30(11): p. 2264-76.
  166. Teslovich, T.M., et al., Biological, clinical and population relevance of 95 loci for blood lipids. Nature, 2010. 466(7307): p. 707-13.
  167. Rip, J., et al., Lipoprotein lipase S447X: a naturally occurring gain-of-function mutation. Arterioscler Thromb Vasc Biol, 2006. 26(6): p. 1236-45.
  168. Ference, B.A., et al., Association of Triglyceride-Lowering LPL Variants and LDL-C-Lowering LDLR Variants With Risk of Coronary Heart Disease. JAMA, 2019. 321(4): p. 364-373.
  169. Pimstone, S.N., et al., Mutations in the gene for lipoprotein lipase. A cause for low HDL cholesterol levels in individuals heterozygous for familial hypercholesterolemia. Arterioscler Thromb Vasc Biol, 1995. 15(10): p. 1704-12.
  170. Burkhardt, R., et al., Trib1 is a lipid- and myocardial infarction-associated gene that regulates hepatic lipogenesis and VLDL production in mice. J Clin Invest, 2010. 120(12): p. 4410-4.
  171. Douvris, A., et al., Functional analysis of the TRIB1 associated locus linked to plasma triglycerides and coronary artery disease. J Am Heart Assoc, 2014. 3(3): p. e000884.
  172. Tg, et al., Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med, 2014. 371(1): p. 22-31.
  173. Jorgensen, A.B., et al., Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med, 2014. 371(1): p. 32-41.
  174. Soufi, M., et al., Mutation screening of the APOA5 gene in subjects with coronary artery disease. J Investig Med, 2012. 60(7): p. 1015-9.
  175. Do, R., et al., Exome sequencing identifies rare LDLR and APOA5 alleles conferring risk for myocardial infarction. Nature, 2015. 518(7537): p. 102-6.
  176. Dewey, F.E., et al., Inactivating Variants in ANGPTL4 and Risk of Coronary Artery Disease. N Engl J Med, 2016. 374(12): p. 1123-33.
  177. Myocardial Infarction, G., et al., Coding Variation in ANGPTL4, LPL, and SVEP1 and the Risk of Coronary Disease. N Engl J Med, 2016. 374(12): p. 1134-44.
  178. Nordestgaard, B.G., R. Wootton, and B. Lewis, Selective retention of VLDL, IDL, and LDL in the arterial intima of genetically hyperlipidemic rabbits in vivo. Molecular size as a determinant of fractional loss from the intima-inner media. Arterioscler Thromb Vasc Biol, 1995. 15(4): p. 534-42.
  179. Norata, G.D., et al., Post-prandial endothelial dysfunction in hypertriglyceridemic subjects: molecular mechanisms and gene expression studies. Atherosclerosis, 2007. 193(2): p. 321-7.
  180. Malloy, M.J. and J.P. Kane, A risk factor for atherosclerosis: triglyceride-rich lipoproteins. Adv Intern Med, 2001. 47: p. 111-36.
  181. Mamo, J.C., S.D. Proctor, and D. Smith, Retention of chylomicron remnants by arterial tissue; importance of an efficient clearance mechanism from plasma. Atherosclerosis, 1998. 141 Suppl 1: p. S63-9.
  182. Havel, R.J. and R.S. Gordon, Jr., Idiopathic hyperlipemia: metabolic studies in an affected family. J Clin Invest, 1960. 39: p. 1777-90.
  183. Benlian, P., et al., Premature atherosclerosis in patients with familial chylomicronemia caused by mutations in the lipoprotein lipase gene. N Engl J Med, 1996. 335(12): p. 848-54.
  184. Zafrir, B., et al., Clinical features and outcomes of severe, very severe, and extreme hypertriglyceridemia in a regional health service. J Clin Lipidol, 2018. 12(4): p. 928-936.
  185. Austin, M.A. and J.E. Hokanson, Epidemiology of triglycerides, small dense low-density lipoprotein, and lipoprotein(a) as risk factors for coronary heart disease. Med Clin North Am, 1994. 78(1): p. 99-115.
  186. Goldberg, I.J., R.H. Eckel, and R. McPherson, Triglycerides and heart disease: still a hypothesis? Arterioscler Thromb Vasc Biol, 2011. 31(8): p. 1716-25.
  187. Brown, W.V., et al., Severe hypertriglyceridemia. J Clin Lipidol, 2012. 6(5): p. 397-408.
  188. Goldberg, A.S. and R.A. Hegele, Severe hypertriglyceridemia in pregnancy. J Clin Endocrinol Metab, 2012. 97(8): p. 2589-96.
  189. Brunzell, J.D., Lipoprotein lipase deficiency and other causes of the chylomicronemia syndrome, in The Metabolic Basis of Inherited Disease, C.R. Scriver, et al., Editors. 1989, McGraw-Hill: New York. p. 1165-1180.
  190. Tremblay, K., et al., Etiology and risk of lactescent plasma and severe hypertriglyceridemia. J Clin Lipidol, 2011. 5(1): p. 37-44.
  191. Lloret Linares, C., et al., Acute pancreatitis in a cohort of 129 patients referred for severe hypertriglyceridemia. Pancreas, 2008. 37(1): p. 13-2.
  192. Cameron, J.L., et al., Acute pancreatitis with hyperlipidemia:The incidence of lipid abnormalities in acute pancreatitis. Annals of Surgery, 1973. 177: p. 483-489.
  193. Farmer, R.G., et al., Hyperlipoproteinemia and pancreatitis. American Journal of Medicine, 1973. 54: p. 161-165.
  194. Wang, Q., et al., Elevated Serum Triglycerides in the Prognostic Assessment of Acute Pancreatitis: A Systematic Review and Meta-Analysis of Observational Studies. J Clin Gastroenterol, 2017. 51(7): p. 586-593.
  195. Mirtallo, J.M., et al., State of the art review: Intravenous fat emulsions: Current applications, safety profile, and clinical implications. Ann Pharmacother, 2010. 44(4): p. 688-700.
  196. Devaud, J.C., et al., Hypertriglyceridemia: a potential side effect of propofol sedation in critical illness. Intensive Care Med, 2012. 38(12): p. 1990-8.
  197. Havel, R.J., Pathogenesis, differentiation and management of hypertriglyceridemia. Adv Intern Med, 1969. 15: p. 117-54.
  198. Yang, F., et al., The role of free fatty acids, pancreatic lipase and Ca+ signalling in injury of isolated acinar cells and pancreatitis model in lipoprotein lipase-deficient mice. Acta Physiol (Oxf), 2009. 195(1): p. 13-28.
  199. Tsuang, W., et al., Hypertriglyceridemic pancreatitis: presentation and management. Am J Gastroenterol, 2009. 104(4): p. 984-91.
  200. Valdivielso, P., A. Ramirez-Bueno, and N. Ewald, Current knowledge of hypertriglyceridemic pancreatitis. Eur J Intern Med, 2014. 25(8): p. 689-94.
  201. Saharia, P., et al., Acute pancreatitis with hyperlipemia: studies with an isolated perfused canine pancreas. Surgery, 1977. 82(1): p. 60-7.
  202. Seplowitz, A.H., S. Chien, and F.R. Smith, Effects of lipoproteins on plasma viscosity. Atherosclerosis, 1981. 38(1-2): p. 89-95.
  203. Chang, Y.T., et al., Association of cystic fibrosis transmembrane conductance regulator (CFTR) mutation/variant/haplotype and tumor necrosis factor (TNF) promoter polymorphism in hyperlipidemic pancreatitis. Clin Chem, 2008. 54(1): p. 131-8.
  204. Ivanova, R., et al., Triglyceride levels and Apolipoprotein E polymorphism in patients with acute pancreatitis. Hepatobiliary Pancreat Dis Int, 2012. 11(1): p. 96-101.
  205. Durrington, P., Dyslipidaemia. Lancet, 2003. 362(9385): p. 717-31.
  206. Parker, F., et al., Evidence for the chylomicron origin of lipids accumulating in diabetic eruptive xanthomas:A correlative lipid biochemical, histochemical and electron microscopic study. Journal of Clinical Investigation, 1970. 49: p. 2172-2187.
  207. Rosenson, R.S., et al., Hypertriglyceridemia and other factors associated with plasma viscosity. Am J Med, 2001. 110(6): p. 488-92.
  208. Inokuchi, R., et al., Hypertriglyceridemia as a possible cause of coma: a case report. J Med Case Rep, 2012. 6: p. 412.
  209. Feingold, K.R., Triglyceride Lowering Drugs, in Endotext, K.R. Feingold, et al., Editors. 2021: South Dartmouth (MA).
  210. Feingold K.R., The Effect of Diet on Cardiovascular Disease and Lipid and Lipoprotein Levels, in Endotext, K.R. Feingold, et al., Editors. 2021: South Dartmouth (MA).
  211. Scandinavian Simvastatin Survival Study Group, Randomized trial of cholesterol lowering in 4444 patients with coronary heart disease: The Scandinavian Simvastatin Survival Study (4S). Lancet, 1994. 344: p. 1383-1389.
  212. Cannon, C.P., et al., Ezetimibe Added to Statin Therapy after Acute Coronary Syndromes. N Engl J Med, 2015.
  213. Koskinen, P., et al., Coronary heart disease incidence in NIDDM patients in the Helsinki Heart Study. Diab Care, 1992. 15: p. 825-829.
  214. ACCORD Study Group, et al., Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med, 2010. 362(17): p. 1563-74.
  215. Keech, A., et al., Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet, 2005. 366(9500): p. 1849-61.
  216. Bezafibrate Infarction Prevention, s., Secondary prevention by raising HDL cholesterol and reducing triglycerides in patients with coronary artery disease. Circulation, 2000. 102(1): p. 21-7.
  217. Camejo, G., Phase 2 clinical trials with K-877 (pemafibrate): A promising selective PPAR-alpha modulator for treatment of combined dyslipidemia. Atherosclerosis, 2017. 261: p. 163-164.
  218. Das Pradhan, A., et al., Triglyceride Lowering with Pemafibrate to Reduce Cardiovascular Risk. N Engl J Med, 2022. 387(21): p. 1923-1934.
  219. Aung, T., et al., Associations of Omega-3 Fatty Acid Supplement Use With Cardiovascular Disease Risks: Meta-analysis of 10 Trials Involving 77917 Individuals. JAMA Cardiol, 2018. 3(3): p. 225-234.
  220. Bhatt, D.L., et al., Cardiovascular Risk Reduction with Icosapent Ethyl for Hypertriglyceridemia. N Engl J Med, 2018.
  221. Nicholls, S.J., et al., Effect of High-Dose Omega-3 Fatty Acids vs Corn Oil on Major Adverse Cardiovascular Events in Patients at High Cardiovascular Risk: The STRENGTH Randomized Clinical Trial. JAMA, 2020. 324(22): p. 2268-2280.
  222. Kalstad, A.A., et al., Effects of n-3 Fatty Acid Supplements in Elderly Patients After Myocardial Infarction: A Randomized, Controlled Trial. Circulation, 2021. 143(6): p. 528-539.
  223. Goff, Z.D. and S.E. Nissen, N-3 polyunsaturated fatty acids for cardiovascular risk. Curr Opin Cardiol, 2022. 37(4): p. 356-363.
  224. Mason, R.P., S.C.R. Sherratt, and R.H. Eckel, Omega-3-fatty acids: Do they prevent cardiovascular disease? Best Pract Res Clin Endocrinol Metab, 2023. 37(3): p. 101681.
  225. Zambon, A., et al., New and Emerging Therapies for Dyslipidemia. Endocrinol Metab Clin North Am, 2022. 51(3): p. 635-653.
  226. Haller, J.F., et al., ANGPTL8 requires ANGPTL3 to inhibit lipoprotein lipase and plasma triglyceride clearance. J Lipid Res, 2017. 58(6): p. 1166-1173.
  227. Rosenson, R.S., et al., Evinacumab in severe hypertriglyceridemia with or without lipoprotein lipase pathway mutations: a phase 2 randomized trial. Nat Med, 2023. 29(3): p. 729-737.
  228. Chaudhry, R., A. Viljoen, and A.S. Wierzbicki, Pharmacological treatment options for severe hypertriglyceridemia and familial chylomicronemia syndrome. Expert Rev Clin Pharmacol, 2018. 11(6): p. 589-598.
  229. Capell, W.H. and R.H. Eckel, Treatment of hypertriglyceridemia. Curr Diab Rep, 2006. 6(3): p. 230-40.
  230. Yuan, G., K.Z. Al-Shali, and R.A. Hegele, Hypertriglyceridemia: its etiology, effects and treatment. CMAJ, 2007. 176(8): p. 1113-20.
  231. Whayne, T.F., Jr. and J.M. Felts, Activation of lipoprotein lipase. Effects of rat serum lipoprotein fractions and heparin. Circ Res, 1970. 27(6): p. 941-51.
  232. Weintraub, M., et al., Continuous intravenous heparin administration in humans causes a decrease in serum lipolytic activity and accumulation of chylomicrons in circulation. J Lipid Res, 1994. 35(2): p. 229-38.
  233. Whayne, T.F., Jr., Concerns about heparin therapy for hypertriglyceridemia. Arch Intern Med, 2010. 170(1): p. 108-9; author reply 109.
  234. Goldberg, I.J., Lipoprotein lipase and lipolysis: central roles in lipoprotein metabolism and atherogenesis. J Lipid Res, 1996. 37(4): p. 693-707.
  235. Aryal, M.R., et al., Acute pancreatitis owing to very high triglyceride levels treated with insulin and heparin infusion. BMJ Case Rep, 2013. 2013.
  236. Khan, A.S., S.U. Latif, and M.A. Eloubeidi, Controversies in the etiologies of acute pancreatitis. JOP, 2010. 11(6): p. 545-52.
  237. Coskun, A., et al., Treatment of hypertriglyceridemia-induced acute pancreatitis with insulin. Prz Gastroenterol, 2015. 10(1): p. 18-22.
  238. Mikhail, N., et al., Treatment of severe hypertriglyceridemia in nondiabetic patients with insulin. Am J Emerg Med, 2005. 23(3): p. 415-7.
  239. Jabbar, M.A., M.I. Zuhri-Yafi, and J. Larrea, Insulin therapy for a non-diabetic patient with severe hypertriglyceridemia. J Am Coll Nutr, 1998. 17(5): p. 458-61.
  240. Thuzar, M., et al., Extreme hypertriglyceridemia managed with insulin. J Clin Lipidol, 2014. 8(6): p. 630-4.
  241. Szczepiorkowski, Z.M., et al., Guidelines on the use of therapeutic apheresis in clinical practice--evidence-based approach from the Apheresis Applications Committee of the American Society for Apheresis. J Clin Apher, 2010. 25(3): p. 83-177.
  242. Stefanutti, C. and U. Julius, Treatment of primary hypertriglyceridemia states - General approach and the role of extracorporeal methods. Atheroscler Suppl, 2015. 18: p. 85-94.
  243. Furuya, T., et al., Plasma exchange for hypertriglyceridemic acute necrotizing pancreatitis: report of two cases. Ther Apher, 2002. 6(6): p. 454-8.
  244. Click, B., et al., The role of apheresis in hypertriglyceridemia-induced acute pancreatitis: A systematic review. Pancreatology, 2015. 15(4): p. 313-20.
  245. Webb, C.B., et al., Effect of TPE vs medical management on patient outcomes in the setting of hypertriglyceridemia-induced acute pancreatitis with severely elevated triglycerides. J Clin Apher, 2021. 36(5): p. 719-726.
  246. Koutroumpakis, E., et al., Management and outcomes of acute pancreatitis patients over the last decade: A US tertiary-center experience. Pancreatology, 2017. 17(1): p. 32-40.
  247. Huang, C., et al., Clinical features and treatment of hypertriglyceridemia-induced acute pancreatitis during pregnancy: A retrospective study. J Clin Apher, 2016. 31(6): p. 571-578.
  248. Wierzbicki, A.S., T.M. Reynolds, and M.A. Crook, Usefulness of Orlistat in the treatment of severe hypertriglyceridemia. Am J Cardiol, 2002. 89(2): p. 229-31.
  249. Tolentino, M.C., et al., Combination of gemfibrozil and orlistat for treatment of combined hyperlipidemia with predominant hypertriglyceridemia. Endocr Pract, 2002. 8(3): p. 208-12.
  250. Davidson, M., et al., The burden of familial chylomicronemia syndrome: interim results from the IN-FOCUS study. Expert Rev Cardiovasc Ther, 2017. 15(5): p. 415-423.
  251. Rouis, M., et al., Therapeutic response to medium-chain triglycerides and omega-3 fatty acids in a patient with the familial chylomicronemia syndrome. Arterioscler Thromb Vasc Biol, 1997. 17(7): p. 1400-6.
  252. Brunzell, J.D., Familial Lipoprotein Lipase Deficiency, in GeneReviews at GeneTests: Medical Genetics Information Resource. 2011, University of Washington: Seattle. p. 1997-2010.
  253. Brunzell JD, D.S., Familial lipoprotein lipase deficiency, Apo CII deficiency and hepatic lipase deficiency., in The Metabolic and Molecular Basis of Inherited Disease, 8th edition2001, McGraw-Hill Book Co.: New York. p. 2789-2816.
  254. Patni, N., C. Quittner, and A. Garg, Orlistat Therapy for Children With Type 1 Hyperlipoproteinemia: A Randomized Clinical Trial. J Clin Endocrinol Metab, 2018. 103(6): p. 2403-2407.
  255. Blackett, P., et al., Lipoprotein abnormalities in compound heterozygous lipoprotein lipase deficiency after treatment with a low-fat diet and orlistat. J Clin Lipidol, 2013. 7(2): p. 132-9.
  256. Tsai, E.C., et al., Potential of essential fatty acid deficiency with extremely low fat diet in lipoprotein lipase deficiency during pregnancy: A case report. BMC Pregnancy Childbirth, 2004. 4(1): p. 27.
  257. Al-Shali, K., et al., Successful pregnancy outcome in a patient with severe chylomicronemia due to compound heterozygosity for mutant lipoprotein lipase. Clin Biochem, 2002. 35(2): p. 125-30.
  258. Carpentier, A.C., et al., Effect of alipogene tiparvovec (AAV1-LPL(S447X)) on postprandial chylomicron metabolism in lipoprotein lipase-deficient patients. J Clin Endocrinol Metab, 2012. 97(5): p. 1635-44.
  259. Sanada, M., et al., Substitution of transdermal estradiol during oral estrogen-progestin therapy in postmenopausal women: effects on hypertriglyceridemia. Menopause, 2004. 11(3): p. 331-6.
  260. Hemelaar, M., et al., Oral, more than transdermal, estrogen therapy improves lipids and lipoprotein(a) in postmenopausal women: a randomized, placebo-controlled study. Menopause, 2003. 10(6): p. 550-8.
  261. Hsu, S.Y., et al., Laparoscopic bariatric surgery for the treatment of severe hypertriglyceridemia. Asian J Surg, 2015. 38(2): p. 96-101.
  262. Gaudet, D., et al., Antisense Inhibition of Apolipoprotein C-III in Patients with Hypertriglyceridemia. N Engl J Med, 2015. 373(5): p. 438-47.

 

Monogenic Disorders Altering HDL Levels

ABSTRACT

 

Very low HDL-C levels (<20mg/dL) may be due to severe elevations in triglycerides, very poorly controlled diabetes, inflammation, infections, malignancy, liver disease, and certain medications such as anabolic steroids. Additionally, variants in multiple genes that each have a small effect but cumulatively lead to a decrease in HDL-C can result in very low HDL-C levels. Finally, rare monogenic disorders such as familial hypoalphalipoproteinemia, Tangier disease, and lecithin acyltransferase (LCAT) deficiency can lead to very low HDL-C levels. In this chapter we discuss the lipid abnormalities and clinical features of these monogenic disorders causing very low HDL-C levels. An elevated concentration of apo A-I and apo A-II is called hyperalphalipoproteinemia (HALP). HALP is classified as moderate (HDL-C levels between 80 and 100 mg/dL) or severe (HDL-C levels > 100 mg/dL). HALP is a heterogeneous condition caused by a variety of genetic and secondary conditions (for example ethanol abuse, primary biliary cirrhosis, multiple lipomatosis, emphysema, exercise, and certain drugs such as estrogens). In many individuals HALP has a polygenic origin. Monogenic HALP includes CETP deficiency, hepatic lipase deficiency, endothelial lipase deficiency, and loss of function mutations in SRB1. In this chapter we discuss the lipid abnormalities and clinical features of these monogenic disorders causing HALP.

 

LOW HDL CONDITIONS

 

The inverse relationship between HDL-C and ASCVD risk is well established but it should be recognized that while this association is consistently observed recent genetic and cardiovascular outcome studies suggest that this association is not causal (1). However, as discussed below major reductions in HDL-C induced by specific monogenic disorders may increase the risk of ASCVD.

 

Isolated low HDL-C levels can occur; however, it is more commonly found in association with hypertriglyceridemia and/or elevated apo B levels, typically as part of the obesity/metabolic syndrome (2). Patients with very low HDL-C (<20 mg/dL) in the absence of severe hypertriglyceridemia, very poorly controlled diabetes, inflammation, infections, malignancy, liver disease, anabolic steroids, or a paradoxical response to PPAR agonists are very rare (<1% of the population) (3,4). These individuals may have a very rare monogenic disorder associated with marked HDL deficiency, including familial hypoalphalipoproteinemia, Tangier disease, and lecithin acyltransferase (LCAT) deficiency. Table 1 summarizes the genetic, lipid, and clinical features of these monogenic low HDL conditions. Inheritance is autosomal co-dominant with heterozygotes having decreases in HDL-C levels approximately midway between normal and homozygotes (3). In some individuals the decrease in HDL-C can be polygenic i.e., variants in multiple genes that each have a small effect but cumulatively lead to a decrease in HDL-C (5).

 

Table 1. Characteristics of Monogenic Low HDL Syndromes

 

Effected genes

Lipids

Clinical features

Familial hypoalpha-lipoproteinemia

apo A-I/apo C-III/ apo A-IV

apo A-I/apo C-III

apo A-I

Apo AI undetectable, marked deficiency in HDL-C, low – normal triglycerides, normal LDL-C

Xanthomas Premature ASCVD Corneal manifestations

Tangier disease

ABCA1

HDL species exclusively preß-1, HDL-C <5 mg/dL

LDL-C low (half normal)

Hepatosplenomegaly

Enlarged tonsils

Neuropathy

ASCVD (6-7th decade)

LCAT deficiency

LCAT

HDL-C <10 mg/dL

apo A-I 20-30 mg/dL

<36% cholesteryl esters

Low LDL-C

Presence of Lp-X particles

FLD develop corneal opacities (“fish eye”), normochromic anemia and proteinuric end stage renal disease

 

FED only develop corneal opacities

Inheritance is autosomal co-dominant with heterozygotes having decreases in HDL-C levels approximately midway between normal and homozygotes (3). FLD- Familial Lecithin: Cholesteryl Ester Acyltransferase Deficiency; FED- Fish Eye Disease

 

Familial Hypoalphalipoproteinemia  

 

Familial hypoalphalipoproteinemia is a heterogeneous group of apolipoprotein A-I (apo A-I) deficiency states. This disorder is the rarest cause of  monogenic severe HDL deficiency (6). These various conditions are characterized by the specific apolipoprotein genes that are affected on the apo A-I/C-III/A-IV gene cluster (3). The genes for these 3 apolipoproteins (apo A-I, apo C-III, and apo A-IV) are grouped together in a cluster on human chromosome 11. In patients with apo A-I/C-III/A-IV deficiency, apoA-1 is undetectable in the plasma and is associated with marked deficiency in HDL-C, low triglyceride levels (due to apo C-III deficiency), and normal LDL-C levels (3). Heterozygotes have plasma HDL-C, apo A-I, apo A-IV, and apo C-III levels that are about 50% of normal (3). This condition is associated with aggressive, premature ASCVD. Additionally, there is evidence of mild fat malabsorption due to deficiency of apo A-IV. Patients with apo A-I/C-III deficiency have undetectable apo A-I and a similar lipid profile as those with apo A-I/C-III/A-IV deficiency (3). This condition is also associated with premature ASCVD. It is distinguished from the former by presence of planar xanthomas and absence of fat malabsorption (since apo A-IV is present). Familial apo A-I deficiency is itself a heterogeneous group of disorders associated with numerous Apo A-I mutations (3). Common manifestations include undetectable plasma Apo A-I, marked HDL deficiency with normal LDL-C and triglyceride levels, xanthomas (planar, tendon, and/or tubero-eruptive depending on the specific gene mutation), and premature ASCVD. Some forms of the disease are also associated with corneal manifestations, including corneal arcus and corneal opacification. One of the interesting manifestations of familial apo A-I deficiency is that levels of apo A-IV and apo E containing HDL particles are only modestly reduced, with preserved electrophoretic mobility and particle size (7).

 

It is notable that familial hypoalphalipoproteinemia is associated with an increased risk of premature ASCVD presumably due to the marked deficiency in Apo A-I and HDL. Given the increased ASCVD risk associated with Apo A-I deficiency, treatment is directed towards aggressive reduction of LDL-C and non-HDL-C levels and reducing other cardiovascular risk factors.

 

Some mutations in Apo A-I are associated with low HDL-C levels and hereditary amyloidosis and are the second most frequent cause of familial amyloidosis (6,8). Note that HDL-C levels are not always decreased in patients with familial amyloidosis secondary to Apo A-I mutations. The N-terminal fragment of the mutated protein is found in the amyloid fragments.

 

Tangier Disease

 

Tangier disease is due to mutations in the gene that codes for ATP-Binding Cassette transporter A1 (ABCA1) and is inherited in an autosomal co-dominant manner (9,10).  Fredrickson first reported this condition in two patients who hailed from Tangier Island in the Chesapeake Bay, for which the disorder is named. ABCA1 facilitates efflux of intracellular cholesterol from peripheral cells to lipid poor A1, the key first step of reverse cholesterol transport (11). As such, this disorder is characterized by severe deficiency of HDL-C (HDL-C <5 mg/dL) and the presence of only thepreß-1 HDL fraction of HDL (10). The poorly lipidated Apo A-I is rapidly catabolized by the kidney. These patients also demonstrate moderate hypertriglyceridemia and low LDL-C levels (10). The decrease in LDL-C is likely due to absence of the transfer of cholesterol from HDL to LDL. Studies have also suggested that an increase in LDL uptake by the liver also occurs (12). The increase in triglycerides may be due to the failure of HDL to provide co-factors that increase lipoprotein lipase activity. Additionally, ABCA1 deficiency in the liver increases triglyceride secretion and hepatic angiopoietin-like protein 3 secretion which could inhibit lipoprotein lipase activity leading to an increase in triglycerides (12,13).

 

Since ABCA1 deficiency impairs free cholesterol efflux from cells, there is accumulation of cholesterol esters in many tissues throughout the body (10). Classically, patients present with hepatosplenomegaly and enlarged yellow-orange hyperplastic tonsils, however, a wide spectrum of phenotypic manifestations is now appreciated with considerable variability in terms of clinical severity and organ involvement (9,10). Peripheral neuropathies are also a common complication and may be relapsing-remitting or chronic progressive (9,10). Tangier disease patients appear to have an increased risk of premature ASCVD, though not as pronounced as those with familial hypoalphalipoproteinemia (3,9,14). When the non-HDL-C levels are greater than 70mg/dL patients with Tangier disease are at higher risk of ASCVD whereas when the non-HDL-C levels are less than 70mg/dL ASCVD is low (9). Less common complications include corneal opacities and hematological manifestations such as thrombocytopenia and hemolytic anemia (9,10).

 

Individuals who are heterozygous for ABCA1 mutations have HDL-C levels that are variable but approximately 50% of normal with normal levels of preß-1 HDL but a deficiency of large α-1 and α-2 HDL particles (10). Cholesterol efflux capacity in heterozygotes has been reported as ~50% of normal. A mutation in one ABCA1 allele has been associated with increased risk of ASCVD in some studies and with no increase in ASCVD risk in other studies (15-20). Different mutations in ABCA1 result in varying HDL-C levels and phenotypes, which might explain the difference in ASCVD risk (21).

 

While Tangier patients manifest characteristically low HDL-C and Apo A-I, this lipid/lipoprotein phenotype is not adequate to make the diagnosis. ABCA1 gene sequence analysis is the preferred test to make the diagnosis of Tangier disease (10). Alternatively, non-denaturing two-dimensional electrophoresis followed by anti-apo A-I immunoblotting demonstrates only preβ1-HDL.

 

Currently, there is no specific treatment for Tangier disease (10). In fact, HDL-C raising therapies such as niacin and fibrates have proven ineffective in patients with this condition (22). Even HDL infusion was not beneficial (23). The major clinical issue in Tangier patients is disabling neuropathy; however, there is no effective intervention to manage this complication (10). Aggressive LDL-C lowering and treatment of other risk factors for atherosclerosis is recommended (10).

 

LCAT Deficiency  

 

LCAT is an enzyme that is bound primarily to HDL, with some also found on LDL (24,25). It facilitates cholesterol esterification by transferring a fatty acid from phosphatidyl choline to cholesterol (24,25). The hydrophobic cholesteryl esters are then sequestered in the core of the lipoprotein particles. LCAT is critical in the maturation of HDL particles. LCAT deficiency is an autosomal co-dominant disorder that manifests as either familial LCAT deficiency (FLD) or fish-eye disease in homozygotes (FED) (24,25). In FLD, mutations in LCAT lead to the inability of LCAT to esterify cholesterol in both HDL and LDL, whereas in FED, mutations in LCAT lead to the inability of LCAT to esterify cholesterol in HDL but the ability of LCAT to esterify cholesterol in LDL is preserved (24,25). Patients with FLD have virtually no cholesterol esters in the circulation while patients with FED have subnormal levels of cholesterol esters carried in apo B containing lipoproteins (24,25). Heterozygotes having decreases in HDL-C levels approximately midway between normal and homozygotes.  

 

Individuals with FLD develop corneal opacities (“fish eye”), normochromic hemolytic anemia (due to cholesterol enrichment of red blood cell membranes), mild thrombocytopenia, and proteinuric end stage renal disease, which is the major cause of morbidity and mortality (24,25). The corneal opacities begin early in life and some patients may need corneal transplants. The rate of development of renal disease is variable but in a large cohort renal failure occurred at a median age of 46 years (26). Patients with FED generally only manifest corneal opacities (24,25).

 

The lipid and lipoprotein profile in patients with FLD usually demonstrates low HDL-C levels (frequently <10 mg/dL) (24,27). In one cohort patients with FED tend to have higher HDL-C levels but in a large systematic review HDL-C levels were similar in patients with FLD and FED (24,27). LDL-C levels tend to be low in FLD and FED while triglyceride levels are increased (24,27). Lipoprotein X (Lp-X) particles are present in patients with FLD but not in patients with FED (24). Lp-X is a multilamellar vesicle with an aqueous core. It is primarily composed of free cholesterol and phospholipid with very little protein (albumin in the core and apolipoprotein C on the surface) and cholesteryl ester.

 

Given the association of Lp-X and kidney disease only with FLD (and not FED) and animal studies demonstrating the nephrotoxicity of Lp-X, it is likely that increased levels of Lp-X results in renal dysfunction in patients with FLD (25,28). Lp-X particles accumulate in the mesangial cells in the glomerulus and are thought to induce inflammation and breakdown of the basement membrane leading to proteinuria. It is notable that after renal transplantation in patients with FLD there is recurrence of renal damage in the transplanted kidney (26).

 

It is unclear as to whether LCAT deficiency is associated with an increased risk of ASCVD (25,29). Atherosclerosis imaging studies have yielded divergent data and the number of patients with FLD or FED studied is limited (25,29). In one study carriers of FLD mutations (i.e., heterozygotes) had a decrease in ASCVD while carriers of FED mutations had an increase in ASCVD (30). This may have been due to higher LDL-C levels in the carriers of FED mutations (30).

 

Current management of FLD focuses on managing the renal dysfunction. The associated kidney disease is traditionally managed with angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, and a low-fat diet (25). Whether lipid lowering drugs are beneficial is unknown. Possible future therapies include enzyme replacement therapy with recombinant human LCAT, liver-directed LCAT gene therapy, small peptide or molecule activators of LCAT, and HDL mimetics (31,32). Infusions of recombinant human LCAT has improved the anemia and most parameters of renal function in a patient with FLD (33). Administration of CER-001, an apolipoprotein A1 (apoA-1)–containing HDL mimetic, has been shown to have beneficial effects on kidney and eye disease in a patient with LCAT deficiency (34).  

 

Approach to the Patient with Low HDL-C Levels

 

When encountering a patient with very low HDL-C levels it is important to first determine if this is a new abnormality or has been present for a long time. If prior HDL-C levels are normal, this excludes a primary monogenic etiology. If the decrease in HDL-C is new, one should consider the possibility of very poorly controlled diabetes, inflammation, infections, malignancy, liver disease, paraproteinemia, anabolic steroids, or a paradoxical response to PPAR agonists. Marked hypertriglyceridemia can also lead to very low HDL-C levels. 

 

In a patient with long-standing very low HDL-C levels without an identifiable secondary cause, one should consider a monogenic etiology. To evaluate potential monogenic causes, a detailed family history, with attention to HDL-C levels, is important. Obtaining lipid levels from relatives is very helpful. A focused physical examination, with particular attention to the skin, eyes, tonsils, and spleen may point to a specific monogenic disorder. Plasma apo A-I levels should be obtained. Individuals with familial hypoalphalipoproteinemia deficiency have undetectable plasma apo A-I. Patients with Tangier disease demonstrate very low apo A-I levels (<5 mg/dL). LCAT deficiency is associated with apo A-I levels that are low but substantially higher than the other monogenic etiologies. Patients with LCAT deficiency also have a higher ratio of free: total cholesterol in plasma and measurement of plasma free (unesterified) cholesterol can be helpful. Two-dimensional gel electrophoresis of plasma followed by immunoblotting with antibodies specific for apo A-I separates lipid-poor preß-HDL from lipid-rich–HDL and can be helpful in differentiating these disorders. Genetic analysis is indicated when a monogenic disorder is suspected.

 

HIGH HDL-C CONDITIONS (HYPERALPHALIPOPROTEINEMIA)

 

An elevated concentration of apo A-I and apo A-II is called hyperalphalipoproteinemia (HALP). HALP is classified as moderate (HDL-C levels between 80 and 100 mg/dL) or severe (HDL-C levels > 100 mg/dL). While it is well recognized that high HDL-C levels are associated with a decrease in ASCVD it should be noted that very high HDL-C levels are paradoxically associated with an increase in ASCVD (35,36).

 

HALP is a heterogeneous condition caused by a variety of genetic and secondary conditions (for example ethanol abuse, primary biliary cirrhosis, multiple lipomatosis, emphysema, exercise, and certain drugs such as estrogens). In many individuals, the very high HDL-C levels have a polygenic origin (5,37). Given the focus of this chapter, monogenic causes of HALP will be reviewed. Monogenic HALP includes CETP deficiency, hepatic lipase deficiency, endothelial lipase deficiency, and loss of function mutations in SRB1. Despite epidemiology that demonstrates an inverse relationship between HDL-C and ASCVD risk, some forms of familial HALP are paradoxically associated with increased cardiovascular risk.

 

HALP is generally identified incidentally after routine assessment of a lipid profile as it is not usually associated with any signs or symptoms. Generally, patients are asymptomatic and no medical therapy is required.

 

Cholesterol Ester Transfer Protein (CETP) Deficiency

 

CETP transfers cholesteryl esters from HDL particles to triglyceride rich lipoproteins and LDL in exchange for triglycerides (11). Individuals who are homozygous for CETP variants have very high HDL-C levels (>100mg/dL) while heterozygotes have moderately increased HDL-C levels (38-41). LDL-C and apo B levels may be normal or modestly decreased. The increase in HDL cholesterol are largely due to the accumulation of cholesterol esters (39). The decrease in LDL-C is due to the failure of cholesterol ester transport from HDL to apo B containing lipoproteins. There is a predominance of small LDL particles. Individuals who are heterozygotes for CETP mutations show modestly elevated HDL-C levels (38,39). In Japanese individuals with HDL-C levels > 100mg/dL 67% were demonstrated to have CETP gene mutations (42). CETP deficiency is the most important and frequent cause of HALP in Japan. CETP deficiency is common in other Asian populations but is relatively rare in other ethnic groups (39). Despite extensive studies the effect of CETP variants on the risk of ASCVD is uncertain (38-40,43). A variety of studies have indicated that a decrease in LDL-C and non-HDL-C levels (i.e. pro-atherogenic lipoproteins) rather than an increase in HDL-C induced by CETP variants underlies a potential beneficial effect on ASCVD (44).  

 

Endothelial Lipase (EL) Deficiency

 

Endothelial lipase (EL) is encoded by the LIPG gene and hydrolyzes phospholipids on HDL resulting in smaller HDL particles that are more rapidly metabolized (11). Genetic variants in LIPG have been identified Iin individuals with elevated HDL-C levels (38,39). As one would predict large HDL particles enriched in phospholipids are observed in individuals deficient in EL (39). Whether variants in LIPG leading to decreased EL activity and increased HDL-C levels reduces ASCVD risk is uncertain (38-40).

 

Hepatic Lipase (HL) Deficiency

 

Hepatic lipase (HL) is encoded by the LIPC gene and mediates the hydrolysis of triglycerides and phospholipids in intermediate density lipoproteins (IDL) and LDL leading to smaller particles (IDL is converted to LDL; LDL is converted from large LDL to small LDL) (11). It also mediates the hydrolysis of triglycerides and phospholipids in HDL resulting in smaller HDL particles (11). Several case reports of families with elevated HDL-C levels (HALP) caused by a genetically defined HL deficiency have been described (39,40). HL deficiency may also be associated with elevated triglycerides and cholesterol with increased intermediate density lipoproteins (IDL) (40,45). Several HL deficient individuals had premature ASCVD likely due to increased levels of apo B containing lipoproteins (40,45). Heterozygotes do not appear to have discrete lipoprotein abnormalities (45).

 

Scavenger Receptor Class B Type I (SR-BI)

 

Scavenger receptor class B type I (SR-BI) is encoded by the SCARB1 gene and facilitates the selective uptake of the cholesterol esters from HDL into the liver, adrenal, ovary, and testes (11). In macrophages and other cells, SR-B1 facilitates the efflux of cholesterol from the cell to HDL particles (11). SR-B1 deficient mice have an increase in atherosclerosis despite elevated HDL-C levels (46). Mutations in SCARB1 associated with decreased SR-B1 have been observed in individuals with high HDL-C levels (47-49). Heterozygotes have intermediate elevations of HDL-C between wild-type and homozygous individuals. Studies have suggested that some but not all mutations in SCARB1 result in an increased risk of ASCVD despite increased HDL-C levels (40,49). A decrease in adrenal function has been reported in some individuals with SCARB1 mutations likely due to a reduced ability of SR-B1 to facilitate cholesterol uptake into the adrenal glands (48,50). Abnormalities in platelet function have also been observed in some patients (50).

 

ACKNOWLEDGEMENTS

 

This work was supported by grants from the Northern California Institute for Research and Education.

 

REFERENCES

 

  1. Kjeldsen EW, Thomassen JQ, Frikke-Schmidt R. HDL cholesterol concentrations and risk of atherosclerotic cardiovascular disease - Insights from randomized clinical trials and human genetics. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159063
  2. Feingold KR. Obesity and Dyslipidemia. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Grossman A, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2023.
  3. Schaefer EJ, Anthanont P, Diffenderfer MR, Polisecki E, Asztalos BF. Diagnosis and treatment of high density lipoprotein deficiency. Prog Cardiovasc Dis 2016; 59:97-106
  4. Geller AS, Polisecki EY, Diffenderfer MR, Asztalos BF, Karathanasis SK, Hegele RA, Schaefer EJ. Genetic and secondary causes of severe HDL deficiency and cardiovascular disease. J Lipid Res 2018; 59:2421-2435
  5. Dron JS, Wang J, Low-Kam C, Khetarpal SA, Robinson JF, McIntyre AD, Ban MR, Cao H, Rhainds D, Dube MP, Rader DJ, Lettre G, Tardif JC, Hegele RA. Polygenic determinants in extremes of high-density lipoprotein cholesterol. J Lipid Res 2017; 58:2162-2170
  6. Zanoni P, von Eckardstein A. Inborn errors of apolipoprotein A-I metabolism: implications for disease, research and development. Curr Opin Lipidol 2020; 31:62-70
  7. Santos RD, Schaefer EJ, Asztalos BF, Polisecki E, Wang J, Hegele RA, Martinez LR, Miname MH, Rochitte CE, Da Luz PL, Maranhao RC. Characterization of high density lipoprotein particles in familial apolipoprotein A-I deficiency. J Lipid Res 2008; 49:349-357
  8. Joy T, Wang J, Hahn A, Hegele RA. APOA1 related amyloidosis: a case report and literature review. Clin Biochem 2003; 36:641-645
  9. Hooper AJ, Hegele RA, Burnett JR. Tangier disease: update for 2020. Curr Opin Lipidol 2020; 31:80-84
  10. Burnett JR, Hooper AJ, McCormick SPA, Hegele RA. Tangier Disease. In: Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Mirzaa G, Amemiya A, eds. GeneReviews((R)). Seattle (WA) 2019.
  11. Feingold KR. Introduction to Lipids and Lipoproteins. In: Feingold KR, Anawalt B, Boyce A, Chrousos G, de Herder WW, Dhatariya K, Dungan K, Grossman A, Hershman JM, Hofland J, Kalra S, Kaltsas G, Koch C, Kopp P, Korbonits M, Kovacs CS, Kuohung W, Laferrere B, McGee EA, McLachlan R, Morley JE, New M, Purnell J, Sahay R, Singer F, Stratakis CA, Trence DL, Wilson DP, eds. Endotext. South Dartmouth (MA) 2021.
  12. Liu M, Chung S, Shelness GS, Parks JS. Hepatic ABCA1 and VLDL triglyceride production. Biochim Biophys Acta 2012; 1821:770-777
  13. Bi X, Pashos EE, Cuchel M, Lyssenko NN, Hernandez M, Picataggi A, McParland J, Yang W, Liu Y, Yan R, Yu C, DerOhannessian SL, Phillips MC, Morrisey EE, Duncan SA, Rader DJ. ATP-Binding Cassette Transporter A1 Deficiency in Human Induced Pluripotent Stem Cell-Derived Hepatocytes Abrogates HDL Biogenesis and Enhances Triglyceride Secretion. EBioMedicine 2017; 18:139-145
  14. Koseki M, Yamashita S, Ogura M, Ishigaki Y, Ono K, Tsukamoto K, Hori M, Matsuki K, Yokoyama S, Harada-Shiba M. Current Diagnosis and Management of Tangier Disease. J Atheroscler Thromb 2021; 28:802-810
  15. Bochem AE, van Wijk DF, Holleboom AG, Duivenvoorden R, Motazacker MM, Dallinga-Thie GM, de Groot E, Kastelein JJ, Nederveen AJ, Hovingh GK, Stroes ES. ABCA1 mutation carriers with low high-density lipoprotein cholesterol are characterized by a larger atherosclerotic burden. Eur Heart J 2013; 34:286-291
  16. van Dam MJ, de Groot E, Clee SM, Hovingh GK, Roelants R, Brooks-Wilson A, Zwinderman AH, Smit AJ, Smelt AH, Groen AK, Hayden MR, Kastelein JJ. Association between increased arterial-wall thickness and impairment in ABCA1-driven cholesterol efflux: an observational study. Lancet 2002; 359:37-42
  17. Schaefer EJ, Zech LA, Schwartz DE, Brewer HB, Jr. Coronary heart disease prevalence and other clinical features in familial high-density lipoprotein deficiency (Tangier disease). Ann Intern Med 1980; 93:261-266
  18. Frikke-Schmidt R, Nordestgaard BG, Stene MC, Sethi AA, Remaley AT, Schnohr P, Grande P, Tybjaerg-Hansen A. Association of loss-of-function mutations in the ABCA1 gene with high-density lipoprotein cholesterol levels and risk of ischemic heart disease. JAMA 2008; 299:2524-2532
  19. Frikke-Schmidt R. Genetic variation in the ABCA1 gene, HDL cholesterol, and risk of ischemic heart disease in the general population. Atherosclerosis 2010; 208:305-316
  20. Iatan I, Alrasadi K, Ruel I, Alwaili K, Genest J. Effect of ABCA1 mutations on risk for myocardial infarction. Curr Atheroscler Rep 2008; 10:413-426
  21. Brunham LR, Singaraja RR, Hayden MR. Variations on a gene: rare and common variants in ABCA1 and their impact on HDL cholesterol levels and atherosclerosis. Annu Rev Nutr 2006; 26:105-129
  22. Serfaty-Lacrosniere C, Civeira F, Lanzberg A, Isaia P, Berg J, Janus ED, Smith MP, Jr., Pritchard PH, Frohlich J, Lees RS, et al. Homozygous Tangier disease and cardiovascular disease. Atherosclerosis 1994; 107:85-98
  23. Schaefer EJ, Anderson DW, Zech LA, Lindgren FT, Bronzert TB, Rubalcaba EA, Brewer HB, Jr. Metabolism of high density lipoprotein subfractions and constituents in Tangier disease following the infusion of high density lipoproteins. J Lipid Res 1981; 22:217-228
  24. Pavanello C, Calabresi L. Genetic, biochemical, and clinical features of LCAT deficiency: update for 2020. Curr Opin Lipidol 2020; 31:232-237
  25. Saeedi R, Li M, Frohlich J. A review on lecithin:cholesterol acyltransferase deficiency. Clin Biochem 2015; 48:472-475
  26. Pavanello C, Ossoli A, Arca M, D'Erasmo L, Boscutti G, Gesualdo L, Lucchi T, Sampietro T, Veglia F, Calabresi L. Progression of chronic kidney disease in familial LCAT deficiency: a follow-up of the Italian cohort. J Lipid Res2020; 61:1784-1788
  27. Mehta R, Elias-Lopez D, Martagon AJ, Perez-Mendez OA, Sanchez MLO, Segura Y, Tusie MT, Aguilar-Salinas CA. LCAT deficiency: a systematic review with the clinical and genetic description of Mexican kindred. Lipids Health Dis 2021; 20:70
  28. Ossoli A, Neufeld EB, Thacker SG, Vaisman B, Pryor M, Freeman LA, Brantner CA, Baranova I, Francone NO, Demosky SJ, Jr., Vitali C, Locatelli M, Abbate M, Zoja C, Franceschini G, Calabresi L, Remaley AT. Lipoprotein X Causes Renal Disease in LCAT Deficiency. PLoS One 2016; 11:e0150083
  29. Norum KR, Remaley AT, Miettinen HE, Strom EH, Balbo BEP, Sampaio C, Wiig I, Kuivenhoven JA, Calabresi L, Tesmer JJ, Zhou M, Ng DS, Skeie B, Karathanasis SK, Manthei KA, Retterstol K. Lecithin:cholesterol acyltransferase: symposium on 50 years of biomedical research from its discovery to latest findings. J Lipid Res2020; 61:1142-1149
  30. Oldoni F, Baldassarre D, Castelnuovo S, Ossoli A, Amato M, van Capelleveen J, Hovingh GK, De Groot E, Bochem A, Simonelli S, Barbieri S, Veglia F, Franceschini G, Kuivenhoven JA, Holleboom AG, Calabresi L. Complete and Partial Lecithin:Cholesterol Acyltransferase Deficiency Is Differentially Associated With Atherosclerosis. Circulation 2018; 138:1000-1007
  31. Freeman LA, Karathanasis SK, Remaley AT. Novel lecithin: cholesterol acyltransferase-based therapeutic approaches. Curr Opin Lipidol 2020; 31:71-79
  32. Vitali C, Rader DJ, Cuchel M. Novel therapeutic opportunities for familial lecithin:cholesterol acyltransferase deficiency: promises and challenges. Curr Opin Lipidol 2023; 34:35-43
  33. Shamburek RD, Bakker-Arkema R, Auerbach BJ, Krause BR, Homan R, Amar MJ, Freeman LA, Remaley AT. Familial lecithin:cholesterol acyltransferase deficiency: First-in-human treatment with enzyme replacement. J Clin Lipidol 2016; 10:356-367
  34. Faguer S, Colombat M, Chauveau D, Bernadet-Monrozies P, Beq A, Delas A, Soler V, Labadens I, Huart A, Benlian P, Schanstra JP. Administration of the High-Density Lipoprotein Mimetic CER-001 for Inherited Lecithin-Cholesterol Acyltransferase Deficiency. Ann Intern Med 2021; 174:1022-1025
  35. Hirata A, Sugiyama D, Watanabe M, Tamakoshi A, Iso H, Kotani K, Kiyama M, Yamada M, Ishikawa S, Murakami Y, Miura K, Ueshima H, Okamura T, Evidence for Cardiovascular Prevention from Observational Cohorts in Japan Research G. Association of extremely high levels of high-density lipoprotein cholesterol with cardiovascular mortality in a pooled analysis of 9 cohort studies including 43,407 individuals: The EPOCH-JAPAN study. J Clin Lipidol 2018; 12:674-684 e675
  36. Madsen CM, Varbo A, Nordestgaard BG. Extreme high high-density lipoprotein cholesterol is paradoxically associated with high mortality in men and women: two prospective cohort studies. Eur Heart J 2017; 38:2478-2486
  37. Motazacker MM, Peter J, Treskes M, Shoulders CC, Kuivenhoven JA, Hovingh GK. Evidence of a polygenic origin of extreme high-density lipoprotein cholesterol levels. Arterioscler Thromb Vasc Biol 2013; 33:1521-1528
  38. Larach DB, Cuchel M, Rader DJ. Monogenic causes of elevated HDL cholesterol and implications for development of new therapeutics. Clin Lipidol 2013; 8:635-648
  39. Giammanco A, Noto D, Barbagallo CM, Nardi E, Caldarella R, Ciaccio M, Averna MR, Cefalu AB. Hyperalphalipoproteinemia and Beyond: The Role of HDL in Cardiovascular Diseases. Life (Basel) 2021; 11
  40. Kardassis D, Thymiakou E, Chroni A. Genetics and regulation of HDL metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159060
  41. Yamashita S, Maruyama T, Hirano K, Sakai N, Nakajima N, Matsuzawa Y. Molecular mechanisms, lipoprotein abnormalities and atherogenicity of hyperalphalipoproteinemia. Atherosclerosis 2000; 152:271-285
  42. Hirano K, Yamashita S, Kuga Y, Sakai N, Nozaki S, Kihara S, Arai T, Yanagi K, Takami S, Menju M, et al. Atherosclerotic disease in marked hyperalphalipoproteinemia. Combined reduction of cholesteryl ester transfer protein and hepatic triglyceride lipase. Arterioscler Thromb Vasc Biol 1995; 15:1849-1856
  43. Yamashita S, Matsuzawa Y. Re-evaluation of cholesteryl ester transfer protein function in atherosclerosis based upon genetics and pharmacological manipulation. Curr Opin Lipidol 2016; 27:459-472
  44. Nicholls SJ, Ray KK, Nelson AJ, Kastelein JJP. Can we revive CETP-inhibitors for the prevention of cardiovascular disease? Curr Opin Lipidol 2022; 33:319-325
  45. Weissglas-Volkov D, Pajukanta P. Genetic causes of high and low serum HDL-cholesterol. J Lipid Res 2010; 51:2032-2057
  46. Trigatti B, Rayburn H, Vinals M, Braun A, Miettinen H, Penman M, Hertz M, Schrenzel M, Amigo L, Rigotti A, Krieger M. Influence of the high density lipoprotein receptor SR-BI on reproductive and cardiovascular pathophysiology. Proc Natl Acad Sci U S A 1999; 96:9322-9327
  47. Brunham LR, Tietjen I, Bochem AE, Singaraja RR, Franchini PL, Radomski C, Mattice M, Legendre A, Hovingh GK, Kastelein JJ, Hayden MR. Novel mutations in scavenger receptor BI associated with high HDL cholesterol in humans. Clin Genet 2011; 79:575-581
  48. Vergeer M, Korporaal SJ, Franssen R, Meurs I, Out R, Hovingh GK, Hoekstra M, Sierts JA, Dallinga-Thie GM, Motazacker MM, Holleboom AG, Van Berkel TJ, Kastelein JJ, Van Eck M, Kuivenhoven JA. Genetic variant of the scavenger receptor BI in humans. N Engl J Med 2011; 364:136-145
  49. Zanoni P, Khetarpal SA, Larach DB, Hancock-Cerutti WF, Millar JS, Cuchel M, DerOhannessian S, Kontush A, Surendran P, Saleheen D, Trompet S, Jukema JW, De Craen A, Deloukas P, Sattar N, Ford I, Packard C, Majumder A, Alam DS, Di Angelantonio E, Abecasis G, Chowdhury R, Erdmann J, Nordestgaard BG, Nielsen SF, Tybjaerg-Hansen A, Schmidt RF, Kuulasmaa K, Liu DJ, Perola M, Blankenberg S, Salomaa V, Mannisto S, Amouyel P, Arveiler D, Ferrieres J, Muller-Nurasyid M, Ferrario M, Kee F, Willer CJ, Samani N, Schunkert H, Butterworth AS, Howson JM, Peloso GM, Stitziel NO, Danesh J, Kathiresan S, Rader DJ, Consortium CHDE, Consortium CAE, Global Lipids Genetics C. Rare variant in scavenger receptor BI raises HDL cholesterol and increases risk of coronary heart disease. Science 2016; 351:1166-1171
  50. Chadwick AC, Sahoo D. Functional genomics of the human high-density lipoprotein receptor scavenger receptor BI: an old dog with new tricks. Curr Opin Endocrinol Diabetes Obes 2013; 20:124-131

 

Pharmacological Causes of Hyperprolactinemia

ABSTRACT

 

Hyperprolactinemia represents a multifaceted endocrine disorder with both physiological and pathological causes. The increased use of anti-psychotic and anti-depressant medications has increased the role pharmaceutical agents play in inducing hyperprolactinemia, being the most frequent cause of hyperprolactinemia in clinical practice. This has particularly impacted females, who demonstrate a higher susceptibility to drug-induced hyperprolactinemia. Of these medications, anti-psychotics, neuroleptic-like medications, anti-depressants, and histamine receptor type 2 antagonists, emerge as the most prominent culprits. Furthermore, opioids, some anti-hypertensive agents, proton pump inhibitors, estrogens, and other less potent hyperprolactinemia-inducing medications are recognized as potential contributors to drug-induced hyperprolactinemia. Many herbal medicines are reported as lactogenic, but their ability to cause hyperprolactinemia remains unclear. This review endeavors to elucidate the intricate mechanisms underlying the induction of hyperprolactinemia by pharmacological agents. We have included available data on the prevalence and extent of drug-induced changes in prolactin levels. We have also included data on herbal agents. We have highlighted where controversial data are identified. Although a detailed exploration of how these medications impact prolactin regulation is beyond the scope of this chapter, this review aims to deepen our understanding of the interplay between pharmacological agents and their effects on prolactin levels, contributing to valuable insights, refined therapeutic approaches, and better patient care.

 

INTRODUCTION

 

The most common cause of consistently high prolactin levels is drug-induced hyperprolactinemia. The overall incidence is higher in women compared to men. Drug-induced causes tripled during the 20-year follow-up period, reflecting the increased prevalence of psychoactive drug use (1). Several drugs have been reported to induce hyperprolactinemia, either by inhibiting dopamine receptors or their actions or by directly stimulating prolactin secretion (2).

 

High levels of prolactin can be attributed to various physiological factors, such as pregnancy and breast-feeding, while minor increases in prolactin levels may also occur during ovulation, after sexual intercourse, during periods of stress, exercise, after food intake, or in association with irritation of the chest wall and breast stimulation. Pathological causes can be related to hypothalamo-pituitary disorders or non-hypothalamo-pituitary disorders. Hypothalamo-pituitary disorders include prolactin-secreting pituitary tumors (including lactotroph tumors, mammo-somatotroph tumors, mature pluri-hormonal PIT1-lineage tumors, immature PIT1-lineage tumors, acidophil stem cell tumors, multi-hormonal pituitary tumors, mixed somatotroph, and lactotroph tumors, and pluri-hormonal tumors) (3); hypothalamic and pituitary stalk compression or damage (non-prolactin-secreting pituitary adenomas, craniopharyngiomas, meningiomas, germinomas, granulomas, metastasis, Rathke cleft cysts, hypophysitis, radiation, surgery, and trauma); infiltrative pituitary disorders; pituitary hyperplasia (McCune-Albright, Carney complex, X-LAG). Other causes include primary hypothyroidism; adrenal insufficiency; systemic diseases such as chronic renal failure and liver cirrhosis; polycystic ovary syndrome; neurogenic causes (chest wall trauma or surgery, herpes zoster); seizures; untreated severe phenylketonuria; pseudocyesis (false pregnancy); autoimmune diseases (lupus, rheumatoid arthritis, multiple sclerosis, systemic sclerosis, Behcet’s disease, polymyositis); cancers (breast, ovarian, colon, hepatocellular) (4). During the diagnostic process of hyperprolactinemia, it is crucial to consider the possibility of macroprolactinoma and the ‘hook’ effect, although the latter is usually not relevant in drug-induced cases (5).

 

Drug-induced hyperprolactinemia is often characterized by prolactin levels ranging from 25 to 100 ng/mL (530-2130 mIU/L). However, certain medications including metoclopramide, risperidone, amisulpride, and phenothiazines can lead to prolactin levels surpassing 200 ng/mL (4255 mIU/L) (6). On similar doses of prolactin-raising anti-psychotics, women with chronic use are more likely to develop hyperprolactinemia than men, reaching significantly higher prolactin levels, with mean levels of 50 ng/mL (1065 mIU/L) (7,8). Younger age was associated with higher prolactin levels in women, but not in men (8,9). Route of drug administration is important, with prolactin levels returning to normal after cessation of the drug: within 2-3 weeks after stopping oral treatment, but no sooner than 6 months after discontinuation of intramuscular depot administration (10).

 

This chapter will encompass a comprehensive discussion of all pharmacological causes as well as some alternative factors contributing to changes in prolactin levels especially hyperprolactinemia.

 

EPIDEMIOLOGY

 

Drug-induced hyperprolactinemia is the most common cause of consistently high prolactin levels. A retrospective follow-up study conducted in Scotland, involving 32,289 hyperprolactinemic individuals from 1993 to 2013, concluded that within the non-pregnancy-related group, the most prominent cause was drug-induced hyperprolactinemia (45.9%), followed by pituitary disorders (25.6%), macroprolactinoma (7.5%), and hypothyroidism (6.1%). Nevertheless, 15% of cases were deemed idiopathic. The overall incidence was higher in women aged 25-44 years old compared to men (1). Female predominance is reported in other studies with a female: male ratio of 5.9:1 and the mean age at diagnosis of hyperprolactinemia is 40 (range 14–85) years (2,11).The position of hyperprolactinemia as a side effect of medications has been assessed in a French Pharmacovigilance Database from 1985 to 2000, which reported 159 cases of hyperprolactinemia out of 182,836 adverse drug reactions (11). The rates of hyperprolactinemia related to therapeutic drug classes were recorded as 31% associated with anti-psychotics, 28% with neuroleptic-like drugs (medications with a similar mechanism of actions as neuroleptics, but used for different purposes, for example movement disorders or anti-emetics), 26% with anti-depressants, 5% with histamine receptor type 2 (H2-receptor) antagonists, and 10% with other drugs.

 

PROLACTIN CONTROL MECHANISMS

 

Prolactin is a polypeptide primarily produced in the anterior pituitary gland, with secondary production occurring in other tissues such as the gonads, mammary gland, endometrium, prostate, lymphocytes, hematopoietic cells, skin, brain, retina, inner ear cochlea, decidua, pancreas, liver, endothelium, and adipose tissue (12–14). In breast and prostate cancer, prolactin has even been proposed as a tumor marker (15,16). Prolactin acts through prolactin receptors (PRLR), which belong to the family of cytokine receptors associated with the non-receptor tyrosine Janus kinase 2. PRLR can activate the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway, MAPK (mitogen-activated protein kinase), PI3 (phosphoinositide 3-kinase), Src kinase, as well as the Nek3 / Vav2 / Rac1 serine / threonine kinase pathway (17). There are different isoforms of this receptor: a long isoform, intermediate isoform, 2 short isoforms S1a and S1b which are formed by alternative splicing and partial deletion of exons 10 and 11, and soluble PRLR (18). These different isoforms are expressed in different tissues, mostly studied in rats. The long isoform is mainly expressed in the adrenal glands, kidneys, mammary glands, small intestine, bile ducts, choroid plexus, and pancreas whereas the short isoform is in the liver and ovaries (19). Prolactin possesses nearly 300 functions apart from lactation including neuroprotection and neurogenesis, offspring recognition by both parents, adipose and weight homeostasis, islet functions, immune regulation, angiogenesis, osmoregulation, and mitogenesis (20).

 

The secretion of prolactin produced by lactotroph cells in the anterior pituitary gland has a circadian rhythm with higher levels during sleep and lower levels during wakefulness (21). Even though pulsatility frequency does not significantly change over 24 hours, the amplitude of pulses is higher during night and day sleep, while wakefulness is associated with an immediate offset of active secretion. Prolactin is lower during the rapid eye movement stage of sleep (22).

 

The synthesis and secretion of prolactin is under the complex control of peptides, steroid hormones, and neurotransmitters, which can act as inhibitory or stimulatory factors, either by a direct effect on lactotroph cells or by indirect pathways through inhibition of dopaminergic tracts, and are widely studied in mammals (2). Dopamine plays a crucial role in inhibiting prolactin secretion. Dopamine can bind the five types of dopamine receptors (G-protein coupled receptors): DRD1, DRD2, DRD3, DRD4 and DRD5, while lactotroph cells express mainly D2 receptors. Dopamine can reach the pituitary via three pathways (Figure 1): through the tuberoinfundibular dopaminergic (TIDA) system, the tuberohypophysial tract (THDA), and the periventricular hypophyseal (PHDA) dopaminergic neurons (23). TIDA neurons originate from the rostral arcuate nucleus of the hypothalamus and release dopamine into the perivascular spaces of the medial eminence and through long portal vessels dopamine reaches the anterior pituitary gland. The THDA neurons originate in the rostral arcuate nucleus and project into the medial and posterior pituitary lobes and release dopamine at these sites. From THDA tract dopamine then reaches lactotroph cells through the short portal vessels (24). PHDA neurons originate in the periventricular nucleus and axons terminate in the intermediate lobe and dopamine release follows the same direction as from the THDA neurons. Prolactin-inhibiting neurons are considered to be a functional unit working synchronously (23). The binding of dopamine to D2 receptors on the plasma membrane of lactotroph cells inhibits prolactin protein, PRL gene transcription, as well as lactotroph proliferation (24). The release of prolactin through exocytosis of prolactin secretory granules is influenced by dopamine through various pathways. Specifically, D2 receptors are coupled with pertussis toxin-sensitive G proteins, which subsequently inhibit adenylate cyclase activity, resulting in decreased levels of cyclic adenosine monophosphate (cAMP) (25).

 

Additionally, the activation of potassium (K+) channels occurs, leading to a reduction in voltage-gated calcium (Ca2+) currents and inhibition of inositol phosphate production. Collectively, these intracellular signaling events culminate in a decrease in the concentration of free calcium ions (Ca2+) resulting in membrane hyperpolarization, ultimately inhibiting the exocytosis of prolactin from its granules (26). The inhibition of PRL gene transcription occurs when D2 receptors are activated, leading to the inhibition of MAPK or protein kinase C pathways. This activation results in a reduction of phosphorylation events on Ets family transcription factors. These transcription factors play a crucial role in the stimulatory responses of thyrotropin-releasing hormone (TRH), insulin, and epidermal growth factor (EGF) on prolactin expression. Moreover, the Ets family transcription factors interact with the PIT1 protein, which is essential for cAMP-mediated PRL gene expression (27). Dopamine exerts anti-mitogenic effects by activating D2 receptors through multiple pathways. These include the inhibition of MAPK (mitogen-activated protein kinase) signaling, protein kinase A signaling, and stimulation of phospholipase D activity. Additionally, dopamine engages a pertussis toxin-insensitive pathway, activates the extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathway, and inhibits the AKT/protein kinase B pathway (28–31).

 

In addition to the dopaminergic inhibitory system, the γ-aminobutyric acid (GABA)-ergic tuberoinfundibular system, culminating in the median eminence, exhibits inhibitory properties, albeit of lesser potency compared to the dopaminergic system, while also having a role in prolactin modulation. GABA-B receptors are discernible both within the anterior pituitary gland, contributing to the maintenance of low prolactin levels, and in TIDA neurons which can be powerfully inhibited by GABA via hyperpolarization, consequently contributing to an elevation in prolactin levels (32,33).

 

Prolactin itself has two negative feedback effects: through ``short-loop feedback regulation`` it enhances the activity of TIDA neurons, where both long and short forms of the PRLR are expressed, with the long isoform being predominant in the arcuate and periventricular nuclei, regulating tyrosine hydroxylase (a rate-limiting enzyme in dopamine synthesis) leading to increase of dopamine release, which inhibits prolactin, as well as autocrine inhibition (34,35). Several other local factors influence prolactin release within pituitary gland as shown in Figure 1.

Figure 1. Prolactin – Central Nervous System Regulation.

 

Other prolactin inhibitory factors include somatostatin, acetylcholine, endothelins, gastrin, and growth hormone, while stimulatory factors include thyrotropin-releasing hormone (TRH) (as seen in primary hypothyroidism), angiotensin II, vasopressin, oxytocin, VIP, galanin, and estrogen.

 

Experiments conducted on rats to elucidate the relationship between the adrenergic system and the regulation of prolactin secretion have focused on stimulating or inhibiting α and β adrenergic receptors. Functional hyperprolactinemia is a complex hormonal interplay of stress-induced neuroendocrine changes involving the dopamine, serotonin and adrenergic systems (36,37). Evidence suggests that the mediobasal hypothalamus and preoptic-anterior hypothalamus harbor the primary adrenoreceptors (38,39). Injecting the α2 agonist clonidine into the mediobasal hypothalamus resulted in a dose-dependent increase in prolactin secretion. This effect was counteracted by the blockade of idazoxan (α2 antagonist). Similarly, the stimulation of prolactin release was induced by isoprenaline (β agonist) and notably attenuated by the β antagonist propranolol. The β2 agonist salbutamol also exhibited efficacy in stimulating prolactin secretion. Conversely, adrenergic agonists such as noradrenaline (mixed α and β), phenylephrine (α1), and tyramine (sympathomimetic) in the mediobasal hypothalamus, failed to elicit an effect on prolactin secretion.

 

Within the preoptic anterior hypothalamus, noradrenaline and adrenaline were found to stimulate prolactin secretion (40). However, the administration of the α1 agonist phenylephrine failed to stimulate prolactin, indirectly suggesting that the stimulatory effect of noradrenaline in the preoptic anterior hypothalamus is likely due to its action at α2 sites. α2 agonism has been shown to reduce the function of tuberoinfundibular dopaminergic neurons leading to increase prolactin production and secretion (41). Consequently, it was inferred that the activation of α2 and β adrenoceptors in the mediobasal hypothalamus and α2 adrenoceptors in the preoptic-anterior hypothalamus, proximal to prolactin-regulating neurons, leads to heightened prolactin secretion, while the action of α1 in the mediobasal hypothalamus may be inhibitory (42).

 

Cholinergic activation may have opposite roles in rodents and humans. Cholinergic agonists suppress prolactin release induced by morphine in rats, suggesting that the central cholinergic system has an inhibitory effect on the prolactin release triggered by morphine or β-endorphine, but this cholinergic inhibition does not occur through catecholaminergic neurons (43). Conversely, in humans, cholinomimetic drugs can increase prolactin levels associated with raised plasma β-endorphin, suggesting a stimulatory interplay of cholinergic factors and endogenous opioids on prolactin levels (44), although circulating opioids may not directly relate to central levels.

 

TIDA neurons express estradiol and progesterone receptors. Estradiol action leads to reduced secretion of dopamine into the portal blood system and mediates a prolactin surge. Progesterone, in addition, suppresses dopamine release being responsible for the plateau phase of the surge (23). Estrogen specifically affects prolactin synthesis by influencing lactotroph cell sensitivity, expression of pituitary dopamine receptor downregulation, and the expression of the prolactin receptor gene (2,34). Ghrelin, a hormone involved in metabolic balance, directly stimulates prolactin secretion at the pituitary level (45).

 

Tachykinins (substance P, neurokinins A, and B, neuropeptide K, neuropeptide ϒ) can act directly on the lactotroph cell and indirectly within the hypothalamus or posterior pituitary. They have a multifaceted impact on prolactin secretion, with both stimulatory and inhibitory effects. They can stimulate prolactin secretion by stimulating and potentiating the release of oxytocin, vasopressin, TRH, VIP, serotonin and glutamate, and by inhibiting GABA. Tachykinins through paracrine actions can directly increase prolactin within the anterior pituitary. They can also increase dopamine but the overall effect is prolactin elevation. Under specific circumstances, the stimulation of dopamine release can be prominent leading to a decrease in prolactin (46). Endogenous opioids are involved in regulating prolactin secretion, particularly during stressful situations, by reducing the activity of tuberoinfundibular dopaminergic neurons mediated by μ-, κ‑, and δ- opioid receptors, resulting in increased prolactin release (47). Prolonged nicotine exposure has been associated with desensitization of dopamine receptors, diminished dopamine turnover, and a decrease in their abundance within the nigrostriatal pathways (48). These alterations have been suggested to contribute to a diminished prolactin response to opiate blockade observed in individuals who smoke. Similar to opioids, histamine has been shown to induce prolactin production predominantly through inhibiting dopaminergic and stimulating serotoninergic and vasopressin-ergic neurons (49).

 

Serotoninergic pathways originating from the dorsal raphe nucleus play a physiological role in mediating nocturnal surges and suckling-induced prolactin rises through a serotonin interaction via serotonin type 1 and 2 receptors (5-hydroxytryptamine receptors, 5HT1, and 5HT2). 5-HT could either release a PRL-releasing factor or inhibit dopamine release. The paraventricular nucleus, where serotoninergic pathways terminate, contains postsynaptic serotonin 5-HT1A, 5-HT2, and 5-HT2C receptor subtypes, and possibly 5-HT3 receptors (50). It was shown that the prolactin-releasing effect of serotonin probably occurs mostly via 5-HT1C / 2 receptors because ritanserin (an elective 5-HT1C / 2 receptor antagonist) opposed this effect (51). Serotonin stimulation of prolactin–releasing factor (PRF) neurons in the paraventricular nucleus leads to PRF release (like VIP and oxytocin) mediating hyperprolactinemia. Moreover, serotoninergic stimulation of GABAergic neurons in the tuberoinfundibular-GABA system has been shown to inhibit TIDA cells which contain 5-HT1A receptors, therefore inhibiting dopamine synthesis/release resulting in increased prolactin secretion (52).

 

Oxytocin, through the posterior pituitary and vasoactive intestinal peptide (VIP) in the anterior pituitary, play significant roles in enhancing PRL gene transcription and modulating dopamine inhibition. Animal studies suggest a potential mediation of VIP by oxytocin to stimulate prolactin secretion (53).

 

The extensive hormonal regulation of prolactin renders it susceptible to various disturbances caused by different classes of medications.

 

CLINICAL CHARACTERISTICS

 

Persistent hyperprolactinemia is associated with disturbances of the gonadal axis leading to interruptions of gonadotrophin-releasing hormone pulsatility and inhibition of luteinizing hormone and follicle-stimulating hormone release (54). Clinical manifestations attributed to hyperprolactinemia predominantly stem from the suppression of the gonadal axis. In premenopausal women, a spectrum of menstrual cycle dysfunctions is observed, spanning from luteal phase shortening to complete amenorrhea, often correlating with elevated prolactin levels. Secondary amenorrhea can be due to hyperprolactinemia in up 30% of patients, and up to 75% of patients with amenorrhea and galactorrhea (55). Beyond these effects, an array of hypoestrogenic indicators may manifest, including symptoms like vaginal dryness, diminished libido, and decreased energy levels. Galactorrhea can be present in up to 80% of females (55,56). In men, the impact of hyperprolactinemia is manifested through a decrease in libido, ranging from diminished sexual desire to oligospermia or even azoospermia attributed to hypogonadotropic hypogonadism. Notably, erectile dysfunction may arise, primarily attributed to the direct inhibitory influence of dopamine, and can be potentially reversed through the administration of dopamine agonists (57). Gynecomastia, on the contrary, is a manifestation of secondary hypogonadism rather than elevated prolactin levels, whereas galactorrhea is rare in men (21).

 

In both genders infertility can be observed, with diminished bone mineral density. In females, bone mineral density is significantly decreased in women with amenorrhea and increases during treatment and menstrual cycle restoration (58). Additionally, in cases where hyperprolactinemia is attributed to a mass, accompanying clinical indications may encompass headaches, visual field disturbances, cranial nerve palsies, and hypopituitarism. Notably, these manifestations may be the only clinical features in post-menopausal women (21,59).

 

PSYCHOTROPIC MEDICATIONS

 

Anti-psychotics and neuroleptic-like drugs are psychotropic medications which primarily exert their anti-psychotic effects through the blockade of DRD2 and D4 receptors in the mesolimbic area. Newer classes of anti-psychotics block 5HT2 and sometimes noradrenergic α1 or α2 receptors (4,35,60). Blockade of D2 receptors in the hypothalamic tuberoinfundibular system and lactotroph cells results in disinhibition of prolactin secretion leading to hyperprolactinemia, being the most common drugs known to induce hyperprolactinemia (61) (Figure 2). On the contrary, strong binding to D2 receptors can extend the half-life of dopamine by approximately 50%. This effect is achieved through two primary mechanisms: direct blockade of the dopamine transporter (DAT) and antagonism of D2 autoreceptors. These processes collectively result in reduced reuptake of dopamine, prolonging its presence in the synaptic area and further stimulating an upregulation of receptors. However, it is important to note that chronic use of anti-psychotics can lead to the reversal of upregulation of DAT (mRNA and protein), potentially contributing to treatment resistance and potentially lower prolactin elevations in the long term. Nonetheless, it is worth mentioning that anti-psychotics typically exhibit a lower affinity for dopamine transporter blockade compared to selective DAT blockers such as nomifensine (62).

Figure 2. Mechanisms of drug-induced hyperprolactinemia with selected examples (adopted from La Torre et al. (2). In addition to opiates, cholinomimetics, PPIs and smoking indirectly also stimulate the opioid receptors. PPIs, Protein pump inhibitors; TCAs, tricyclic anti-depressants; MAO, monoamine oxidase; SSRIs, selective serotonin reuptake inhibitors; SNRIs, serotonin-noradrenaline reuptake inhibitors.

 

The potency of anti-psychotics and neuroleptic-like medications to induce a rise in prolactin levels varies (Table 1). The level of prolactin increase depends on the anti-psychotic drug (different affinity and selectivity for dopamine receptors; blood-brain barrier penetrating capability, degree of serotoninergic inhibition), the dose administered, and the patient's age and sex (34,35). Lastly, polymorphisms in genes related to dopamine receptors (such as DRD1, DRD2, DRD3) (63), dopamine transporters (SLC6A3), and dopamine-metabolizing enzymes (such as monoamine oxidase and catechol-O-methyltransferase) have been associated with individual variations in response to anti-psychotic treatment and the development of side effects, including hyperprolactinemia (64).

 

Although direct evidence establishing the involvement of adrenergic receptors in hyperprolactinemia caused by antipsychotic and antidepressant medications remains unproven, indirect indications, as elucidated in Figure 1, suggest the potential implication of these receptors. It is plausible that adrenergic receptors might play a partial role in the hyperprolactinemia induced by these medications.

           

Table 1. Medications and Their Ability to Cause Hyperprolactinemia

Cluster Name

Subclass mechanism of action

Medications

Prolactin increment

Frequency of prolactin increment (61,65)

Anti-psychotics

First generation anti-psychotics

Antagonize/block dopamine receptors, especially D2 receptors. Can block α1 adrenergic receptors.

Butaperazine

UP to 2-3-fold normal range with dozes 60 mg/daily. Higher in women (66)

High

Chlorpromazine

Up to 3-fold with initiation of treatment, up to 2-fold in long-term treatment) (67)

Moderate/ High

 

Flupenthixol

Up to 2-3-fold during the first month, and normalization in the next few months (68)

High

Fluphenazine

Up to 3-fold with initiation of treatment, up to 2-fold in long-term treatment (67). Up to 40-foldof the upper end of the normal range (69)

High

 

 

Haloperidol

Up to 9-fold at the beginning of treatment (3-fold in long-term treatment) (70)

High

Loxapine

Up to 3-foldof the upper end of the normal range in women (66)

Moderate

Perphenazine

Up to 40-fold of the upper end of the normal range (69)

Moderate

Pimozide

?

Moderate

Prochlorperazine

?

?

Promazine

Up to 4-fold of the upper end of the normal range (69)

 

Thiordiazine

Up to 3-fold with initiation of treatment, up to 2-fold in long-term treatment) (67)

High

Thiothixene

Up to 3-fold with initiation of treatment, up to 2-fold in long-term treatment (71)

Moderate/ High

 

Trifluoperazine

?

Moderate

Veralipride

Up to 10 time increment, transient (72)

High

Zuclopenthixole

?

?

Second generation anti-psychotics

Dopamine receptors blockade especially D2 receptors, serotonin (5-HT) receptor blockade, glutamate modulation, can antagonize α1 or α2 adrenergic receptors and histamine receptors.

Amisulpiride

 

Up to 10-fold at the beginning of treatment and remained elevated during treatment but lower levels (68)

Case reports

Aripiprazole

Reduce prolactin levels (73)

Case reports / No effect/ Reduced prolactin

Asenapine

Up to 2-fold increment and rarely with higher doses up to 4-fold (35)

Low, Moderate

 

 

Brexpiprazole

Mild increment (74)

 

Low

Clozapine

Mild (up to 2-fold) and transient (75)

 

Case reports or No effect

Iloperidone

Mild increment, transient (76)

Case reports or No effect

Levosulpiride

Up to 15-fold normal range (77)

Case reports / Moderate for galactorrhea (78)

 

Lurasidone

Up to 10-fold normal range (79)/ no effect (80)

Case reports or No effect

Molindone

?

Moderate

 

Olanzapine

Mild (up to 2-fold) and transient (75)

Low

 

Paliperidone

2-10-fold for depot formulations (81)

High

Perospirone

None (82)

None/ Case reports

Quetiapine

Mild and transient (75)

Low

 

Risperidone

2-10-fold

High (83)

 

Sertindole

Mild and transient (75)

?

Sulpiride

Up to 6-7-fold from baseline, dose dependent effect (84)

High

 

Thiethylperazine

?

?

Ziprasidone

Up to 4-fold from baseline and transient (35,75)

Low

 

Neuroleptic-like medications

 

Block D2 receptors

Domperidone

Up to 10-fold (85,86)

High

Droperidol

Significant increment after 10 minutes of administration, with peak at 20 minutes (87)

?

Metoclopramide

Up to 15-fold (2)

High

 

Anti-depressants

TCAs

Block the reuptake of both serotonin and noradrenaline.

Amitriptyline

2-fold increment on dosage 200/300mg (88)

Low

Amoxapine

3,5-fold to baseline (89)

High

Clomipramine

Up to 3-fold increment from baseline (90)

High

Desipramine

Just above the normal limit with 100 mg oral administration (91)

Low, Controversial

Imipramine

Up to 4-fold normal range (69)

Controversial

Nortriptyline

2-fold in the first 2 weeks in one patient (88)

None or Low

SSRI

Block the reuptake of serotonin.

Citalopram/

Escitalopram

Up to 3-fold increment (52)

None or Low (rare reports), Controversial data

Fluoxetine

Fluvoxamine

Paroxetine

Sertraline

SNRI

Block the reuptake of both serotonin and noradrenaline.

Duloxetine

Up to 2-fold normal range (92)

Case reports

Milnacipran

Not increased risk of hyperprolactinemia (93)

None

Venlafaxine

Up to 2-fold normal range, dose related (94)

Case reports

MAO inhibitors

Inhibit the enzyme. Monoamine oxidase, which breaks down serotonin, noradrenaline, and dopamine, though increasing their levels.

Clorgyline

Up to 2-fold from baseline (95)

Low

Pargyline

Up to 3-fold from baseline (95)

Low

Phenelzine

Unclear elevation, galactorrhea (96)

Low/ Case reports

Atypical anti-depressants

Inhibit noradrenaline and dopamine reuptake.

Bupropion

No significant change (80)

Case reports

Increases the release of both serotonin and noradrenaline.

Mirtazapine

No significant change (80)

Case reports

Serotonin modulators

Modulate serotonin receptors in the brain to enhance serotonin transmission.

Indoramine

 (97)

Case report

Nefazodone

Mild increment from baseline only at acute administration (98)

None/ Case reports

Trazodone

Up to1.5-fold from baseline (99)

None, Low

Vortioxetine

Up to 2-fold elevation (100)

Case reports

Selective noradrenaline reuptake inhibitor

Inhibit reuptake of norephinephrine.

Reboxetine

Up to 2-fold from baseline (101)

Case reports

NMDA receptor antagonist

Block NMDA receptors though influencing glutamate neurotransmission.

Esketamine

?

None

Gastric acid reducers

H2 receptor antagonists

H2 receptor antagonists.

Cimetidine

Up to 3-fold after 400 mg IV infusion (102)

Low

Ranitidine

Mild increment only in high IV doses (103)

Low

Protein pump inhibitors (PPIs)

Inhibit the activity of the proton pump (H+/K+ ATPase) in the stomach's parietal cells.

Esomeprazole

?

Case reports or No effect

Lansoprazole

4-fold increment from baseline (104)

Omeprazole

No significant change (105)

Pantoprazole

No significant change (106)

Rabeprazole

No significant change (107)

Opioids

 

They activate opioid receptors. Main types of opioid receptors: mu (μ), delta (δ), and kappa (κ).

Apomorphine

By acting as dopamine agonist it lowers prolactin (108)

None

Heroin

Elevated in addiction (within normal range) compared to healthy control or during abstinence (109)

Moderate in addicted patients that have values over 25 ng/mL

Methadone

Mild increment, transient increases for several hours following the administration (110)

?

Morphine

Up to 2-fold increment from baseline (111)

High

Antihypertensives

 

It decreases the release of noradrenaline.

Methyldopa

3-4-fold (65) up to 40-fold normal range (69)

Moderate

Inhibit the storage of neurotransmitters like noradrenaline and serotonin in nerve cells, though decreasing their release.

Reserpine

2.5-fold increment from baseline (112) Up to 40-fold normal range (69)

High

Block calcium channels in cardiac and smooth muscle cells.

Verapamil

2-fold (113)

Low

Estrogens

 

By using as contraceptives they suppress sexual axis.

Estradiol infusion

3-4-fold, dose-dependent, way of administration is important (oral and IV) (114)

Low

 

Estradiol withdrawal

?

 

Gonadotropins and GNRH agonists

 

Same as endogenous components, used for fertility induction.

hCG

Up to 4-fold increment, transient (115)

High

hMG

Up to 2.7-fold increment from baseline, transient (116)

High, Transient

GnRH agonist.

Leuprolide acetate

1.5-fold higher prolactin in compared to hMG alone, transient (117)

High

Other drugs

Benzodiazepines

Enhances the effects of GABA in the brain.

Diazepam

Mild, dose-dependent (118)

Controversial

Anxiolytics

Serotonin receptor agonist.

Buspirone

2-fold (119)

Case report or No effect

α-2 adrenergic agonist.

Clonidine

?

Case reports

Anticonvulsant

Block sodium channels in nerve cells.

Carbamazepine

Less than 2-fold in sleep entrained (120,121)

?

Phenytoin

Controversial, it can also lower prolactin levels (122)

?

Enhances the effects of GABA in the brain.

Phenobarbital

Controversial (123)

 

Valproic Acid

Controversial, it can also lower prolactin (124)

Case reports

Mood stabilizer

Decrease dopamine release and glutamate, increase GABA inhibition.

Lithium Carbonate

Controversial, no effect (183)

None

Antimigraine medication

Calcium channel blocker.

Flunarizine

Mild increment, up to 1.5-fold from baseline (125)

Case reports

Weight loss medications

Increase the release of serotonin and inhibit its reuptake.

Fenfluramine

Mild increment within normal range in previews non-hyperprolactinemic patients (126)

High

Inhibit the reuptake of serotonin, noradrenaline, and dopamine.

Sibutramine

4-fold (2,127)

Case report

Anticholinesterase inhibitors

Reversible acetylcholinesterase inhibitor.

Physostigmine salicylate

Less than 100 ng/mL (44).

Low

Prokinetic medication

Stimulate serotonin receptors in the gut.

Cisapride

High increment (up to 200 ng/dL) but in co-administration of other drug inducing hyperprolactinemia (128)

Case reports

Antihistaminic with sedative and antiemetic properties

Block histamine receptors.

Promethazine

?

?

Central Nervous System Stimulants

Increase the release and reduce the reuptake of noradrenaline and dopamine in the brain.

Amphetamine

Mild, only during withdrawal (129)

?

Methylphenidate

No effect (130)

Case reports/ No effect

ADHD medication

α -2 adrenergic agonist.

Guanfascine

Controversial, it can also lower prolactin (131)

Case reports

Decongestant

Sympathomimetic amine, predominantly α-1 agonist

Pseudoephedrine

Lower prolactin levels (132)

Case reports

Rheumatoid arthritis medications

Reduce inflammation, modify immune response.

Bucillamine

Mild increment within normal range (133)

Case report

Penicillamine

?

Case reports

Osteoporosis medication

Monoclonal antibody that inhibit the receptor activator of nuclear factor kappa-B ligand (RANKL).

Denosumab

?

Case reports

Substance of abuse

Blocks the reuptake of noradrenaline, dopamine, and serotonin in the brain.

Cocaine

Decrease prolactin levels (134)

Mild increment only during withdrawal (129)

Case reports

Increases the release and inhibits the reuptake of serotonin and to some extent, dopamine and noradrenaline.

Ecstasy

Mild or no effect (135)

?

Stimulates nicotinic acetylcholine receptors, leading to the release of neurotransmitters like dopamine and noradrenaline.

Smoking

Mild increment, transient (136)

Moderate

Anti-HIV medications

Protease inhibitors that prevent the cleavage of viral proteins and thereby inhibiting viral replication.

Ritonavir / Saquinavir

Mild (137)

Case reports

Radiotherapy

Use of high-energy radiation to damage the DNA within the targeted cells.

Intracranial radiotherapy

?

Moderate (138)

             

Frequency of increase to abnormal prolactin levels with chronic use: high: >50%; moderate: 25 to 50%; low: <25%; none or low: case reports. The effect may be dose-dependent. Drugs marked with blue have controversial data or decrease prolactin levels as explained in the table. Where we could not identify reliable data for the parameters in the table we added a question mark. *First-generation anti-psychotics, non-selective dopamine receptors antagonists. **Second-generation anti-psychotics.

 

Anti-Psychotics

 

Anti-psychotics are traditionally classified as first- and second-generation, but more recently a new classification taxonomy has been developed by McCutcheon et al. to express different receptor affinity of different anti-psychotics. Due to the impossibility to include in this new classification all drugs that cause hyperprolactinemia, we have used the old classification (Table 1) (139,140).

 

The first-generation anti-psychotics are typically associated with more severe hyperprolactinemia (2-3-fold increment), whereas second-generation drugs have lower D2 affinity and stronger blockade of 5HT2A receptors leading to milder prolactin elevations (1-2-fold), except risperidone, paliperidone, and amisulpiride. Amisulpiride has the greatest potential to cause hyperprolactinemia of all anti-psychotics (4).

 

The first-generation anti-psychotics, such as fluphenazine and haloperidol, act as non-selective dopamine receptors antagonists (2,10). The therapeutic effects on psychotic symptoms occur through D2 and D4 receptor binding in the mesolimbic area, while side effects are mediated by D2 blockade in the striatal area (linked to extrapyramidal effects) and in the hypothalamic infundibular system (linked to hyperprolactinemia) in more than 50% of patients. A clinical trial involving 69 patients examined the effects of various anti-psychotic medications on prolactin levels, including chlorpromazine, depot haloperidol, fluphenazine, zuclopenthixol, sulpiride, pimozide, droperidol, and flupenthixol. The study found a significant elevation in prolactin levels only in females, with a mean level of 1106 mIU/L (52 ng/mL) compared to the normal range of <480 mIU/L (22.6 ng/mL). In males, the mean prolactin levels were within the normal range, which may be attributed to the significantly lower total daily dose of chlorpromazine used in males (199.0-220.1 mg/day) compared to females (384.4-302.48 mg/day, P<0.05) (7).

 

Second-generation anti-psychotics with lower D2 affinity led to milder prolactin elevations (1-2-fold), except for paliperidone, risperidone, and amisulpiride whose effect on prolactin is similar to the first-generation neuroleptics. Chlorpromazine, loxapine, olanzapine and quetiapine have variable effects on prolactin secretion, while aripiprazole, clozapine, iloperidone, lurasidone have little or no effect on prolactin secretion (35).

 

An important factor contributing to variations in the induction of hyperprolactinemia by different anti-psychotic medications is the blood-brain barrier. Permeability glycoprotein transporter (P-gp), coded by the ABCB1 gene, is expressed in various tissues including in the cells of the blood-brain barrier. P-gp plays a role in actively transporting hydrophobic drugs with a molecular weight greater than 400 Da out of the brain, thus protecting the brain from these medications; therefore, this protein can change drug bioavailability (141,142).

 

The affinity of risperidone, paliperidone, and amisulpiride (prolactin rises up to 10-fold with these drugs) for P-gp is approximately twice that of olanzapine and chlorpromazine (prolactin rise is up to 3-fold with these drugs), and four times greater than haloperidol and clozapine (prolactin rise can be high initially but usually reduces with time) (143). The higher affinity of risperidone, paliperidone, and amisulpiride to P-gp could, among other mechanisms, partly explain the greater induction of hyperprolactinemia by these drugs, as P-gp does not allow them to enter the brain via the blood-brain barrier. Therefore, the portal circulation of the anterior pituitary delivers a somewhat higher concentration of these drugs to the lactotrophs, which are located outside the blood-brain barrier, to inhibit the D2 receptors (144).

 

Aripiprazole can act as a partial agonist at D2 receptors and display partial agonist activity at 5HT1A receptors, while also acting as an antagonist at 5HT2A receptors. Antagonism at these receptors can help to normalize prolactin levels since 5HT2A receptor activation has been associated with increased prolactin release. That is why it is considered a prolactin secretion modulator (145). Its role in prolactin levels has been investigated in a study involving both retrospective and prospective components (146). The retrospective part of the study included 30 patients undergoing risperidone treatment, when it was observed that after 6 months of treatment, prolactin levels remained high although somewhat lower than at the start of observation. In the prospective part of the study, 30 other patients were divided into two groups: one group receiving risperidone alone at a daily dosage of 2-4 mg and the other group receiving a combination of risperidone and aripiprazole at a daily dosage of 5-10 mg. The group receiving adjunctive aripiprazole exhibited significantly lower serum prolactin levels compared to the risperidone-only group at weeks1 (914±743 vs 1567±1009 mU/L), 2 (750±705 vs 1317±836 mUI/L) and 6 (658±590 vs 1557±882 mUI/L). Notably, during aripiprazole treatment, prolactin levels at weeks 1, 2, and 6 were significantly lower than at baseline (P< 0.05) (at baseline patients were treated with risperidone as monotherapy), suggesting that aripiprazole may effectively alleviate risperidone-induced hyperprolactinemia. Similar findings supporting the role of aripiprazole in reducing prolactin levels have been reported in other studies (147). Combination therapy presents a promising therapeutic approach for adjunctive treatment or for transitioning from risperidone to mitigate hyperprolactinemia (146).

 

More recently, a new medication SEP-363856, a trace amine-associated receptor 1 (TAAR1) and 5HT1A agonist, has been developed to treat schizophrenia. Its mechanism of action is not based on D2 antagonism, and has a favorable effectiveness and tolerability profile, without causing hyperprolactinemia (148). This category of medication serves as compelling evidence for the significant involvement of dopamine receptors in drug-induced hyperprolactinemia, and it is a future viable therapeutic choice for patients experiencing adverse effects associated with hyperprolactinemia.

 

DRUG-INDUCED HYPERPROLACTINEMIA IN PEDIATRIC PATIENTS

 

Anti-psychotic medications have been found to induce hyperprolactinemia in the pediatric population as well as in adults. In a trial involving 35 children and adolescents with early-onset psychosis, primarily diagnosed with childhood-onset schizophrenia or psychotic disorder not otherwise specified, prolactin levels were measured after a 3-week washout period, as well as after 6 weeks of treatment with haloperidol, olanzapine, and clozapine (149). Following the 6-week treatment period, haloperidol (9 of 10 patients – mean age 13.4 years) and olanzapine (7 of 10 patients– mean age 15.9 years) resulted in prolactin levels above the upper limit of normal. The mean increase was 5.2-fold for haloperidol and 2.4-fold for olanzapine. The prolactin response did not show statistically significant differences between females and males treated with haloperidol and olanzapine. Clozapine (22 patients, mean age 14.7) caused a small but significant rise in females (1.2-fold) but levels remained in the normal range for all patients. There was no rise in males. Why this difference between females and males is only on clozapine remains unclear and difficult to explain. However, in a study involving 36 girls aged 8-17 years, mean prolactin levels were higher in girls compared to boys, with the most significant increase occurring around the age of 13y, correlating with menarche. A highly significant correlation was found between increases in plasma prolactin and estradiol levels between the ages of 11 and 13 years. Girls with long menstrual cycles (>28 days) between the ages of 14 and 16 years had higher prolactin levels (p<0.05) (150). Even though the mean age of patients on clozapine was above 13 years, we do not possess information on the duration of the menstrual cycle of those girls, as if it is longer, the physiologic estrogenization because of the longer menstrual cycle can impact the range of prolactin elevation. In any case, the population sample size was relatively small to draw definitive conclusions and to provide answers why this happens only in clozapine patients (149).The authors concluded that the prolactin response in male children and adolescents treated with haloperidol or olanzapine was significantly higher than that observed in adult males. However, the prolactin response in female children and adolescents after haloperidol treatment did not differ significantly from that of adult females in similar studies, possibly due to the adult similarity of estrogen status seen in female adolescents (149). Aripiprazole in another study showed a lesser prolactin increase than olanzapine, quetiapine, and risperidone, similar to the adult population (151).

 

In another clinical trial involving 396 children and adolescents (aged 14.0 ± 3.1 years), the impact of anti-psychotic medications on prolactin levels was studied. The medications involved risperidone, olanzapine, quetiapine, and aripiprazole. Risperidone caused the highest incidence of hyperprolactinemia (93.5%) and had the highest peak prolactin levels (median = 56.1 ng/mL) followed in order by olanzapine, quetiapine, and aripiprazole. Menstrual disturbances were the most prevalent side effect (28.0%), particularly with risperidone (35.4%). Notably, severe hyperprolactinemia was associated with decreased libido, erectile dysfunction, and galactorrhea (152).

 

In conclusion, a comprehensive meta-analysis comprising 32 randomized controlled trials with a total of 4643 participants, with an average age of 13 years, has demonstrated that risperidone, paliperidone, and olanzapine are associated with a significant increase in prolactin levels among children and adolescents. Conversely, aripiprazole is linked to a notable decrease in prolactin levels in this age group. It is worth noting that haloperidol was not included in these studies, resulting in an absence of evidence regarding its prolactin-related effects in this population (153).

 

These findings underscore that haloperidol, risperidone, paliperidone and olanzapine are potent inducers of hyperprolactinemia in children and adolescents, mirroring observations in the adult population. A comprehensive listing of medications associated with hyperprolactinemia in children can be found in Table2.

 

Table 2. Drugs Reported to Induce Hyperprolactinemia in Children and Adolescents

Medication class

 

High

>50 percent of patients

Moderate

25-50 percent of patients

Low

<25 percent of patients

Case reports

Anti-psychotics, first-generation 'typical'

Fluphenazine (154)

Haloperidol (149,155)

 

Chlorpromazine (156)

Loxapine (157)

Pimozide (158,159)

 

 

Anti-psychotics, second-generation 'atypical'

Paliperidone (160,161)

Risperidone (152,155,162–164)

 

Asenapine (165)

Molindone (166)

Olanzapine (149,152)

Lurasidone (167,168)

Ziprasidone (169)

Quetiapine (152,162)

 

 

Clozapine (149)

Aripiprazole* (152,170)

Amisulpride (171)

Brexpiprazole (172)

Anti-depressants

Clomipramine (173)

 

 

Desipramine (174)

Bupropion (175)

Citalopram (176)

Escitalopram (177)

Fluoxetine (178)

Sertraline (179)

Duloxetine (177)

Paroxetine (180)

Venlafaxine (181)

Anti-emetics and gastrointestinal medications

Metoclopramide (182–184)

Domperidone (185,186)

 

 

 

Omeprazole (187)

Lansoprazole (187)

Cisapride

Others

Fenfluramine (188)

 

 

Estrogens (189)

Triptorelin (190)

Clonidine (191)

Methylphenidate (181)

Guanfacine (181)

Valproic acid (181)

Penicillamine (181)

*Aripiprazole is a partial agonist at the type 2 dopamine receptor and display partial agonist activity at the type 1A serotonin receptor (5HT1A) and antagonist at 5HT2A receptor. It can be used in combination with other psychotropic medications to reduce prolactin levels. Aripiprazole itself can sometimes cause mild hyperprolactinemia (192).

 

CLINICAL MANAGEMENT OF ANTI-PSYCHOTIC-INDUCED HYPERPROLACTINEMIA

 

Drug-induced hyperprolactinemia should be considered in the differential diagnosis of elevations of prolactin levels, sometimes greater than 200 µg/L (4260 mIU/L). Particularly when serum prolactin levels exceed 80-100 µg/L (1700-2130 mIU/L), pituitary magnetic resonance imaging (MRI) should be performed to rule out the presence of any underlying pituitary or hypothalamic masses that may contribute to hyperprolactinemia (193). According to the guidelines from the Endocrine Society, in symptomatic patients suspected of having drug-induced hyperprolactinemia, it is recommended the first test to diagnose drug-induced hyperprolactinemia is the discontinuation of the medication for 3 days or switch to an alternative drug (e.g. a prolactin-sparing anti-psychotic (e.g. aripiprazole), or an anti-psychotic with lower dopamine antagonist potency (Table 1) followed by retesting of serum prolactin levels.

 

However, any discontinuation or substitution of anti-psychotic agents should be done in consultation with the patient's psychiatric physician. If discontinuation is not possible or if the onset of hyperprolactinemia does not coincide with therapy initiation, obtaining a pituitary MRI is recommended (despite prolactin levels) to differentiate between medication-induced hyperprolactinemia and hyperprolactinemia caused by a pituitary or hypothalamic mass (194).

 

Before initiating treatment with an anti-psychotic medication, it is advised that clinicians inquire about the patient's previous treatment experience, sexual dysfunction, menstrual history (including irregularities and menopausal status), as well as any history of galactorrhea. Additionally, obtaining a baseline prolactin level is recommended before starting treatment (195). This pre-treatment screening for hyperprolactinemia can help determine whether subsequently elevated prolactin levels are due to medication-induced factors (196) and make the diagnosis easier without the need to perform further imaging.

 

While treatment of hyperprolactinemia in patients receiving anti-psychotics may not always be necessary, in cases where clinical hypogonadism is evident, several options are available (193). The initial step is to discontinue the drug if clinically feasible. If discontinuation is not possible, switching to a similar anti-psychotic that does not cause hyperprolactinemia is suggested. If neither of these options is feasible, cautious administration of a dopamine agonist may be considered in consultation with the patient's physician (194). It is worth noting that these interventions only result in the normalization of prolactin levels in approximately half of the patients receiving anti-psychotics, and careful psychiatric monitoring is required due to the possibility of psychosis exacerbation with dopamine agonists (193).

 

For patients with long-term hypogonadism demonstrated by hypogonadal symptoms or low bone mass, the use of estrogen or testosterone replacement is recommended. In rare instances where patients receiving anti-psychotics also present with a pituitary tumor, treatment options primarily revolve around tumor-specific interventions, considering especially optic chiasm compression. When a non-functioning tumor is suspected, the above options to eliminate the drug-induced component of hyperprolactinemia should be considered, with supervised short-term cessation of the medication to clarify drug-induced or tumor-derived hyperprolactinemia (193).

 

In addition to the interventions mentioned earlier, addressing fertility concerns in patients with drug-induced hyperprolactinemia may require further measures. Normalization of prolactin levels through medication adjustment or dopamine agonist therapy can often lead to the restoration of fertility. In situations where fertility is not spontaneously regained, fertility treatment with gonadotrophins or assisted reproductive procedures may be necessary.

 

NEUROLEPTIC-LIKE MEDICATIONS

 

Metoclopramide and domperidone are anti-emetic and gastrointestinal motility agents known also as neuroleptic-like medications which can increase pituitary prolactin secretion and breast milk production by a dopamine antagonistic action (Figure 1).

 

Metoclopramide is a central and peripheral D2 receptor antagonist (197). Its administration is followed by an acute increase in prolactin levels up to 15-fold above the baseline that persists in chronic administration of the drug (2). Even though the majority of patients treated with metoclopramide develop hyperprolactinemia (Table 1), related symptoms such as amenorrhea, galactorrhea, gynecomastia, and impotence remain unclear (2).

 

Domperidone is a peripheral D2 antagonist (it does not cross the blood-brain barrier) used for treating intestinal motility disorders, especially for the prevention of gastrointestinal discomfort with dopaminergic treatment in Parkinson's disease (198), as it antagonizes the D2 receptors in the upper gastrointestinal tract. It also reaches the D2 receptors on lactotroph cells inducing hyperprolactinemia, and can be used for stimulating lactation, for example in women with preterm infants, or an adoptive parent (199,200), and even in transgender women who wish to breastfeed (201,202). It can cause a 10-fold elevation in prolactin levels with normalization of prolactin levels after three days (85,86). Neuroleptic-like medications are summarized in Tables 1 and 3.

 

Table 3. H2 Receptor Antagonists, Opioids, Anti-Hypertensives, PPIs, Estrogens, and Other Drugs and Their Ability to Cause Hyperprolactinemia.

Medication class

High

>50% of patients

Moderate

25-50% of patients

Low

<25% of patients

Case reports

Anti-emetic and gastrointestinal

Domperidone

Metoclopramide

 

Prochlorperazine

 

Esomeprazole

Omeprazole

Lansoprazole

Cisapride

H2-receptor antagonists

 

 

Cimetidine

Ranitidine

 

Anti-hypertensives

 

Methyldopa

Verapamil

 

Others

Fenfluramine

Opioids

 

Estrogens

Protease inhibitors

Cocaine Bucillamine

Clonidine

Methylphenidate

Guanfascine

Valproic Acid

Penicillamine

 

ANTI-DEPRESSANTS

 

Anti-depressants can be classified based on their structure and mechanism of action into tricyclic anti-depressants (TCAs), selective serotonin reuptake inhibitors (SSRIs), serotonin-noradrenaline reuptake inhibitors (SNRIs), monoamine oxidase (MAO) inhibitors, atypical anti-depressants, serotonin modulators, selective noradrenaline reuptake inhibitor, and NMDA (N-Methyl-D-Aspartate) receptor antagonists (203). Data on their ability to cause hyperprolactinemia are controversial, as described below. The two main mechanisms, both related to elevated serotoninergic tonus, explain how anti-depressants can induce hyperprolactinemia by indirect modulation of prolactin release by serotonin and serotonin stimulation of GABAergic neurons (50) Adrenergic receptors involvement in their ability to cause hyperprolactinemia remains unclear. (Figure 1).

 

Data on the incidence of hyperprolactinemia with anti-depressant medications, especially SSRIs, MAO inhibitors, and some TCAs, suggest that they can cause modest and generally asymptomatic hyperprolactinemia (4,193). Their ability to cause hyperprolactinemia is summarized in Table 1.

 

TCAs

 

TCAs, such as amitriptyline, desipramine, clomipramine, and amoxapine, can induce sustained mild hyperprolactinemia (2) (Table 1). They manifest their mechanism of action by blocking the reuptake of noradrenaline and serotonin, through increasing serotoninergic tonus, leading to hyperprolactinemia. Amitriptyline's ability to cause hyperprolactinemia seems to be dose-dependent. A dosage of 150-250 mg/day to 5 patients showed no effect on prolactin levels after 3-7 weeks (204), whereas a dosage of 200-300 mg/day caused a 2-fold increment of prolactin levels in two of nine chronic treated patients (88). In 13 patients with depression taking amitriptyline or desipramine, prolactin levels were studied after intravenous injection of tryptophan (serotonin precursor). It was observed that tryptophan-induced prolactin elevation was significantly increased compared with a preceding placebo period (205). A similar tryptophan test was performed with clomipramine 20 mg vs placebo in 6 normal subjects. Levels of prolactin increased after tryptophan infusion in pretreated patients with clomipramine (206),suggesting serotonin involvement in this hyperprolactinemia.

 

The effect of the traditional TCA imipramine on serum prolactin levels is controversial. A five-week double-blind study on patients with depression taking imipramine did not show any significant change in serum prolactin (207). However, another study in young healthy men showed that imipramine’s effect on prolactin is dose-dependent (usually therapeutic dose is 50-150 mg): oral administration of 100 mg, but not of 40 mg, led to a consistent rise in prolactin levels after 3 hours of administration, but the rise was mild (maximum 25.85 ng/mL (550 mUI/L) (91).

 

Data regarding nortriptyline's ability to induce sustained hyperprolactinemia are lacking. Anyway, over a 4-6 week treatment of 8 patients with nortriptyline up to 150 mg/daily, no difference was found between placebo and the medication group. In only one patient was observed a transitional 2-fold elevation in the first 2 weeks (88).

 

Amoxapine, which is an anti-depressant with neuroleptic properties as well, is found to increase prolactin levels in 10 patients approximately 3.5-fold compared to baseline and more than desipramine in 12 patients, where almost no difference with baseline was observed (89). The proposed mechanism of hyperprolactinemia involves the blockade of the D2 receptor in tuberoinfundibular neurons or the anterior pituitary gland (2).

 

SSRIs

 

SSRIs enhance serotonin activity via inhibition of neuronal serotonin reuptake. This could be the most prominent mechanism leading to a prolactin elevation. A review of 13 case reports showed prolactin levels between 28 and 60 ng/mL (595 and 1276 mUI/L) (52). In a French study, 27 of 159 cases (17%) had SSRI-induced hyperprolactinemia with sertraline being the most prominent, followed by fluoxetine, paroxetine and fluvoxamine. Only citalopram was found not to increase prolactin levels significantly (208). However, in another study fluoxetine, paroxetine, and fluvoxamine were found again to elevate prolactin levels, but they also found citalopram to induce hyperprolactinemia. In this study duloxetine, milnacipran, and sertraline (which was the most prominent in the previous study) were not associated with an increased risk of hyperprolactinemia (93). Fluoxetine-induced hyperprolactinemia was found in patients with major depression (4.5% of men and 22.2% of women) following 12 weeks of fluoxetine treatment (209).Differences in the ability to cause hyperprolactinemia can be attributed to the variations in the affinity of the SSRIs for dopamine, histamine, and GABA receptors.

 

The Nurses’ Health Study and its follow-up study assessed anti-depressant use and circulating prolactin levels in 610 women (including 267 anti-depressant users) with two measurements of prolactin an average of 11 years apart (210). In this study, mean prolactin levels were similar among SSRI users (13.2 µg/L, (280 mUI/L), 95% CI 12.2-14.4), users of other classes of anti-depressants (12.7 µg/L (270 mUI/L), 95% CI 11.0-14.6), and non-users (13.1µg/L, (278 mUI/L), 95% CI 12.8-13.4) (210). However, the duration and dosage of anti-depressant use at the time of prolactin sampling had not been assessed, as the participants had only responded as current anti-depressant users/non-users on a questionnaire.

 

MAO Inhibitors

 

Monoamine oxidase is an enzyme responsible for breaking down neurotransmitters such as serotonin, noradrenaline, and dopamine in the brain. Inhibition of this enzyme is expected to increase the levels of all those neurotransmitters. Even though increased dopamine is suspected to be related to lower prolactin levels, probably the serotonin increment prevails and that is why this class of medications is related to hyperprolactinemia; or dopamine and serotonin increment neutralize each other and no difference in prolactin levels is seen.

 

MAO inhibitors with serotoninergic activity (pargyline and cordyline) can cause modest and generally asymptomatic hyperprolactinemia (2,61). Phenelzine was observed to persistently increase prolactin levels in 4 of11 patients, which returned to normal during a placebo week and rose again in all 4 patients after treatment restart (88).

 

Atypical Anti-Depressants

 

Mirtazapine's mechanism of action is different from other anti-depressants, as it does not inhibit the reuptake of serotonin or noradrenaline, but it increase the release of serotonin and noradrenaline (211). In any case, prolactin levels did not show any difference in 8 healthy male subjects pre- and post-mirtazapine 15 mg oral administration (212).

 

Serotonin Modulators

 

Trazodone acts through dual inhibition of serotonin reuptake and serotonin type 2 receptors, coupled with antagonism of histamine and α-1-adrenergic receptors (213). In 12 patients with depression, 150 mg of trazodone for 3 weeks caused significantly higher prolactin levels after 12 hours, 1 week, and 2 weeks of treatment compared to baseline. Higher levels were after the first week 15,3+/-8,5 ng/mL (325 +/- 180 mUI/L) compared to baseline 9,1 +/- 5,6 ng/mL (194+/-119 mUI/L) (99).

 

Selective Noradrenaline Reuptake Inhibitor

 

Reboxetine is a selective noradrenaline reuptake inhibitor that was shown to increase prolactin in healthy men after acute administration, but this effect can be reversed if reboxetine is simultaneously administered with α2-blocker mirtazapine, suggesting a role of α2-receptors in the enhancement of prolactin release after reboxetine (214). In any case, conflicting data are present even with this drug, with one other study showing no difference between pre-treatment and after-treatment prolactin levels in patients taking up to 8 mg reboxetine for 4 weeks in 17 patients (215). This discrepancy can be due to the limited number of patients or other neuroendocrine mechanisms still less explored.

 

SNRI medications are described to cause only mild and rare elevations on prolactin levels, whereas (94), esketamine (NMDA receptor antagonist)is not described to cause hyperprolactinemia.

 

Summary

 

In summary, controversial data are available on anti-depressant-induced hyperprolactinemia. Routine monitoring of prolactin levels in patients taking anti-depressants is not recommended unless symptoms related to prolactin increase (in premenopausal women: menstrual cycle dysfunction leading to amenorrhea, oligomenorrhoea, anovulatory cycles, low libido, and energy; in men: erectile dysfunction, decreased energy and libido, decreased muscle mass, decreased body hair; and for both of them osteopenia, galactorrhea and infertility) occur (50,193). If the prolactin serum level is elevated (> 25 µg/L (531 mUI/L), in these patients a differential diagnosis is needed. The proposed approach is to withdraw the anti-depressant drug slowly over 2 weeks and replace it with another anti-depressant less likely to cause hyperprolactinemia, then reassess symptoms and prolactin levels after 2-4 weeks (193). If the serum prolactin remains elevated, other causes of hyperprolactinemia should be addressed by an endocrinologist. Another approach is to perform a pituitary MRI if the replacement of the anti-depressant is difficult to manage.

 

GASTRIC ACID REDUCERS

 

Histamine-Receptor Inhibitors

 

Histamine, a CNS neurotransmitter, binds to both H1 and H2 receptors. It can stimulate prolactin secretion via H1 receptors by inhibiting the dopaminergic system. On the contrary, histamine can also inhibit prolactin secretion via H2 receptors using a non-dopaminergic mechanism involving β-endorphin, vasoactive intestinal peptide, vasopressin, or TRH (216), all of which act as prolactin-releasing factors (49) (Figure 1).

 

In the French Pharmacovigilance Study, H2 receptor antagonists were found to contribute to 5% of drug-induced hyperprolactinemia, primarily with ranitidine (odds ratio = 4.43; 95% CI: 1.82–10.8) (11). Other studies have shown that H2-receptor antagonists such as cimetidine and ranitidine can elevate prolactin levels (217,218). Specifically, cimetidine caused a three-fold increase in prolactin levels after a 400 mg IV infusion, although this effect was not observed with oral administration of 800 mg cimetidine in healthy individuals (102) (Table 1,3).

 

Interestingly, it has been observed that systemic administration of the H2 agonist impromidine does not prevent cimetidine-induced hyperprolactinemia. In contrast, pre-administration of benzodiazepines or GABA lowered the prolactin response. This suggests that cimetidine-induced hyperprolactinemia may be mediated through neurotransmitters in the GABA-ergic system (219).

 

Proton-Pump Inhibitors

 

Proton-pump inhibitors (PPIs) have been recently reviewed regarding the risk of hyperprolactinemia and related sexual disorders observed with long-term use (220). The exact mechanism by which PPIs increase prolactin levels is not fully understood; possible explanations include inhibition of dopamine receptors, interference with other dopamine receptors, involvement of the serotoninergic pathway, modulation of the opioid pathway, and a potential role in decreasing prolactin clearance (220) (Figure 2). In addition, PPIs can increase gastrin levels in chronic use especially in females using high doses (221). As gastrin can act as prolactin-inhibitory factor (108), this antagonistic effect may explain the relatively mild hyperprolactinemia occurring with this class of drugs. Esomeprazole has a mild inhibitory effect on CYP3A4, which leads to decreased metabolism of estrogen, thereby increasing serum estrogen levels which can stimulate the production of prolactin (222). Gynecomastia, impotence, irregular menses, and galactorrhea have been described with PPI use. Hyperprolactinemia occurred less often than sexual disorders, and most cases of hyperprolactinemia were reported with omeprazole, esomeprazole, and lansoprazole use (e.g. 4-fold increment with lansoprazole) (104) (Table 1,3). Pantoprazole and rabeprazole were only sporadically associated with hyperprolactinemia. The authors assert that the occurrence of sexual dysfunction in individuals using PPIs, despite having normal prolactin levels, may be attributed to the development of low vitamin B12 levels, hypomagnesaemia and iron deficiency resulting from PPI usage. These nutritional deficiencies have been implicated in the manifestation of sexual disorders in other studies (220,223,224).

 

OPIOIDS

 

Endogenous opioids, morphine, and related drugs, activate ε-, μ-, κ- and δ- opioid receptors in the hypothalamus, modulating pituitary hormone secretion. They do not possess direct effects on pituitary cells (225,226). They inhibit the gonadal axis via ε receptors (GnRH suppression) and stimulate prolactin production by reducing the activity of tuberoinfundibular dopaminergic neurons via μ, κ and δ opioid receptors – mostly μ receptors as the μ receptor opioid antagonist naloxone prior to morphine and methadone use prevents opioid-induced hyperprolactinemia (227). Indirect stimulation of prolactin release can be mediated by stimulating prolactin-releasing factors production (the serotoninergic pathways discussed above) (228) (Figure 2). Hyperprolactinemia can then lead to additional gonadal axis suppression. In addition, opioids can modulate the corticotroph axis via κ and δ opioid receptors and the somatotroph axis via μ, κ and δ opioid receptors (226). Additionally, opioids manifest negative effects on bone health through direct inhibition of osteoblasts by opioids, gonadal axis suppression, altered mental status, and other comorbidities (chronic conditions, smoking, alcohol use) (229).

 

Acute intravenous or intra-ventricular administration of endogenous opioids leads to a rapid plasma prolactin increase in a dose-dependent manner (44,225). Chronic use of opioids effects on prolactin can vary: oral opioids for chronic pain increase prolactin, but morphine administered intrathecally for chronic non-cancer pain had no effect on prolactin (226). Hyperprolactinemia induced by opioids can be symptomatic: painful gynecomastia, galactorrhea, and hypogonadism have been reported in chronic opioid users. These can be alleviated with discontinuation or reduction of opioid dose, and sometimes dopamine agonists such as bromocriptine (226). Methadone induces a transient increase in prolactin levels, whereas chronic methadone users have normal basal prolactin levels (110), (Table 1, 3).

 

On the contrary, in a study involving six patients with hyperprolactinemia and amenorrhea, the use of naltrexone, an opioid antagonist, was investigated to determine if blocking endogenous opioids could improve the sexual axis. On the first day of naltrexone administration, significant increases were observed in the mean concentration of luteinizing hormone (LH), LH pulse amplitude, and estradiol levels compared to the control day. This indicated a prompt partial reactivation of the hypothalamic-pituitary-gonadal axis as a result of naltrexone, leading to heightened gonadotrophin levels and subsequent release of estradiol. However, it was found that the effect of opioid antagonism did not result in a sustained increase in estradiol secretion with chronic treatment. Additionally, prolactin levels continued to increase over time (mean prolactin level 255 ± 121 microgram/L), despite the initial improvement in the gonadal axis. This study demonstrated that although prolactin-induced suppression of the gonadal axis can be reversed to some extent by acute opioid antagonism, it is not an effective treatment for revitalizing the gonadal axis in the long term. Possible explanations for this lack of sustained effect include desensitization of the hypothalamic-pituitary unit for the effects of opioid receptor blockade and other disruptors of the axis, which may counteract the positive effects of opioid antagonism (230,231).

 

For endocrinopathies caused by opioids, including hyperprolactinemia, potential management choices include reducing or discontinuing opioid usage whenever feasible and exploring alternative pain relief therapies for chronic pain situations. Hormonal replacement therapy can be considered for hypogonadism and hypoadrenalism (226).

 

ANTIHYPERTENSIVES

 

Some antihypertensive medications including α-methyldopa, verapamil, labetalol, and reserpine, have been associated with hyperprolactinemia. This phenomenon is attributed mainly to the potential inhibition of dopaminergic pathways, highlighting the complex interplay between anti-hypertensive therapy and endocrine function. Other mechanisms are drug-specific and will be explained below.

 

α -methyldopa is an α-adrenergic inhibitor that leads to the suppression of monoamine synthesis, including noradrenaline, dopamine, and serotonin, which likely contributes to its anti-hypertensive effect. It causes hyperprolactinemia through the inhibition of dopamine synthesis by competitive inhibition of DOPA decarboxylase which transforms L-dopa into dopamine (61) (Figure 2). Long-term treatment resulted in elevated basal prolactin levels (3-4-fold), while a single dose of 750-1000mg reaches a peak of high prolactin level after 4-6 hours of administration (232). Gynecomastia is the most common endocrine side effect.

 

Calcium channel blockers are other drugs studied for their potential to cause hyperprolactinemia. The dihydropyridine class was found to have no effects on prolactin levels. Whereas, from the non-dihydropyridine class, which mainly blocks L-type of calcium channel receptors in the heart, only verapamil was found to cause 2-fold persistent hyperprolactinemia (and galactorrhea),while drug discontinuation reversed hyperprolactinemia in all patients (113). A clinical trial suggested that verapamil acts by reducing dopamine release in the tuberoinfundibular pathway through calcium influx inhibition (Figure 2), possibly by N-calcium channels which are known to be involved in the regulation of dopamine release and other neurotransmitters (233).

 

Labetalol is an α- and β-adrenoceptor blocker anti-hypertensive that has been reported to increase prolactin levels when administered intravenously, but not when administered orally (100 or 200 mg) as labetalol cross the blood-brain-barrier only in negligible amounts. The increase in prolactin release caused by intravenous labetalol is not readily explained by its interference with adrenergic receptors. The exact mechanism underlying this effect of the drug is currently unclear, but it is possible that labetalol's ability to block dopamine activity (anti-dopaminergic activity) might be involved in this response (234) (Figure 2). Pre-treatment with levodopa and carbidopa can prevent prolactin response after labetalol (235), suggesting dopamine pathway involvement suppression inside the blood-brain-barrier.

 

Reserpine is a rauwolfia alkaloid previously used for the treatment of hypertension as well as psychosis, schizophrenia, and tardive dyskinesia; it reduces dopamine by inhibiting their hypothalamic storage in secretory granules (236), and by blockade of vesicular monoamine transporter type 2 in monoamine neurons (237), leading to hyperprolactinemia. Prolactin levels are higher during treatment with reserpine than 6 weeks after discontinuation of the drug. Increased incidence of gynecomastia and breast cancer has also been reported among patients on anti-hypertensive therapy with reserpine (236). The ability of anti-hypertensives to cause hyperprolactinemia is summarized in Tables 1 and 3.

 

ESTROGENS

 

Estrogens stimulate prolactin secretion by several mechanisms: They bind to specific intracellular lactotroph cells receptors, though enhancing prolactin gene transcription and synthesis (238). They also inhibit tuberoinfundibular dopamine synthesis, stimulate lactotroph cell hyperplasia, downregulate dopamine receptor expression, and modify lactotroph responsiveness to other regulators (23,239) (Figure 2). Estrogen-induced hyperprolactinemia is dependent on the degree of estrogenization. Higher levels of estrogens in pregnancy and during ovulation increase prolactin levels with the last, contributing to a higher normal range of prolactin in pre-menopausal women.

 

Studies documenting the incidence of hyperprolactinemia showed that women on oral contraceptives were reported to have higher prolactin levels by 12% to 30% (240,241). Some, but not all, studies suggest that there is a dose-dependent effect (4). No increase in basal prolactin levels is reported during therapy with modern contraceptives with lower amounts of estrogen (242)or estrogen plus cyproterone acetate alone (243).

 

In transgender patients, estradiol or ethinyl estradiol treatment, the prolactin level rise was dependent on the dose of estrogen, duration of exposure, and alteration of SHBG levels. Estradiol infusion at levels above 10,000 pg/mL for as short as 6-7 hours significantly elevated prolactin levels by 3- to 4-fold, whereas ethinyl estradiol 2 mg/day for 1 month did not consistently elevate prolactin in all patients, which can be due to its ability to increase SHBG binding and maintaining free portion in the normal range (114) (Table 1, 3).

 

For women on post-menopausal hormone replacement therapies over 2.5 years, serum prolactin measured were within the normal range (244). In another study on 75 women, who were randomly assigned to three groups: control (receiving placebo), transdermal hormonal replacement (biphasic 17β-estradiol and progesterone, natural hormones), and oral ethinyl-estradiol and desogestrel, prolactin levels significantly increased in the oral group, but not in the transdermal group. There was a significant difference in hormone levels: in the oral group, estradiol levels increased five times and estrone levels eleven times. In the transdermal group, estrone and estradiol levels were increased three times (245).

 

GONADOTROPHINS AND GNRH AGONISTS

 

In addition to the known prolactin function in lactation, several studies have suggested other benefits of prolactin in oocyte development, formation of corpus luteum and its survival, steroidogenesis and implantation (246). In natural cycles there is a transient increase in late follicular phase of prolactin, but this increment is higher in stimulated cycles (246). In a cohort study were included 79 patients; 60 individuals underwent in vitro fertilization, 14 received clomiphene citrate treatment, and five patients with premature ovarian failure were administered estradiol. During the course of human menopausal gonadotrophin (hMG) treatment, a notable increase in both serum estradiol and prolactin concentrations were observed from early to late follicular days (P < 0.01). Specifically, prolactin levels increased from an initial mean value of 367±38 mIU/L (17.25±1.8 ng/mL) to 991±84 mIU/L (46.6±4 ng/mL) (Table 1). Bromocriptine effectively mitigated the increase in prolactin levels but was associated with a significant elevation in estradiol levels (P < 0.05) because prolactin itself works as a controller of estradiol increment. Clomiphene treatment led to a significant increase in serum estradiol levels (P < 0.01) but a significant decrease in serum prolactin concentrations during the late follicular phase (P < 0.01), indicating disruption of the estradiol-prolactin feedback mechanism. Among patients with premature ovarian insufficiency, serum prolactin concentrations increased concomitantly with rising serum of estradiol concentrations (after estradiol administration). Additionally, it was observed that the presence of prolactin significantly reduced estradiol production by granulosa cells (P < 0.05) (116).

 

An increment of prolactin levels is found even after hCG administration with a maximum prolactin level of 93.2 ng/mL; 1983 mIU/L (115).Notably, knowing that prolactin is a stress hormone, during assisted procedures it is increased, but this is a transitory increment without consequences in fertility outcome (247).

 

Not only gonadotrophins but also GnRH agonists are widely used during invitro fertilization to maintain a controlled and synchronized ovarian stimulation. Use of leuprolide acetate (GnRH agonist) concomitantly with hMG, resulted in higher prolactin and estradiol levels in comparison with patients receiving only hMG (prolactin 24.2 vs 16.8 ng/mL; 515 vs 358 mIU/L) (117). In another randomized study, along protocol with 0.1 mg subcutaneous triptorelin starting from day 10 of the preceding stimulation cycle and short protocol, where 0.1 mg subcutaneous triptorelin is given in the stimulating cycle, were compared. Prolactin levels were measured at 9 am in the first day of hCG administration. The long protocol correlated with higher prolactin levels (31.3 ± 16.9 vs 23.7 ± 11 ng/mL; 666 ± 359 vs 504 ± 234 mIU/L) (248).

 

In children, GnRH agonists are used in precocious puberty (CPP)as well as growth hormone deficiency (GHD)who do not properly respond to exogenous growth hormone treatment. In a study involving 119 children with CPP and 93 with GHD, treated with triptorelin or leuprolide, prolactin levels were measured before and every six months for 6 years for CPP group and for 2 years for GHD group. Moreover, prolactin levels were checked after 6 and 12 months of treatment withdrawal. In this study was concluded that even though prolactin levels were higher in triptorelin treated patients (only 3.8% developed hyperprolactinemia in triptorelin group which was solved after withdrawal – baseline 12.5 ± 3.7 ng/mL (266 ± 79 mIU/L) to max 45.6 ± 4.5 ng/mL; 970 ± 96 mIL/L), no significant difference was found in prolactin in basal condition and during GNRH agonist treatment in CPP and GHD (190)(Table 2).

 

OTHER DRUGS

 

A lot of other drugs have been reported to cause mild (less than 2-fold increment) increases in prolactin levels. A synthesized visualization of these mechanisms is shown in Figure 2.

 

The acute administration of buspirone, an anxiolytic medication, was investigated in a study involving 8 healthy volunteers. The findings revealed an increase in plasma prolactin levels across all participants compared to the baseline levels observed in 8 control subjects. During the study, blood samples were collected at 30-minute intervals over a duration of 2 hours. The zenith of prolactin levels was observed between minutes 90 and 120 for all individuals, with the maximum elevation reaching 37 ng/mL (787 mIU/L) (249). It is noteworthy that the augmentation of prolactin is believed to exhibit a dose-dependent relationship. Furthermore, it was observed that chronic usage of buspirone did not lead to significant alterations in prolactin levels, indicating a potential adaptation to the acute changes induced by the medication. The underlying mechanism responsible for this phenomenon is posited to involve both serotoninergic and dopaminergic implications (119).

 

Carbamazepine, a widely used anticonvulsant, was examined in a cohort comprising 4 patients with complex partial seizures undergoing chronic carbamazepine treatment (200 mg administered three times daily). Blood samples were collected at intervals of 2 hours. Additionally, a group of 5 patients with untreated epileptic seizures participated, wherein a thyrotrophin-releasing hormone (TRH) stimulation test was performed both prior to and 35-50 days post the administration of 200 mg carbamazepine three times daily. Blood samples were obtained 10, 30, and 60 minutes following intravenous injection of 200µg TRH. Furthermore, 4 normal volunteer subjects were included in the study. On the first day, a placebo was administered, followed by the administration of 400 mg carbamazepine at 8 AM on the second day. Blood samples were collected at baseline on both days and subsequently at hourly intervals until 4 PM. After a span of two weeks, a nocturnal study was conducted, spanning from 6 PM to 6 AM. The investigation revealed that there were no discernible alterations in spontaneous prolactin release or TRH-stimulated prolactin levels. However, a slight increase in sleep-entrained prolactin values was observed, while retaining the secretory circadian rhythm. Given that the release of prolactin during sleep is largely attributed to serotoninergic activity, it is plausible that the modest increment (less than 2-fold) may implicate serotoninergic modulation (working as a serotonin-releasing factor and reuptake inhibitor) facilitated by carbamazepine (120,121).

 

Sympathomimetic amines fenfluramine and sibutramine, formerly used for appetite suppression due to their stimulatory effect on the synaptic concentration of serotonin, have been shown to induce hyperprolactinemia as a result of increased serotoninergic activity and postsynaptic stimulation of 5HT2Areceptors. In a case report, after starting sibutramine, a 38-year-old female patient developed hyperprolactinemia (prolactin levels 46 and 89.6 ng/mL (978 and 1906 mUI/L) with amenorrhea and galactorrhea. Discontinuation of sibutramine, confirmed by a sella MRI, led to rapid normalization of prolactin levels within 15 days, and symptoms resolved during a 90-day follow-up (2,127).

 

Cholinomimetic drugs have been reported controversially in the literature regarding their ability to cause hyperprolactinemia. However, in collaborative studies from the National Institute of Mental Health and the University of California, San Diego, three separate experiments were conducted involving volunteers of different genders and ages. In the first experiment, nine volunteers received physostigmine salicylate at 33 µg/kg, while in the second experiment, eleven male volunteers were given 22 µg/kg of physostigmine salicylate. The third experiment involved six volunteers receiving 3 mg of arecoline hydrobromide. Placebo saline was administered in all experiments as well. It was shown that intravenous injection of physostigmine or arecoline can elevate prolactin correlating with raised β-endorphin levels in the blood. Prolactin elevation was less than 100 ng/mL (2127 mUI/L). Cholinergic activation in the hypothalamus, particularly focusing on β-endorphin, might help in explaining how peptides modify primary neurochemical effects on hormone regulation in the hypothalamus and pituitary (44).

 

Bucillamine, an analogue of D-penicillamine used as an antirheumatic drug in Japan, has been reported to induce hyperprolactinemia (109 ng/mL (2319 mUI/L)) after 30 months of treatment start, associated with gynecomastia and galactorrhea in one case report. The mechanism remains unclear (133).

 

‘Ecstasy’ (MDMA) was shown to increase prolactin secretion in rhesus monkeys by stimulating serotonin release and by direct-acting as a 5HT2A agonist (250); In nine studies, five of them observed an increase in prolactin levels due to the intervention. However, in the remaining studies, there was no significant change in prolactin levels, and these unresponsive results tended to occur when a lower dose of the intervention was used on average. This suggests a potential relationship between the dosage of the intervention and its effect on prolactin levels (135).

 

Smoking, particularly the consumption of high-nicotine cigarettes, has been associated with a significant acute elevation in prolactin levels, ranging from 50% to 78% above the baseline, within 6 minutes after smoking. These elevated levels persist for approximately 42 minutes and return to baseline within 120 minutes of initiating smoking (136). The underlying mechanism probably involves the stimulation of rapid prolactin release through the augmentation of endogenous opioids, which subsequently inhibits dopamine release (251). However, prolonged nicotine exposure leads to desensitization of dopamine receptors, and lowers dopamine turnover (48) probably contributing to hyperprolactinemia. It has been hypothesized that the increased incidence of osteopenia and osteoporosis could be at least partly related to this effect (252).

 

Recently, an association has been reported between HIV-1 protease inhibitors and the adverse effect of galactorrhea and hyperprolactinemia in four HIV-1 infected women treated with indinavir, nelfinavir, ritonavir, or saquinavir. The cause of this unexpected toxicity could be attributed to several possible mechanisms: 1) Protease inhibitors may enhance the stimulatory effects of prolactin due to their inhibition of the cytochrome P450 system, leading to longer half-life of prolactin; 2) opportunistic infections in AIDS patients may induce cytokine-driven prolactin production by pituitary or immune cells; 3) protease inhibitors might exert direct endocrine effects on the pituitary or hypothalamus (253,254). To explore mechanisms of hyperprolactinemia induced by protease inhibitors, experiments were conducted using rat pituitary cells and hypothalamic neuronal endings. The results showed that both ritonavir and saquinavir could directly stimulate prolactin secretion, while not affecting dopamine release. This suggests that these protease inhibitors might interact with specific mammalian proteins in the anterior pituitary involved in prolactin secretion, leading to the observed galactorrhea and hyperprolactinemic effect (137).

 

Regarding chemotherapy and immunosuppression, there are some controversial data on the effect of chemotherapy and immunosuppression on prolactin levels, as significant prolactin increases are not frequent and usually mild (2). Prolactin and growth hormone have been involved as part of a cytokine system in the recovery of the immune response after chemotherapy and bone marrow transplantation (255). In a study of 20 breast cancer patients undergoing high-dose chemotherapy and autologous stem-cell transplantation, plasma prolactin levels increased within and 30 days after transplant, yet still remaining within the normal range. The use of antiemetic drugs further raised prolactin levels. Patients in continuous complete remission after transplantation exhibited higher prolactin levels, while elevated prolactin did not impact disease-free survival, suggesting potential for further research into post-transplant immune response (256).

 

Radiotherapy for intracranial germ cell tumors was shown to induce hyperprolactinemia with a prevalence of 35.3% (138).

 

Other drug inducing hyperprolactinemia are described in Table 1.

 

DRUGS REPORTED TO DECREASE PROLACTIN LEVELS OR HAVE AN EQUIVOCAL EFFECT

 

Several medications, beyond the established treatments like cabergoline, bromocriptine, carbidopa and levodopa, have reported effects on reducing prolactin levels. For instance, pseudoephedrine, an α-adrenergic stimulant primarily affecting α1 receptors, shares structural similarities with amphetamine and moderately stimulating dopamine release in the brain by acting on D2 receptors in the pituitary, consequently lowering prolactin levels. Studies have indicated pseudoephedrine's potential to decrease milk production, at least partly attributed to its effect on prolactin levels through dopaminergic actions in the pituitary (132). Moreover, indirect evidence suggests that α-1 receptors stimulation leads to decreased prolactin levels (41).

 

Amphetamine was seen to produce a poor prolactin suppressant effect in either normal- or hyperprolactinemic subjects. The proposed mechanism of prolactin lowering potential is due to their ability to stimulate the release of dopamine (257). However, during the withdrawal period of cocaine use, hyperprolactinemia has been observed, probably due to a decrease in dopamine levels, leading to dysregulation in the dopamine system and increased prolactin. Moreover, during withdrawal, prolactin can be secreted as a stress hormone (129).

 

Guanafascine, an α2 adrenergic agonist, used to treat ADHD, has been shown to decrease prolactin levels. In a longitudinal study spanning three years involving 15 patients diagnosed with hyperprolactinemia, the noteworthy suppressive impact of guanfacine on prolactin levels suggests potential involvement of hypothalamic or extrahypothalamic adrenergic pathways in the intricate regulation of prolactin secretion (131). Even though α2 stimulation has been shown to increase prolactin levels in rats, this is not fully understood in humans making the explanation in this case confusing (42).

 

The impact of benzodiazepines (BDZ) on prolactin secretion is a subject of debate. Research findings have yielded conflicting results. Some studies conducted on both non-epileptic patients and healthy volunteers have not detected significant modifications in prolactin levels following BDZ treatment (258). A study on 30 adolescent patients with schizophrenia with gradually increasing doses of diazepam to a maximum of 100-400 mg/day, with 4 weeks of treatment, showed that only doses higher than 250 mg/day give a significant but mild increase in prolactin levels. Proposed mechanisms are inhibition of TIDA neurons by activation of the GABA system, or activation of the endorphin-ergic system leading to hyperprolactinemia (118). On the contrary, diazepam was found to suppress the secretion of prolactin in vitro through one of two mechanisms: it either strengthens the direct inhibitory action of GABA on prolactin release, or it hinders a benzodiazepine-sensitive Ca2+-calmodulin dependent protein kinase at micromolar concentrations leading to a reduction of prolactin secretion (259).

 

Moreover, phenytoin, an anticonvulsant impeding sodium channels in nerve cells, have generated conflicting data regarding their impact on prolactin levels. In animal studies, phenytoin showcased a rapid decline in both prolactin release and mRNA concentrations, functioning as a partial T3 agonist by binding to T3 nuclear receptors (260). However, clinical observations revealed elevated resting levels of prolactin in phenytoin-treated patients compared to untreated counterparts. Remarkably, responses to metoclopramide and bromocriptine remained unaltered, indicating a limited effect of phenytoin on the D2 receptors present on lactotrophs (261). Even the conclusions drawn from these findings remain contentious. Evidence suggests that phenytoin treatment may enhance the growth hormone response to levodopa, implying a phenytoin-induced dopaminergic activity at the hypothalamic-pituitary level (122). More specifically, it is postulated that phenytoin might enhance dopamine receptor sensitivity by inhibiting the Ca2+ calmodulin complex. This effect could contribute to reduced prolactin secretion (122). On the contrary, other studies have not demonstrated any notable alterations in prolactin levels due to phenytoin administration (262). The discordant outcomes surrounding phenytoin's impact on prolactin levels underscore the complexity of its effects and necessitate further investigation for conclusive insights into its mechanisms of action.

 

In another comprehensive study involving 126 subjects, both males and females, with generalized or partial epilepsy receiving phenobarbital as monotherapy or in combination with phenytoin or benzodiazepines, a distinct pattern emerged. Specifically, the administration of phenobarbital, either alone or in combination, resulted in elevated prolactin levels, but this elevation was found to be statistically significant only in the male participants. Notably, knowing that an epileptic attack itself can cause hyperprolactinemia, those data remain confusing. The proposed mechanism in this study is phenobarbital interaction with GABA receptors, leading to increased prolactin levels (263). However other studies do not show any change in prolactin levels (123).

 

Valproic acid, an anticonvulsant working as a central stimulant of GABAergic neurons, has demonstrated the ability to reduce prolactin basal levels as well as TRH-stimulated prolactin levels. This is indirect proof of the synergically acting of GABA neurons with dopaminergic tracts (124). However, in another study, no effect of valproic acid on prolactin levels was noticed during the night (264).

 

Lithium carbonate, a pharmaceutical agent employed as a mood stabilizer, has undergone investigation in different studies, as it decreases dopamine release and glutamate, and increases inhibitory GABA (265). One of them encompassed a longitudinal examination involving 9 patients diagnosed with bipolar disorder. The focus of this study was the assessment of plasma prolactin levels before and 12 hours after the evening administration of lithium. Evaluations were conducted on days 1, 6, 8, 13, 30, 60, and 90. Notably, this investigation yielded no discernible correlation between lithium concentration and prolactin levels, and no statistically significant alterations in prolactin levels were observed. The second part of this study adopted a cross-sectional design, involving 26 patients with an established history of long-term lithium treatment spanning durations of 3 months to 20 years. A comparative analysis revealed that prolactin levels, measured at 9 AM following a one-hour period of rest, did not demonstrate elevation in comparison to 16 controls. It is noteworthy that in both studies, lithium concentrations ranged from 0.4 to 1.4 mmol/L (normal range 0.5-1.2 mmol/L) (266). Additionally, the administration of lithium did not exert an impact on the plasma prolactin response to thyrotrophin-releasing hormone (TRH) stimulation compared to pre-treatment levels (267). The combined findings from these investigations provide compelling evidence that lithium does not contribute to hyperprolactinemia, thereby distinguishing it from medications with such an effect.

Cocaine has been shown to decrease prolactin levels beginning at 30-min following cocaine administration reaching statistical significance at the 90- and 120-minute time points (134).

 

Those medications are mentioned in Table 1. Their mechanism of altering prolactin levels is summarized in Figure 2.

 

HERBAL MEDICINES AFFECTING PROLACTIN LEVELS

 

In Table 4 is list of herbal medicines has been used traditionally to stimulate lactation (268). However, firm scientific evidence that they actually induce hyperprolactinemia is scarce.

Table 4. Lactogenic Herbs (268)

Family name

Species name

Common name

Amaryllidaceae

Allium sativum

Garlic

Annonaceae

Xylopia aethiopica

 

African Pepper or Ethiopian Pepper

Asclepiadaceae

Secamoneafzelii

 

-

Costaceae

 

Costusafer

 

African Ginger

Euphorbiaceae

 

Euphorbia hirta

 

Asthma Plant or Tawa-Tawa

Euphorbia thymifolia

 

Petty Spurge

Hymenocardiaacida

 

African Almond or Honeytree

Plagiostylesafricana

 

-

Ricinus communis

 

Castor Bean Plant

Leguminosae

 

Tamarindus indica

 

Tamarind

Acacia nicolita

 

-

Desmodiumadscendens

 

-

Malvaceae

 

Hibiscus sabdariffa

 

Roselle or Red Sorrel

Gossypium herbaceum

 

Cotton Plant

Moraceae

 

 Milicia excelsa

 

African Teak or Iroko

Ficus species

Ficus or Fig trees

Musaceae

 

Musa paradisiaca

 

Plantain

Ranunculaceae

 

Nigella sativa

 

Black Cumin or Black Seed

 

Actaea (Cimiciguga) racemose

Black Cohosh

Solanaceae

 

Solanum torvum

 

Turkey Berry or Devil's Fig

Verbanaceae

 

Lippia multiflora

 

Bush Tea or False Green Tea

Zingiberaceae

 

Aframomummelegueta

 

Grains of Paradise or Alligator Pepper

Fabaceae

Trifolium pratense

Red Clover

Trigonella foenum-graecum

 

Fenugreek

Apiaceae

Foeniculum vulgare

Fennel

Some herbs are known to decrease prolactin levels. For example, chaste tree (Vitex agnus-castus) decrease prolactin levels by activating to D2-receptors and suppressing prolactin release, as shown in in vitro experiments on lactotroph cell cultures and in in vivo animal experiments (269). Another herb, Mucuna pruriens, which is a natural source of l-dihydroxyphenylalanine (a dopamine precursor) is found to decrease prolactin levels in humans (270). Vitamin B6 (pyridoxine), by acting as a coenzyme in dopamine synthesis and aspartame, a sweetener metabolized in phenylalanine (dopamine precursor), have been shown to interfere with milk production by reducing prolactin levels (271). Ashgawanda (Withania somnifera) is found to decrease prolactin levels up to 12% (272). Moreover, oral zinc is found to decrease prolactin levels below the normal range in all 17 subjects with normal prolactin levels, in scenario of increased zinc levels in the blood (273).

 

While none of the mentioned herbs are currently established within clinical guidelines for specifically lactogenic or prolactin-reducing purposes, ongoing research and anecdotal evidence suggest potential roles for these botanicals as adjunctive therapies.

 

CONCLUSION

 

In summary, this review underscores the significant role of drug-induced hyperprolactinemia in causing higher prolactin levels and provides detailed insights into how pharmaceutical agents contribute to this effect. However, understanding the complex mechanisms behind drug-induced hyperprolactinemia is still a work in progress. More research is needed to delve deeper into these mechanisms and gain better insights. These efforts will contribute to refining treatment strategies and improving patient care.

 

REFERENCES

 

  1. Soto-Pedre E, Newey PJ, Bevan JS, Greig N, Leese GP. The epidemiology of hyperprolactinaemia over 20 years in the Tayside region of Scotland: the Prolactin Epidemiology, Audit and Research Study (PROLEARS). Clin Endocrinol (Oxf). 2017 Jan;86 (1):60–7.
  2. Torre DL, Falorni A. Pharmacological causes of hyperprolactinemia. Ther Clin Risk Manag. 2007 Oct;3 (5):929–51.
  3. Asa SL, Mete O, Perry A, Osamura RY. Overview of the 2022 WHO Classification of Pituitary Tumors. Endocr Pathol. 2022 Mar 1;33 (1):6–26.
  4. Samperi I, Lithgow K, Karavitaki N. Hyperprolactinaemia. J Clin Med. 2019 Dec 13;8 (12):2203.
  5. Vilar L, Vilar CF, Lyra R, Freitas M da C. Pitfalls in the Diagnostic Evaluation of Hyperprolactinemia. Neuroendocrinology. 2019;109 (1):7–19.
  6. Ajmal A, Joffe H, Nachtigall LB. Psychotropic-induced hyperprolactinemia: a clinical review. Psychosomatics. 2014;55 (1):29–36.
  7. Smith S, Wheeler MJ, Murray R, O’Keane V. The effects of antipsychotic-induced hyperprolactinaemia on the hypothalamic-pituitary-gonadal axis. J Clin Psychopharmacol. 2002 Apr;22 (2):109–14.
  8. Kinon BJ, Gilmore JA, Liu H, Halbreich UM. Prevalence of hyperprolactinemia in schizophrenic patients treated with conventional antipsychotic medications or risperidone11This work was sponsored by Eli Lilly and Company. Psychoneuroendocrinology. 2003 Apr 1;28:55–68.
  9. Halbreich U, Kahn LS. Hyperprolactinemia and schizophrenia: mechanisms and clinical aspects. J Psychiatr Pract. 2003 Sep;9 (5):344–53.
  10. Wieck A, Haddad P. Hyperprolactinaemia caused by antipsychotic drugs. BMJ. 2002 Feb 2;324 (7332):250–2.
  11. Petit A, Piednoir D, Germain ML, Trenque T. Hyperprolactinémies d’origine médicamenteuse : étude cas/non-cas dans la banque nationale de pharmacovigilance. Therapies. 2003 Mar 1;58 (2):159–63.
  12. Kavarthapu R, Dufau ML. Prolactin receptor gene transcriptional control, regulatory modalities relevant to breast cancer resistance and invasiveness. Frontiers in Endocrinology [Internet]. 2022 [cited 2023 Jul 14];13. Available from: https://www.frontiersin.org/articles/10.3389/fendo.2022.949396
  13. Cabrera-Reyes EA, Limón-Morales O, Rivero-Segura NA, Camacho-Arroyo I, Cerbón M. Prolactin function and putative expression in the brain. Endocrine. 2017 Aug 1;57 (2):199–213.
  14. Marano RJ, Ben-Jonathan N. Minireview: Extrapituitary Prolactin: An Update on the Distribution, Regulation, and Functions. Mol Endocrinol. 2014 May;28 (5):622–33.
  15. Dagvadorj A, Collins S, Jomain JB, Abdulghani J, Karras J, Zellweger T, et al. Autocrine prolactin promotes prostate cancer cell growth via Janus kinase-2-signal transducer and activator of transcription-5a/b signaling pathway. Endocrinology. 2007 Jul;148 (7):3089–101.
  16. Jacobson EM, Hugo ER, Borcherding DC, Ben-Jonathan N. Prolactin in breast and prostate cancer: molecular and genetic perspectives. Discov Med. 2011 Apr;11 (59):315–24.
  17. Bouilly J, Sonigo C, Auffret J, Gibori G, Binart N. Prolactin signaling mechanisms in ovary. Molecular and Cellular Endocrinology. 2012 Jun 5;356 (1):80–7.
  18. Abramicheva PA, Smirnova OV. Prolactin Receptor Isoforms as the Basis of Tissue-Specific Action of Prolactin in the Norm and Pathology. Biochemistry Moscow. 2019 Apr 1;84 (4):329–45.
  19. Ben-Jonathan N, LaPensee CR, LaPensee EW. What can we learn from rodents about prolactin in humans? Endocr Rev. 2008 Feb;29 (1):1–41.
  20. Ben-Jonathan N, MERSHONf JL, Allen DL, Steinmetz RW. Extrapituitary Prolactin: Distribution, Regulation, Functions, and Clinical Aspects. 1996;17 (6).
  21. Prabhakar VKB, Davis JRE. Hyperprolactinaemia. Best Practice & Research Clinical Obstetrics & Gynaecology. 2008 Apr 1;22 (2):341–53.
  22. Spiegel K, Follenius M, Simon C, Saini J, Ehrhart J, Brandenberger G. Prolactin secretion and sleep. Sleep. 1994 Feb;17 (1):20–7.
  23. Grattan DR. 60 YEARS OF NEUROENDOCRINOLOGY: The hypothalamo-prolactin axis. J Endocrinol. 2015 Aug;226 (2):T101–22.
  24. Freeman ME, Kanyicska B, Lerant A, Nagy G. Prolactin: structure, function, and regulation of secretion. Physiol Rev. 2000 Oct;80 (4):1523–631.
  25. Gonzalez-Iglesias AE, Murano T, Li S, Tomić M, Stojilkovic SS. Dopamine Inhibits Basal Prolactin Release in Pituitary Lactotrophs through Pertussis Toxin-Sensitive and -Insensitive Signaling Pathways. Endocrinology. 2008 Apr 1;149 (4):1470–9.
  26. Vallar L, Meldolesi J. Mechanisms of signal transduction at the dopamine D2 receptor. Trends in Pharmacological Sciences. 1989 Feb 1;10 (2):74–7.
  27. Day RN, Liu J, Sundmark V, Kawecki M, Berry D, Elsholtz HP. Selective Inhibition of Prolactin Gene Transcription by the ETS-2 Repressor Factor *. Journal of Biological Chemistry. 1998 Nov 27;273 (48):31909–15.
  28. Booth AK, Gutierrez-Hartmann A. Signaling Pathways Regulating Pituitary Lactotrope Homeostasis and Tumorigenesis. In: Diakonova P Maria, editor. Recent Advances in Prolactin Research [Internet]. Cham: Springer International Publishing; 2015 [cited 2023 Jul 16]. p. 37–59. (Advances in Experimental Medicine and Biology). Available from: https://doi.org/10.1007/978-3-319-12114-7_2
  29. Suzuki S, Yamamoto I, Arita J. Mitogen-activated protein kinase-dependent stimulation of proliferation of rat lactotrophs in culture by 3’,5’-cyclic adenosine monophosphate. Endocrinology. 1999 Jun;140 (6):2850–8.
  30. Senogles SE. D2 dopamine receptor-mediated antiproliferation in a small cell lung cancer cell line, NCI-H69. Anticancer Drugs. 2007 Aug;18 (7):801–7.
  31. Radl D, Mei CD, Chen E, Lee H, Borrelli E. Each individual isoform of the dopamine D2 receptor protects from lactotroph hyperplasia. Molecular Endocrinology. 2013;27 (6):953–65.
  32. Grandison L, Cavagnini F, Schmid R, Invitti SC, Guidotti A. gamma-Aminobutyric acid- and benzodiazepine-binding sites in human anterior pituitary tissue. J Clin Endocrinol Metab. 1982 Mar;54 (3):597–601.
  33. Ammari R, Broberger C. Pre- and post-synaptic modulation by GABAB receptors of rat neuroendocrine dopamine neurones. Journal of Neuroendocrinology. 2020;32 (11):e12881.
  34. Fitzgerald P, Dinan TG. Prolactin and dopamine: what is the connection? A review article. J Psychopharmacol. 2008 Mar;22 (2 Suppl):12–9.
  35. Peuskens J, Pani L, Detraux J, De Hert M. The effects of novel and newly approved antipsychotics on serum prolactin levels: a comprehensive review. CNS Drugs. 2014 May;28 (5):421–53.
  36. de-Castro-e-Silva E, Ramalho MJ, Midlej M, Cobas C, Machado A, Antunes-Rodrigues J. Desipramine blocks stress-induced prolactin release in rats: role of central beta-2 adrenoceptors. Braz J Med Biol Res. 1990;23 (2):199–204.
  37. Sonino N, Navarrini C, Ruini C, Fallo F, Boscaro M, Fava GA. Life events in the pathogenesis of hyperprolactinemia. Eur J Endocrinol. 2004 Jul;151 (1):61–5.
  38. Willoughby JO, Day TA, Menadue MF, Jervois PM, Blessing WW. Adrenoceptors in the preoptic-anterior hypothalamic area stimulate secretion of prolactin but not growth hormone in the male rat. Brain Res Bull. 1986 May;16 (5):697–704.
  39. Day TA, Jervois PM, Menadue MF, Willoughby JO. Catecholamine mechanisms in medio-basal hypothalamus influence prolactin but not growth hormone secretion. Brain Res. 1982 Dec 16;253 (1–2):213–9.
  40. Koshiyama H, Kato Y, Shimatsu A, Murakami Y, Hattori N, Ishikawa Y, et al. Possible involvement of endogenous opioid peptides in prolactin secretion induced by alpha 2-adrenergic stimulation in rats. Proc Soc Exp Biol Med. 1989 Nov;192 (2):105–8.
  41. Fuxe K, Hökfelt T. Participation of Central Monoamine Neurons in the Regulation of Anterior Pituitary Function with Special Regard to the Neuro-Endocrine Role of Tubero-Infundibular Dopamine Neurons. In: Bargmann W, Scharrer B, editors. Aspects of Neuroendocrinology. Berlin, Heidelberg: Springer; 1970. p. 192–205.
  42. Kapoor R, Chapman IM, Willoughby JO. α2 and β Adrenoceptors in the Mediobasal Hypothalamus and α2 Adrenoceptors in the Preoptic-Anterior Hypothalamus Stimulate Prolactin Secretion in the Conscious Male Rat. Journal of Neuroendocrinology. 1993;5 (2):189–93.
  43. Muraki T, Tokunaga Y, Nakadate T, Kato R. Inhibition by cholinergic agonists of the prolactin release induced by morphine. Naunyn Schmiedebergs Arch Pharmacol. 1979 Sep;308 (3):249–54.
  44. Risch SC, Janowsky DS, Siever LJ, Judd LJ, Rausch JL, Huey LY, et al. Correlated cholinomimetic-stimulated beta-endorphin and prolactin release in humans. Peptides. 1982;3 (3):319–22.
  45. Lanfranco F, Motta G, Baldi M, Gasco V, Grottoli S, Benso A, et al. Ghrelin and anterior pituitary function. Front Horm Res. 2010;38:206–11.
  46. Debeljuk L, Lasaga M. Tachykinins and the control of prolactin secretion. Peptides. 2006 Nov;27 (11):3007–19.
  47. Shaw D, al’Absi M. Blunted opiate modulation of prolactin response in smoking men and women. Pharmacol Biochem Behav. 2010 Mar;95 (1):1–5.
  48. Netter P, Toll C, Lujic C, Reuter M, Hennig J. Addictive and nonaddictive smoking as related to responsivity to neurotransmitter systems. Behav Pharmacol. 2002 Sep;13 (5–6):441–9.
  49. Knigge U, Warberg J. Neuroendocrine functions of histamine. Agents Actions Suppl. 1991;33:29–53.
  50. Coker F, Taylor D. Antidepressant-induced hyperprolactinaemia. CNS Drugs. 2010 Jul 1;24 (7):563–74.
  51. Rittenhouse PA, Levy AD, Li Q, Bethea CL, Van de Kar LD. Neurons in the hypothalamic paraventricular nucleus mediate the serotonergic stimulation of prolactin secretion via 5-HT1c/2 receptors. Endocrinology. 1993 Aug;133 (2):661–7.
  52. Emiliano ABF, Fudge JL. From galactorrhea to osteopenia: rethinking serotonin-prolactin interactions. Neuropsychopharmacology. 2004 May;29 (5):833–46.
  53. Samson WK, Bianchi R, Mogg RJ, Rivier J, Vale W, Melin P. Oxytocin mediates the hypothalamic action of vasoactive intestinal peptide to stimulate prolactin secretion. Endocrinology. 1989 Feb;124 (2):812–9.
  54. Al-Chalabi M, Bass AN, Alsalman I. Physiology, Prolactin. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Aug 6]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK507829/
  55. Schlechte J, Sherman B, Halmi N, vanGilder J, Chapler F, Dolan K, et al. Prolactin-Secreting Pituitary Tumors in Amenorrheic Women: A Comprehensive Study*. Endocrine Reviews. 1980 Jul 1;1 (3):295–308.
  56. Vance ML, Thorner MO. Prolactinomas. Endocrinol Metab Clin North Am. 1987 Sep;16 (3):731–53.
  57. Carter JN, Tyson JE, Tous G, Van Vliet S, Faiman C, Friesen HG. Prolactin-Secreting Tumors and Hypogonadism in 22 Men. N Engl J Med. 1978 Oct 19;299 (16):847–52.
  58. Biller BM, Baum HB, Rosenthal DI, Saxe VC, Charpie PM, Klibanski A. Progressive trabecular osteopenia in women with hyperprolactinemic amenorrhea. The Journal of Clinical Endocrinology & Metabolism. 1992 Sep 1;75 (3):692–7.
  59. Thapa S, Bhusal K. Hyperprolactinemia. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Jun 19]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK537331/
  60. Montejo ÁL, Arango C, Bernardo M, Carrasco JL, Crespo-Facorro B, Cruz JJ, et al. Spanish consensus on the risks and detection of antipsychotic drug-related hyperprolactinaemia. Rev Psiquiatr Salud Ment. 2016;9 (3):158–73.
  61. Molitch ME. Medication-Induced Hyperprolactinemia. Mayo Clinic Proceedings. 2005 Aug 1;80 (8):1050–7.
  62. Amato D, Kruyer A, Samaha AN, Heinz A. Hypofunctional Dopamine Uptake and Antipsychotic Treatment-Resistant Schizophrenia. Front Psychiatry. 2019 May 28;10:314.
  63. Thompson J, Thomas N, Singleton A, Piggott M, Lloyd S, Perry EK, et al. D2 dopamine receptor gene (DRD2) Taq1 A polymorphism: reduced dopamine D2 receptor binding in the human striatum associated with the A1 allele. Pharmacogenetics. 1997 Dec;7 (6):479–84.
  64. Osmanova DZ, Freidin MB, Fedorenko OY, Pozhidaev IV, Boiko AS, Vyalova NM, et al. A pharmacogenetic study of patients with schizophrenia from West Siberia gets insight into dopaminergic mechanisms of antipsychotic-induced hyperprolactinemia. BMC Med Genet. 2019 Apr 9;20 (Suppl 1):47.
  65. Molitch ME. Drugs and prolactin. Pituitary. 2008 Jun 1;11 (2):209–18.
  66. Gruen PG, Sachar EJ, Altman N, Langer G, Tabrizi MA, Halpern FS. Relation of plasma prolactin to clinical response in schizophrenic patients. Arch Gen Psychiatry. 1978 Oct;35 (10):1222–7.
  67. Goodnick PJ, Santana O, Rodriguez L. Antipsychotics: impact on prolactin levels. Expert Opinion on Pharmacotherapy. 2002 Oct 1;3 (10):1381–91.
  68. Schlösser R, Gründer G, Anghelescu I, Hillert A, Ewald-Gründer S, Hiemke C, et al. Long-term effects of the substituted benzamide derivative amisulpride on baseline and stimulated prolactin levels. Neuropsychobiology. 2002;46 (1):33–40.
  69. Turkington RW. Prolactin Secretion in Patients Treated With Various Drugs: Phenothiazines, Tricyclic Antidepressants, Reserpine, and Methyldopa. Archives of Internal Medicine. 1972 Sep 1;130 (3):349–54.
  70. Spitzer M, Sajjad R, Benjamin F. Pattern of development of hyperprolactinemia after initiation of haloperidol therapy. Obstet Gynecol. 1998 May;91 (5 Pt 1):693–5.
  71. Crowley TJ, Hydinger-Macdonald M. Motility, Parkinsonism, and prolactin with thiothixene and thioridazine. Arch Gen Psychiatry. 1981 Jun;38 (6):668–75.
  72. Vercellini P, Sacerdote P, Trespidi L, Manfredi B, Panerai AE, Crosignani PG. Veralipride for hot flushes induced by a gonadotropin-releasing hormone agonist: a controlled study. Fertil Steril. 1994 Nov;62 (5):938–42.
  73. Cosi C, Carilla-Durand E, Assié MB, Ormiere AM, Maraval M, Leduc N, et al. Partial agonist properties of the antipsychotics SSR181507, aripiprazole and bifeprunox at dopamine D2 receptors: G protein activation and prolactin release. Eur J Pharmacol. 2006 Mar 27;535 (1–3):135–44.
  74. Ivkovic J, Lindsten A, George V, Eriksson H, Hobart M. Effect of Brexpiprazole on Prolactin: An Analysis of Short- and Long-Term Studies in Schizophrenia. J Clin Psychopharmacol. 2019;39 (1):13–9.
  75. Stanniland C, Taylor D. Tolerability of atypical antipsychotics. Drug Saf. 2000 Mar;22 (3):195–214.
  76. Weiden PJ. Iloperidone for the treatment of schizophrenia: an updated clinical review. Clin Schizophr Relat Psychoses. 2012 Apr;6 (1):34–44.
  77. Kuchay MS, Mithal A. Levosulpiride and Serum Prolactin Levels. Indian J Endocrinol Metab. 2017;21 (2):355–8.
  78. Lozano R, Concha MP, Montealegre A, de Leon L, Villalba JO, Esteban HL, et al. Effectiveness and safety of levosulpiride in the treatment of dysmotility-like functional dyspepsia. Ther Clin Risk Manag. 2007 Mar;3 (1):149–55.
  79. Suthar N, Aneja J. Lurasidone-induced Parkinsonism and Hyperprolactinemia. Indian J Psychol Med. 2019;41 (2):192–4.
  80. Pacchiarotti I, Murru A, Kotzalidis GD, Bonnin CM, Mazzarini L, Colom F, et al. Hyperprolactinemia and medications for bipolar disorder: Systematic review of a neglected issue in clinical practice. European Neuropsychopharmacology. 2015 Aug 1;25 (8):1045–59.
  81. Einarson TR, Hemels MEH, Nuamah I, Gopal S, Coppola D, Hough D. An analysis of potentially prolactin-related adverse events and abnormal prolactin values in randomized clinical trials with paliperidone palmitate. Ann Pharmacother. 2012 Oct;46 (10):1322–30.
  82. Togo T, Iseki E, Shoji M, Oyama I, Kase A, Uchikado H, et al. Prolactin levels in schizophrenic patients receiving perospirone in comparison to risperidone. J Pharmacol Sci. 2003 Mar;91 (3):259–62.
  83. Brunelleschi S, Zeppegno P, Risso F, Cattaneo CI, Torre E. Risperidone-associated hyperprolactinemia: evaluation in twenty psychiatric outpatients. Pharmacol Res. 2003 Oct;48 (4):405–9.
  84. Weizman A, Maoz B, Treves I, Asher I, Ben-David M. Sulpiride-induced hyperprolactinemia and impotence in male psychiatric outpatients. Prog Neuropsychopharmacol Biol Psychiatry. 1985;9 (2):193–8.
  85. da Silva OP, Knoppert DC, Angelini MM, Forret PA. Effect of domperidone on milk production in mothers of premature newborns: a randomized, double-blind, placebo-controlled trial. CMAJ. 2001 Jan 9;164 (1):17–21.
  86. Koch MW, Liu WQ, Camara-Lemarroy C, Zhang Y, Pike GB, Metz L, et al. Domperidone-induced elevation of serum prolactin levels and immune response in multiple sclerosis. J Neuroimmunol. 2019 Sep 15;334:576974.
  87. Schettini G, Mastronardi P, Scanni E, Pinto M, Forgione A, Florio T, et al. [Antiemetic effect of the levo isomer of sulpiride (L-sulpiride) in humans]. Minerva Anestesiol. 1989 May;55 (5):239–43.
  88. Meltzer HY, Fang VS, Tricou BJ, Robertson A. Effect of antidepressants on neuroendocrine axis in humans. Adv Biochem Psychopharmacol. 1982;32:303–16.
  89. Anton RF, Sexauer JD, Randall CL. Amoxapine elevates serum prolactin in depressed men. Journal of Affective Disorders. 1983 Nov 1;5 (4):305–10.
  90. Cordes J, Kahl KG, Werner C, Henning U, Regenbrecht G, Larisch R, et al. Clomipramine-induced serum prolactin as a marker for serotonin and dopamine turnover: results of an open label study. Eur Arch Psychiatry Clin Neurosci. 2011 Dec;261 (8):567–73.
  91. Nutt D, Middleton H, Franklin M. The neuroendocrine effects of oral imipramine. Psychoneuroendocrinology. 1987;12 (5):367–75.
  92. Özkan HM. [Duloxetine Associated Galactorrhea and Hyperprolactinemia: A Case Report]. Turk Psikiyatri Derg. 2020;31 (4):294–6.
  93. Trenque T, Herlem E, Auriche P, Dramé M. Serotonin Reuptake Inhibitors and Hyperprolactinaemia. Drug-Safety. 2011 Dec 1;34 (12):1161–6.
  94. Yang MS, Cheng WJ, Huang MC. Dose-related hyperprolactinemia induced by venlafaxine. Prog Neuropsychopharmacol Biol Psychiatry. 2009 Jun 15;33 (4):733–4.
  95. Slater SL, Lipper S, Shiling DJ, Murphy DL. Elevation of plasma-prolactin by monoamine-oxidase inhibitors. Lancet. 1977 Aug 6;2 (8032):275–6.
  96. Segal M, Heys RF. Inappropriate lactation. Br Med J. 1969 Oct 25;4 (5677):236.
  97. Pradalier A, Vincent D, Barzegar C. [Hyperprolactinemia induced by indoramin]. Therapie. 1998;53 (5):500–2.
  98. Ae W, Pj C. Attenuation of the prolactin-stimulating and hyperthermic effects of nefazodone after subacute treatment. J Clin Psychopharmacol. 1994;14:268–73.
  99. Otani K, Yasui N, Kaneko S, Ishida M, Ohkubo T, Osanai T, et al. Trazodone treatment increases plasma prolactin concentrations in depressed patients. Int Clin Psychopharmacol. 1995 Jun;10 (2):115–7.
  100. Ozkan HM. Galactorrhea and hyperprolactinemia during vortioxetine use: case report. Turkish Journal of Biochemistry. 2019 Feb 1;44 (1):105–7.
  101. Schüle C, Baghai T, Schmidbauer S, Bidlingmaier M, Strasburger CJ, Laakmann G. Reboxetine acutely stimulates cortisol, ACTH, growth hormone and prolactin secretion in healthy male subjects. Psychoneuroendocrinology. 2004 Feb;29 (2):185–200.
  102. Burland WL, Gleadle RI, Lee RM, Rowley-Jones D, Groom GV. Prolactin responses to cimetidine. Br J Clin Pharmacol. 1979 Jan;7 (1):19–21.
  103. Delitala G, Devilla L, Pende A, Canessa A. Effects of the H2 receptor antagonist ranitidine on anterior pituitary hormone secretion in man. Eur J Clin Pharmacol. 1982;22 (3):207–11.
  104. Duwicquet F, Gras-Champel V, Masmoudi K. Hyperprolactinémie avec galactorrhée induites par le lansoprazole : à propos d’un cas. Therapies. 2017 Dec 1;72 (6):691–3.
  105. MacGilchrist AJ, Howden CW, Kenyon CJ, Beastall GH, Reid JL. The effects of omeprazole on endocrine function in man. Eur J Clin Pharmacol. 1987;32 (4):423–5.
  106. Dammann HG, Bethke T, Burkhardt F, Wolf N, Khalil H, Luehmann R. Effects of pantoprazole on endocrine function in healthy male volunteers. Aliment Pharmacol Ther. 1994 Oct;8 (5):549–54.
  107. Dammann HG, Burkhardt F, Wolf N. The effects of oral rabeprazole on endocrine and gastric secretory function in healthy volunteers. Aliment Pharmacol Ther. 1999 Sep;13 (9):1195–203.
  108. Müller EE, Locatelli V, Cella S, Peñalva A, Novelli A, Cocchi D. Prolactin-Lowering and -Releasing Drugs Mechanisms of Action and Therapeutic Applications: Drugs. 1983 Apr;25 (4):399–432.
  109. Chan V, Wang C, Yeung RT. Effects of heroin addiction on thyrotrophin, thyroid hormones and porlactin secretion in men. Clin Endocrinol (Oxf). 1979 Jun;10 (6):557–65.
  110. Bart G, Borg L, Schluger JH, Green M, Ho A, Kreek MJ. Suppressed prolactin response to dynorphin A1-13 in methadone-maintained versus control subjects. J Pharmacol Exp Ther. 2003 Aug;306 (2):581–7.
  111. Delitala G, Grossman A, Besser GM. The participation of hypothalamic dopamine in morphine-induced prolactin release in man. Clin Endocrinol (Oxf). 1983 Oct;19 (4):437–44.
  112. Camanni E, Strumia E, Portaleone P, Molinatti GM. Prolactin secretion during reserpine and syrosingopine treatment. Eur J Clin Pharmacol. 1981;20 (5):347–9.
  113. Romeo JH, Dombrowski R, Kwak YS, Fuehrer S, Aron DC. Hyperprolactinaemia and verapamil: prevalence and potential association with hypogonadism in men. Clinical Endocrinology. 1996;45 (5):571–5.
  114. Goh HH, Ratnam SS. Effect of estrogens on prolactin secretion in transsexual subjects. Arch Sex Behav. 1990 Oct;19 (5):507–16.
  115. Kamel A, Halim AA, Shehata M, AlFarra S, El-Faissal Y, Ramadan W, et al. Changes in serum prolactin level during intracytoplasmic sperm injection, and effect on clinical pregnancy rate: a prospective observational study. BMC Pregnancy Childbirth. 2018 May 9;18 (1):141.
  116. Takiguchi S, Nakamura Y, Yamagata Y, Takayama H, Harada A, Sugino N, et al. Role of transient hyperprolactinemia in the late follicular phase of the gonadotropin‐stimulated cycle. Reprod Med Biol. 2002 Dec 11;1 (2):69–74.
  117. Meldrum DR, Cedars MI, Hamilton F, Huynh D, Wisot A, Marr B. Leuprolide acetate elevates prolactin during ovarian stimulation with gonadotropins. J Assist Reprod Genet. 1992 Jun 1;9 (3):251–3.
  118. Weizman A, Tyano S, Wijsenbeek H, Ben David M. High dose diazepam treatment and its effect on prolactin secretion in adolescent schizophrenic patients. Psychopharmacology (Berl). 1984;82 (4):382–5.
  119. Cowen PJ, Anderson IM, Grahame-Smith DG. Neuroendocrine effects of azapirones. J Clin Psychopharmacol. 1990 Jun;10 (3 Suppl):21S-25S.
  120. Bonuccelli U, Murialdo G, Martino E, Lecchini S, Bonura ML, Bambini G, et al. Effects of carbamazepine on prolactin secretion in normal subjects and in epileptic subjects. Clin Neuropharmacol. 1985;8 (2):165–74.
  121. Dailey JW, Reith ME, Steidley KR, Milbrandt JC, Jobe PC. Carbamazepine-induced release of serotonin from rat hippocampus in vitro. Epilepsia. 1998 Oct;39 (10):1054–63.
  122. D’Alessandro R, Cortelli P, Gallassi R, Ghisoli E, Montanaro N, Zoni E, et al. Phenytoin-induced increase in growth hormone response to levodopa in adult males. J Neurol Neurosurg Psychiatry. 1984 Jul;47 (7):715–9.
  123. Murialdo G, Galimberti CA, Gianelli MV, Rollero A, Polleri A, Copello F, et al. Effects of valproate, phenobarbital, and carbamazepine on sex steroid setup in women with epilepsy. Clin Neuropharmacol. 1998;21 (1):52–8.
  124. Giroud M, Marchal G, Baleydier-Giroud F, Desgres J, Dumas R. [GABAergic control of hypophyseal gonadotropic function and prolactin. Demonstration by valproic acid]. J Gynecol Obstet Biol Reprod (Paris). 1985;14 (2):163–9.
  125. Li W, Liu R, Liu W, Li G, Chen C. The effect of topiramate versus flunarizine on the non-headache symptoms of migraine. Int J Neurosci. 2023 Jan;133 (1):19–25.
  126. Scarduelli C, Mattei AM, Brambilla G, Zavaglia C, Adelasco P, Cavioni V, et al. Effect of Fenfluramine Oral Administration on Serum Prolactin Levels in Healthy and Hyperprolactinemic Women. Gynecol Obstet Invest. 1985 Feb 1;19 (2):92–6.
  127. Soares Leaes CG, Pereira-Lima JFS, da Costa Oliveira M. A case of sibutramine-induced hyperprolactinemia. Neuro Endocrinol Lett. 2011;32 (5):616–8.
  128. Ziambaras K, Dagogo-Jack S. Tumor-grade hyperprolactinemia induced by multiple medications in the setting of renal failure. Endocr Pract. 1999;5 (3):139–42.
  129. Lago JA, Kosten TR. Stimulant withdrawal. Addiction. 1994 Nov;89 (11):1477–81.
  130. Janowsky DS, Leichner P, Parker D, Judd L, Huey L, Clopton P. Methylphenidate and serum prolactin in man. Psychopharmacology. 1978 Jan 1;58 (1):43–7.
  131. Hauger-Klevene JH, Pinkas MB, Gerber S. Blood pressure and prolactin: effects of guanfacine. Three-year follow-up study. Hypertension. 1981;3 (6 Pt 2):II-222–5.
  132. Aljazaf K, Hale TW, Ilett KF, Hartmann PE, Mitoulas LR, Kristensen JH, et al. Pseudoephedrine: effects on milk production in women and estimation of infant exposure via breastmilk. Br J Clin Pharmacol. 2003 Jul;56 (1):18–24.
  133. Mori T, Yokogawa N, Higuchi R, Tsujino M, Shimada K, Sugii S. Bucillamine-induced gigantomastia with galactorrhea and hyperprolactinaemia. Mod Rheumatol Case Rep. 2020 Jan;4 (1):122–5.
  134. Elman I, Lukas SE. Effects of cortisol and cocaine on plasma prolactin and growth hormone levels in cocaine-dependent volunteers. Addictive Behaviors. 2005 May 1;30 (4):859–64.
  135. Dumont GJH, Verkes RJ. A review of acute effects of 3,4-methylenedioxymethamphetamine in healthy volunteers. J Psychopharmacol. 2006 Mar;20 (2):176–87.
  136. Mendelson JH, Sholar MB, Mutschler NH, Jaszyna-Gasior M, Goletiani NV, Siegel AJ, et al. Effects of Intravenous Cocaine and Cigarette Smoking on Luteinizing Hormone, Testosterone, and Prolactin in Men. J Pharmacol Exp Ther. 2003 Oct 1;307 (1):339–48.
  137. Orlando G, Brunetti L, Vacca M. Ritonavir and Saquinavir directly stimulate anterior pituitary prolactin secretion, in vitro. Int J Immunopathol Pharmacol. 2002;15 (1):65–8.
  138. Xiang B, Zhu X, He M, Wu W, Pang H, Zhang Z, et al. Pituitary Dysfunction in Patients with Intracranial Germ Cell Tumors Treated with Radiotherapy. Endocrine Practice. 2020 Dec 1;26 (12):1458–68.
  139. McCutcheon RA, Harrison PJ, Howes OD, McGuire PK, Taylor DM, Pillinger T. Data-Driven Taxonomy for Antipsychotic Medication: A New Classification System. Biol Psychiatry. 2023 Apr 14;S0006-3223 (23)01200-3.
  140. Chokhawala K, Stevens L. Antipsychotic Medications. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Jun 7]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK519503/
  141. Kelly RJ, Robey RW, Chen CC, Draper D, Luchenko V, Barnett D, et al. A pharmacodynamic study of the P-glycoprotein antagonist CBT-1® in combination with paclitaxel in solid tumors. Oncologist. 2012;17 (4):512.
  142. Schinkel AH. P-Glycoprotein, a gatekeeper in the blood-brain barrier. Adv Drug Deliv Rev. 1999 Apr 5;36 (2–3):179–94.
  143. Boulton DW, DeVane CL, Liston HL, Markowitz JS. In vitro P-glycoprotein affinity for atypical and conventional antipsychotics. Life Sci. 2002 May 31;71 (2):163–9.
  144. El-Mallakh RS, Watkins J. Prolactin Elevations and Permeability Glycoprotein. Prim Care Companion CNS Disord. 2019 May 16;21 (3):27149.
  145. Burris KD, Molski TF, Xu C, Ryan E, Tottori K, Kikuchi T, et al. Aripiprazole, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther. 2002 Jul;302 (1):381–9.
  146. Jiang XJ, Wu FX, Zhang JP, Shi L, Hu JQ, Zhu HZ, et al. Effects of Risperidone and Aripiprazole on Serum Levels of Prolactin, Testosterone and Estradiol in Female Patients with Schizophrenia. Drug Res (Stuttg). 2018 Jul;68 (7):410–4.
  147. Lee BJ, Lee SJ, Kim MK, Lee JG, Park SW, Kim GM, et al. Effect of Aripiprazole on Cognitive Function and Hyperprolactinemia in Patients with Schizophrenia Treated with Risperidone. Clin Psychopharmacol Neurosci. 2013 Aug 28;11 (2):60–6.
  148. Spoelstra SK, Bruggeman R, Knegtering H. [An antipsychotic without dopamine receptor blockade?]. Tijdschr Psychiatr. 2021;63 (11):804–9.
  149. Wudarsky M, Nicolson R, Hamburger SD, Spechler L, Gochman P, Bedwell J, et al. Elevated Prolactin in Pediatric Patients on Typical and Atypical Antipsychotics. Journal of Child and Adolescent Psychopharmacology. 1999 Jan;9 (4):239–45.
  150. Aubert ML, Sizonenko PC, Paunier L. Human Prolactin during puberty and first menstrual cycles in girls. Pediatr Res. 1978 Feb;12 (2):157–157.
  151. Uttley L, Kearns B, Ren S, Stevenson M. Aripiprazole for the Treatment and Prevention of Acute Manic and Mixed Episodes in Bipolar I Disorder in Children and Adolescents: A NICE Single Technology Appraisal. PharmacoEconomics. 2013 Nov 1;31 (11):981–90.
  152. Koch MT, Carlson HE, Kazimi MM, Correll CU. Antipsychotic-Related Prolactin Levels and Sexual Dysfunction in Mentally Ill Youth: A 3-Month Cohort Study. Journal of the American Academy of Child & Adolescent Psychiatry. 2023 Sep 1;62 (9):1021–50.
  153. Krøigaard SM, Clemmensen L, Tarp S, Pagsberg AK. A Meta-Analysis of Antipsychotic-Induced Hypo- and Hyperprolactinemia in Children and Adolescents. J Child Adolesc Psychopharmacol. 2022 Sep;32 (7):374–89.
  154. Bunker MT, Marken PA, Schneiderhan ME, Ruehter VL. Attenuation of antipsychotic-induced hyperprolactinemia with clozapine. J Child Adolesc Psychopharmacol. 1997;7 (1):65–9.
  155. Rosenbloom AL. Hyperprolactinemia with Antipsychotic Drugs in Children and Adolescents. Int J Pediatr Endocrinol. 2010;2010:159402.
  156. Apter A, Dickerman Z, Gonen N, Assa S, Prager-Lewin R, Kaufman H, et al. Effect of chlorpromazine on hypothalamic-pituitary-gonadal function in 10 adolescent schizophrenic boys. Am J Psychiatry. 1983 Dec 1;140 (12):1588–91.
  157. Hellings JA, Arnold LE, Han JC. Dopamine antagonists for treatment resistance in autism spectrum disorders: review and focus on BDNF stimulators loxapine and amitriptyline. Expert Opin Pharmacother. 2017 Apr;18 (6):581–8.
  158. Simeon J, Lawrence S, Simeon S. Effects of pimozide on prolactin in children [proceedings]. Psychopharmacol Bull. 1979 Apr 1;15 (2):40–2.
  159. Sallee FR, Dougherty D, Sethuraman G, Vrindavanam N. Prolactin monitoring of haloperidol and pimozide treatment in children with Tourette’s syndrome. Biol Psychiatry. 1996 Nov 15;40 (10):1044–50.
  160. Savitz A, Lane R, Nuamah I, Singh J, Hough D, Gopal S. Long-Term Safety of Paliperidone Extended Release in Adolescents with Schizophrenia: An Open-Label, Flexible Dose Study. J Child Adolesc Psychopharmacol. 2015 Sep;25 (7):548–57.
  161. Gopal S, Lane R, Nuamah I, Copenhaver M, Singh J, Hough D, et al. Evaluation of Potentially Prolactin-Related Adverse Events and Sexual Maturation in Adolescents with Schizophrenia Treated with Paliperidone Extended-Release (ER) for 2 Years: A Post Hoc Analysis of an Open-Label Multicenter Study. CNS Drugs. 2017 Sep 1;31 (9):797–808.
  162. Stevens JR, Kymissis PI, Baker AJL. Elevated prolactin levels in male youths treated with risperidone and quetiapine. J Child Adolesc Psychopharmacol. 2005 Dec;15 (6):893–900.
  163. Anderson GM, Scahill L, McCracken JT, McDougle CJ, Aman MG, Tierney E, et al. Effects of short- and long-term risperidone treatment on prolactin levels in children with autism. Biol Psychiatry. 2007 Feb 15;61 (4):545–50.
  164. Troost PW, Lahuis BE, Hermans MH, Buitelaar JK, van Engeland H, Scahill L, et al. Prolactin release in children treated with risperidone - Impact and role of CYP2D6 metabolism. Journal of Clinical Psychopharmacology. 2007 Feb;27 (1):52–7.
  165. Stepanova E, Grant B, Findling RL. Asenapine Treatment in Pediatric Patients with Bipolar I Disorder or Schizophrenia: A Review. Paediatr Drugs. 2018 Apr;20 (2):121–34.
  166. Stocks JD, Taneja BK, Baroldi P, Findling RL. A phase 2a randomized, parallel group, dose-ranging study of molindone in children with attention-deficit/hyperactivity disorder and persistent, serious conduct problems. J Child Adolesc Psychopharmacol. 2012 Apr;22 (2):102–11.
  167. Gjessing Jensen K. Severe Hyperprolactinemia during Lurasidone Treatment in a 16-year Old Girl with Schizophrenia - A Case Report. Scand J Child Adolesc Psychiatr Psychol. 2022 Jan;10 (1):87–8.
  168. FDA. Review and Evaluation of Clinical Data NDA #200603 [Internet]. 2019. Available from: https://www.fda.gov/media/133816/download
  169. Malone RP, Delaney MA, Hyman SB, Cater JR. Ziprasidone in adolescents with autism: an open-label pilot study. J Child Adolesc Psychopharmacol. 2007 Dec;17 (6):779–90.
  170. Yoo HK, Joung YS, Lee JS, Song DH, Lee YS, Kim JW, et al. A multicenter, randomized, double-blind, placebo-controlled study of aripiprazole in children and adolescents with Tourette’s disorder. J Clin Psychiatry. 2013 Aug;74 (8):e772-780.
  171. Varol Tas F, Guvenir T. Amisulpride treatment of adolescent patients with schizophrenia or schizo-affective disorders. Eur Child Adolesc Psychiatry. 2009 Aug;18 (8):511–3.
  172. FDA. Brexpiprazole in Pediatric Patients with Schizophrenia Aged 13 to 17 [Internet]. 2021. Available from: https://www.fda.gov/media/155985/download?attachment
  173. Hanna GL, McCracken JT, Cantwell DP. Prolactin in childhood obsessive-compulsive disorder: clinical correlates and response to clomipramine. J Am Acad Child Adolesc Psychiatry. 1991 Mar;30 (2):173–8.
  174. Overtoom CCE, Verbaten MN, Kemner C, Kenemans JL, van Engeland H, Buitelaar JK, et al. Effects of methylphenidate, desipramine, and L-dopa on attention and inhibition in children with Attention Deficit Hyperactivity Disorder. Behavioural Brain Research. 2003;145 (1–2):7–15.
  175. Conners CK, Casat CD, Gualtieri CT, Weller E. Bupropion hydrochloride in attention deficit disorder with hyperactivity. Journal of the American Academy of Child & Adolescent Psychiatry. 1996;35 (10):1314–21.
  176. Türkoğlu S. Citalopram-Induced Galactorrhea in an Adolescent. Clin Neuropharmacol. 2016;39 (6):331.
  177. McGrane IR, Morefield CM, Aytes KL. Probable Galactorrhea Associated with Sequential Trials of Escitalopram and Duloxetine in an Adolescent Female. J Child Adolesc Psychopharmacol. 2019 Dec;29 (10):788–9.
  178. Iancu I, Ratzoni G, Weitzman A, Apter A. More fluoxetine experience. J Am Acad Child Adolesc Psychiatry. 1992 Jul 1;31 (4):755–6.
  179. Ekinci N, Güneş S, Kalinli M, Ekinci Ö. Sertraline-Related Amenorrhea in an Adolescent. Clin Neuropharmacol. 2019;42 (3):99–100.
  180. Alacqua M, Trifirò G, Arcoraci V, Germanò E, Magazù A, Calarese T, et al. Use and tolerability of newer antipsychotics and antidepressants: a chart review in a paediatric setting. Pharm World Sci. 20070621st ed. 2008 Jan;30 (1):44–50.
  181. FDA. FDA Adverse Event Reporting System (FAERS) Public Dashboard. 2023.
  182. Ijaiya K. Prolactin response to exercise, metoclopramide and other provacative agents in children. Eur J Pediatr. 1980 Sep;134 (3):231–7.
  183. Preeyasombat C, Mahachoklertwattana P, Sriphrapradang A, Choubtam L. Prolactin (PRL) release in normal and growth hormone deficient children after oral metoclopramide. J Med Assoc Thai. 1993 Oct;76 Suppl 2:34–41.
  184. Nicolson R, Craven-Thuss B, Smith J, McKinlay BD, Castellanos FX. A Randomized, Double-Blind, Placebo-Controlled Trial of Metoclopramide for the Treatment of Tourette’s Disorder. Journal of the American Academy of Child and Adolescent Psychiatry. 2005 Jul 1;44 (7):640.
  185. Deprettere AR, Van Acker KJ, Du Caju MVL. Increased serum prolactin but normal TSH during prolonged domperidone treatment in children. Eur J Pediatr. 1987 Mar 1;146 (2):189–91.
  186. Cho E, Ho S, Gerber P, Davidson AGF. Monitoring of serum prolactin in pediatric patients with cystic fibrosis who are receiving domperidone. Can J Hosp Pharm. 2009 Mar;62 (2):119–26.
  187. Jabbar A, Khan R, Farrukh SN. Hyperprolactinaemia induced by proton pump inhibitor. J Pak Med Assoc. 2010 Aug;60 (8):689–90.
  188. Stoff DM, Pasatiempo AP, Yeung J, Cooper TB, Bridger WH, Rabinovich H. Neuroendocrine responses to challenge with dl-fenfluramine and aggression in disruptive behavior disorders of children and adolescents. Psychiatry Research. 1992 Sep 1;43 (3):263–76.
  189. Tack LJW, Heyse R, Craen M, Dhondt K, Bossche HV, Laridaen J, et al. Consecutive Cyproterone Acetate and Estradiol Treatment in Late-Pubertal Transgender Female Adolescents. J Sex Med. 2017 May;14 (5):747–57.
  190. Massart F, Parrino R, Placidi G, Massai G, Federico G, Saggese G. Prolactin secretion before, during, and after chronic gonadotropin-releasing hormone agonist treatments in children. Fertil Steril. 2005 Sep;84 (3):719–24.
  191. Mendhekar DN. Clonidine-induced gynecomastia and hyperprolactinemia in a 6-year-old child. J Clin Psychiatry. 2005 Dec;66 (12):1616–7.
  192. Koch MT, Carlson HE, Kazimi MM, Correll CU. Antipsychotic-Related Prolactin Levels and Sexual Dysfunction in Mentally Ill Youth: A 3-Month Cohort Study. Journal of the American Academy of Child & Adolescent Psychiatry. 2023 Sep 1;62 (9):1021–50.
  193. Chanson P. Treatments of psychiatric disorders, hyperprolactinemia and dopamine agonists. Best Practice & Research Clinical Endocrinology & Metabolism. 2022 Dec 1;36 (6):101711.
  194. Melmed S, Casanueva FF, Hoffman AR, Kleinberg DL, Montori VM, Schlechte JA, et al. Diagnosis and treatment of hyperprolactinemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab. 2011 Feb;96 (2):273–88.
  195. Peveler RC, Branford D, Citrome L, Fitzgerald P, Harvey PW, Holt RIG, et al. Antipsychotics and hyperprolactinaemia: clinical recommendations. J Psychopharmacol. 2008 Mar;22 (2 Suppl):98–103.
  196. Byerly M, Suppes T, Tran QV, Baker RA. Clinical implications of antipsychotic-induced hyperprolactinemia in patients with schizophrenia spectrum or bipolar spectrum disorders: recent developments and current perspectives. J Clin Psychopharmacol. 2007 Dec;27 (6):639–61.
  197. Isola S, Hussain A, Dua A, Singh K, Adams N. Metoclopramide. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Jun 20]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK519517/
  198. Lertxundi U, Domingo-Echaburu S, Soraluce A, García M, Ruiz-Osante B, Aguirre C. Domperidone in Parkinson’s disease: a perilous arrhythmogenic or the gold standard? Curr Drug Saf. 2013 Feb;8 (1):63–8.
  199. Asztalos EV, Campbell-Yeo M, da Silva OP, Ito S, Kiss A, Knoppert D, et al. Enhancing Human Milk Production With Domperidone in Mothers of Preterm Infants. J Hum Lact. 2017 Feb;33 (1):181–7.
  200. Glover K, Casey JJ, Gilbert M. Case Report: Induced Lactation in an Adoptive Parent. afp. 2023 Feb;107 (2):119–20.
  201. Weimer AK. Lactation Induction in a Transgender Woman: Macronutrient Analysis and Patient Perspectives. J Hum Lact. 2023 Aug;39 (3):488–94.
  202. Reisman T, Goldstein Z. Case Report: Induced Lactation in a Transgender Woman. Transgend Health. 2018;3 (1):24–6.
  203. Sheffler ZM, Patel P, Abdijadid S. Antidepressants. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Jun 9]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK538182/
  204. Meltzer HY, Piyakalmala S, Schyve P, Fang VS. Lack of effect of tricyclic antidepressants on serum prolactin levels. Psychopharmacology. 1977 Jan 1;51 (2):185–7.
  205. Charney DS, Heninger GR, Sternberg DE. Serotonin Function and Mechanism of Action of Antidepressant Treatment: Effects of Amitriptyline and Desipramine. Archives of General Psychiatry. 1984 Apr 1;41 (4):359–65.
  206. Anderson IM, Cowen PJ. Clomipramine enhances prolactin and growth hormone responses to L-tryptophan. Psychopharmacology (Berl). 1986;89 (1):131–3.
  207. Cooper DS, Gelenberg AJ, Wojcik JC, Saxe VC, Ridgway EC, Maloof F. The Effect of Amoxapine and Imipramine on Serum Prolactin Levels. AMA Archives of Internal Medicine. 1981 Jul;141 (8):1023–5.
  208. Petit A, Piednoir D, Germain ML, Trenque T. Hyperprolactinémies d’origine médicamenteuse : étude cas/non-cas dans la banque nationale de pharmacovigilance. Therapies. 2003 Mar 1;58 (2):159–63.
  209. Papakostas GI, Miller KK, Petersen T, Sklarsky KG, Hilliker SE, Fava M. Serum Prolactin Levels Among Outpatients With Major Depressive Disorder During the Acute Phase of Treatment With Fluoxetine. J Clin Psychiatry. 2006 Jun 15;67 (6):19359.
  210. Reeves KW, Okereke OI, Qian J, Tworoger SS, Rice MS, Hankinson SE. Antidepressant use and circulating prolactin levels. Cancer Causes Control. 2016 Jul;27 (7):853–61.
  211. Jilani TN, Gibbons JR, Faizy RM, Saadabadi A. Mirtazapine. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Aug 12]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK519059/
  212. Laakmann G, Schüle C, Baghai T, Waldvogel E. Effects of mirtazapine on growth hormone, prolactin, and cortisol secretion in healthy male subjects. Psychoneuroendocrinology. 1999 Oct;24 (7):769–84.
  213. Shin JJ, Saadabadi A. Trazodone. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 [cited 2023 Aug 12]. Available from: http://www.ncbi.nlm.nih.gov/books/NBK470560/
  214. Schüle C, Baghai T, Laakmann G. Mirtazapine Decreases Stimulatory Effects of Reboxetine on Cortisol, Adrenocorticotropin and Prolactin Secretion in Healthy Male Subjects. Neuroendocrinology. 2004;79 (1):54–62.
  215. Moeller O, Hetzel G, Rothermundt M, Michael N, Nyhuis PW, Suslow T, et al. Oral citalopram and reboxetine challenge tests before and after selective antidepressant treatment. J Psychiatr Res. 2003;37 (3):261–2.
  216. Knigge UP. Histaminergic regulation of prolactin secretion. Dan Med Bull. 1990 Apr;37 (2):109–24.
  217. Perret G, Hugues JN, Louchahi M, Varoquaux O, Modigliani E. Effect of a short-term oral administration of cimetidine and ranitidine on the basal and thyrotropin-releasing hormone-stimulated serum concentrations of prolactin, thyrotropin and thyroid hormones in healthy volunteers. A double-blind cross-over study. Pharmacology. 1986;32 (2):101–8.
  218. Rampello L, Raffaele R, Nicoletti G, Pira FL, Vecchio I, Malaguarnera M, et al. Neurobehavioral Syndrome Induced by H2-Receptor Blocker Withdrawal: Possible Role of Prolactin. Clinical Neuropharmacology. 1997 Feb;20 (1):49–54.
  219. Sibilia V, Netti C, Guidobono F, Pagani F, Pecile A. Cimetidine-induced prolactin release: possible involvement of the GABA-ergic system. Neuroendocrinology. 1985 Mar;40 (3):189–92.
  220. Ashfaq M, Haroon MZ, Alkahraman YM. Proton pump inhibitors therapy and risk of hyperprolactinemia with associated sexual disorders. Endocrine Regulations. 2022 Apr 1;56 (2):134–47.
  221. Helgadóttir H, Lund SH, Gizurarson S, Metz DC, Björnsson ES. Predictors of gastrin elevation following proton pump inhibitor therapy. J Clin Gastroenterol. 2020 Mar;54 (3):227–34.
  222. Pipaliya N, Solanke D, Rathi C, Patel R, Ingle M, Sawant P. Esomeprazole induced galactorrhea: a novel side effect. Clin J Gastroenterol. 2016 Feb 1;9 (1):13–6.
  223. Bennett M. Vitamin B12 deficiency, infertility and recurrent fetal loss. J Reprod Med. 2001 Mar 1;46 (3):209–12.
  224. Toprak O, Sarı Y, Koç A, Sarı E, Kırık A. The impact of hypomagnesemia on erectile dysfunction in elderly, non-diabetic, stage 3 and 4 chronic kidney disease patients: a prospective cross-sectional study. Clin Interv Aging. 2017;12:437–44.
  225. Van Vugt DA, Meites J. Influence of endogenous opiates on anterior pituitary function. Fed Proc. 1980 Jun;39 (8):2533–8.
  226. Fountas A, Chai ST, Kourkouti C, Karavitaki N. Mechanisms in endocrinology: endocrinology of opioids. European Journal of Endocrinology. 2018 Oct 1;179 (4):R183–96.
  227. Panerai AE, Petraglia F, Sacerdote P, Genazzani AR. Mainly mu-opiate receptors are involved in luteinizing hormone and prolactin secretion. Endocrinology. 1985 Sep;117 (3):1096–9.
  228. Shin SH, Obonsawin MC, Van Vugt DA, Baby N, Jhamandas K. Morphine can stimulate prolactin release independent of a dopaminergic mechanism. Can J Physiol Pharmacol. 1988 Nov;66 (11):1381–5.
  229. Fountas A, Van Uum S, Karavitaki N. Opioid-induced endocrinopathies. Lancet Diabetes Endocrinol. 2020 Jan;8 (1):68–80.
  230. Matera C, Freda PU, Ferin M, Wardlaw SL. Effect of chronic opioid antagonism on the hypothalamic-pituitary-ovarian axis in hyperprolactinemic women. J Clin Endocrinol Metab. 1995 Feb;80 (2):540–5.
  231. de Wit W, Schoute E, Schoemaker J. Chronic naltrexone treatment induces desensitization of the luteinizing hormone pulse generator for opioid blockade in hyperprolactinemic patients. J Clin Endocrinol Metab. 1995 May;80 (5):1739–42.
  232. Steiner J, Cassar J, Mashiter K, Dawes I, Fraser TR, Breckenridge A. Effects of methyldopa on prolactin and growth hormone. Br Med J. 1976 May 15;1 (6019):1186–8.
  233. Kelley SR, Kamal TJ, Molitch ME. Mechanism of verapamil calcium channel blockade-induced hyperprolactinemia. Am J Physiol. 1996 Jan;270 (1 Pt 1):E96-100.
  234. Barbieri C, Ferrari C, Caldara R, Crossignani RM, Bertazzoni A. Endocrine and metabolic effects of labetalol in man. J Cardiovasc Pharmacol. 1981;3 (5):986–91.
  235. Barbieri C, Larovere MT, Mariotti G, Ferrari C, Caldara R. Prolactin Stimulation by Intravenous Labetalol Is Mediated Inside the Central Nervous System. Clinical Endocrinology. 1982;16 (6):615–9.
  236. Lee PA, Kelly MR, Wallin JD. Increased Prolactin Levels During Reserpine Treatment of Hypertensive Patients. JAMA. 1976 May 24;235 (21):2316–7.
  237. Veselinović T, Schorn H, Vernaleken IB, Schiffl K, Klomp M, Gründer G. Impact of different antidopaminergic mechanisms on the dopaminergic control of prolactin secretion. J Clin Psychopharmacol. 2011 Apr 1;31 (2):214–20.
  238. Frantz AG. Prolactin. N Engl J Med. 1978 Jan 26;298 (4):201–7.
  239. Arita J, Kimura F. Direct Inhibitory Effect of Long Term Estradiol Treatment on Dopamine Synthesis in Tuberoinfundibular Dopaminergic Neurons: In Vitro Studies Using Hypothalamic Slices. Endocrinology. 1987 Aug 1;121 (2):692–8.
  240. Reyniak JV, Wenof M, Aubert JM, Stangel JJ. Incidence of hyperprolactinemia during oral contraceptive therapy. Obstet Gynecol. 1980 Jan;55 (1):8–11.
  241. Luciano AA, Sherman BM, Chapler FK, Hauser KS, Wallace RB. Hyperprolactinemia and contraception: a prospective study. Obstet Gynecol. 1985 Apr;65 (4):506–10.
  242. Jernström H, Knutsson M, Taskila P, Olsson H. Plasma prolactin in relation to menstrual cycle phase, oral contraceptive use, arousal time and smoking habits. Contraception. 1992 Dec;46 (6):543–8.
  243. Acién P, Mauri M, Gutierrez M. Clinical and hormonal effects of the combination gonadotrophin-releasing hormone agonist plus oral contraceptive pills containing ethinyl-oestradiol (EE) and cyproterone acetate (CPA) versus the EE-CPA pill alone on polycystic ovarian disease-related hyperandrogenisms. Hum Reprod. 1997 Mar;12 (3):423–9.
  244. Foth D, Römer T. Prolactin serum levels in postmenopausal women receiving long-term hormone replacement therapy. Gynecol Obstet Invest. 1997;44 (2):124–6.
  245. Stanosz S, Zochowska E, Safranow K, Sieja K, Stanosz M. Influence of modified transdermal hormone replacement therapy on the concentrations of hormones, growth factors, and bone mineral density in women with osteopenia. Metabolism. 2009 Jan;58 (1):1–7.
  246. Iancu ME, Albu AI, Albu DN. Prolactin Relationship with Fertility and In Vitro Fertilization Outcomes—A Review of the Literature. Pharmaceuticals. 2023 Jan;16 (1):122.
  247. Trikoilis Ν, Mavromatidis G, Tzafetas M, Deligeoroglou Ε, Tzafetta M, Loufopoulos A, et al. The association of in vitro fertilization/intracytoplasmic sperm injection results with anxiety levels and stress biomarkers: An observational, case-control study✰. Journal of Gynecology Obstetrics and Human Reproduction. 2022 Jan 1;51 (1):102254.
  248. Kamel MA, Zabel G, Bernart W, Neulen J, Breckwoldt M. Comparison between prolactin, gonadotrophins and steroid hormones in serum and follicular fluid after stimulation with gonadotrophin-releasing hormone agonists and human menopausal gonadotrophin for an in-vitro fertilization programme. Hum Reprod. 1994 Oct;9 (10):1803–6.
  249. Meltzer HY, Flemming R, Robertson A. The Effect of Buspirone on Prolactin and Growth Hormone Secretion in Man. Archives of general psychiatry. 1983 Oct;40 (10):1099–102.
  250. Murnane KS, Kimmel HL, Rice KC, Howell LL. The neuropharmacology of prolactin secretion elicited by 3,4-methylenedioxymethamphetamine (“ecstasy”): A concurrent microdialysis and plasma analysis study. Horm Behav. 2012 Feb;61 (2):181–90.
  251. Xue Y, Domino EF. Tobacco/Nicotine and Endogenous Brain Opioids. Prog Neuropsychopharmacol Biol Psychiatry. 2008 Jul 1;32 (5):1131–8.
  252. Li H, Wallin M, Barregard L, Sallsten G, Lundh T, Ohlsson C, et al. Smoking-Induced Risk of Osteoporosis Is Partly Mediated by Cadmium From Tobacco Smoke: The MrOS Sweden Study. J Bone Miner Res. 2020 Aug;35 (8):1424–9.
  253. Hutchinson J, Murphy M, Harries R, Skinner CJ. Galactorrhoea and hyperprolactinaemia associated with protease-inhibitors. Lancet. 2000 Sep 16;356 (9234):1003–4.
  254. Montero A, Fernandez MA, Cohen JE, Luraghi MR, Sen L. Prolactin levels in the cerebrospinal fluid of patients with HIV infection and AIDS. Neurol Res. 1998 Jan;20 (1):2–4.
  255. Redelman D, Welniak LA, Taub D, Murphy WJ. Neuroendocrine hormones such as growth hormone and prolactin are integral members of the immunological cytokine network. Cell Immunol. 2008;252 (1–2):111–21.
  256. Hinterberger-Fischer M, Ogris E, Kier P, Bauer K, Kittl E, Habertheuer KH, et al. Elevation of plasma prolactin in patients undergoing autologous blood stem-cell transplantation for breast cancer: is its modulation a step toward posttransplant immunotherapy? Am J Clin Oncol. 2000 Aug;23 (4):325–9.
  257. DeLeo V, Cella SG, Camanni F, Genazzani AR, Müller EE. Prolactin lowering effect of amphetamine in normoprolactinemic subjects and in physiological and pathological hyperprolactinemia. Horm Metab Res. 1983 Sep;15 (9):439–43.
  258. Wilson JD, King DJ, Sheridan B. Tranquillisers and plasma prolactin. Br Med J. 1979 Jan 13;1 (6156):123–4.
  259. Schettini G, Cronin MJ, O’Dell SB, MacLeod RM. The benzodiazepine agonist diazepam inhibits basal and secretagogue-stimulated prolactin release in vitro. Brain Research. 1984 Jan 23;291 (2):343–9.
  260. Davis JR, Lynam TC, Franklyn JA, Docherty K, Sheppard MC. Tri-iodothyronine and phenytoin reduce prolactin messenger RNA levels in cultured rat pituitary cells. J Endocrinol. 1986 Jun;109 (3):359–64.
  261. Elwes RD, Dellaportas C, Reynolds EH, Robinson W, Butt WR, London DR. Prolactin and growth hormone dynamics in epileptic patients receiving phenytoin. Clin Endocrinol (Oxf). 1985 Sep;23 (3):263–70.
  262. Dana-Haeri J, Oxley J, Richens A. Pituitary responsiveness to gonadotrophin-releasing and thyrotrophin-releasing hormones in epileptic patients receiving carbamazepine or phenytoin. Clin Endocrinol (Oxf). 1984 Feb;20 (2):163–8.
  263. Bonuccelli U, Murialdo G, Rossi G, Bonura ML, Polleri A, Murri L. Prolactin Secretion in Epileptic Subjects Treated with Phenobarbital: Sex Differences and Circadian Periodicity. Epilepsia. 1986;27 (2):142–8.
  264. Cavallo A, Moore DC, Nahori A, Beaumanoir A, Sizonenko PC. Plasma prolactin and cortisol concentrations in epileptic patients during the night. Arch Neurol. 1984 Nov;41 (11):1179–82.
  265. Malhi GS, Tanious M, Das P, Coulston CM, Berk M. Potential mechanisms of action of lithium in bipolar disorder. Current understanding. CNS Drugs. 2013 Feb;27 (2):135–53.
  266. Lanng Nielsen J, Amdisen A, Darling S, Pedersen EB. Plasma prolactin during lithium treatment. Neuropsychobiology. 1977;3 (1):30–4.
  267. Tanimoto K, Maeda K, Yamaguchi N, Chihara K, Fujita T. Effect of lithium on prolactin responses to thyrotropin releasing hormone in patients with manic state. Psychopharmacology (Berl). 1981;72 (2):129–33.
  268. Bekoe EO, Kitcher C, Gyima NAM, Schwinger G, Frempong M. Medicinal Plants Used as Galactagogues. In: Pharmacognosy - Medicinal Plants [Internet]. IntechOpen; 2018 [cited 2023 Oct 12]. Available from: https://www.intechopen.com/chapters/64667
  269. Wuttke W, Jarry H, Christoffel V, Spengler B, Seidlová-Wuttke D. Chaste tree (Vitex agnus-castus)--pharmacology and clinical indications. Phytomedicine. 2003 May;10 (4):348–57.
  270. Prasad SK, Qureshi TN, Qureshi S. Mucuna pruriens seed powder feeding influences reproductive conditions and development in Japanese quail Coturnix coturnix japonica. Animal. 2009 Feb;3 (2):261–8.
  271. Jacobson H. Anti-Lactogenics – Herbs, Meds, Vitamins, and Foods that can undermine milk production - Hilary Jacobson [Internet]. 2022 [cited 2023 Nov 24]. Available from: https://hilaryjacobson.com/course_work/anti-lactogenics-herbs-meds-vitamins-and-foods-that-can-undermine-milk-production/, https://hilaryjacobson.com/course_work/anti-lactogenics-herbs-meds-vitamins-and-foods-that-can-undermine-milk-production/
  272. Mahdi AA, Shukla KK, Ahmad MK, Rajender S, Shankhwar SN, Singh V, et al. Withania somnifera Improves Semen Quality in Stress-Related Male Fertility. Evid Based Complement Alternat Med. 2009 Sep 29;
  273. Brandão Néto J, de Mendonça BB, Shuhama T, Marchini JS, Madureira G, Pimenta WP, et al. Zinc: an inhibitor of prolactin (PRL) secretion in humans. Horm Metab Res. 1989 Apr;21 (4):203–6.

 

Familial Hypercholesterolemia

ABSTRACT

 

Familial hypercholesterolemia (FH) is a prevalent, autosomal co-dominant disorder of lipid metabolism that results in elevated low-density lipoprotein cholesterol (LDL-C) levels and premature atherosclerosis. Screening for and identifying heterozygous FH in childhood is critical, given its high prevalence and asymptomatic presentation. Furthermore, treatment of FH in childhood is effective at lowering LDL-C levels and has the potential to reduce atherosclerotic cardiovascular disease (ASCVD) events in adulthood. Selective screening based on family history had previously been recommended to identify children and adolescents with FH or other lipid disorders. However, studies indicated that many individuals with heterozygous FH were missed with this approach, and therefore in 2011 the National Heart, Lung, and Blood Institute Expert Panel recommended universal screening of children and adolescents between ages 9 and 11 years and again at ages 17 to 21 years, in addition to selective screening, in order to identify pediatric individuals with heterozygous FH. This approach was affirmed in the 2018 American College of Cardiology and the American Heart Association (ACC/AHA) Cholesterol Guidelines, along with endorsing cascade screening as another reasonable approach to identifying children with FH. Once FH is diagnosed, prompt treatment with lifestyle modification should be initiated. When lifestyle interventions are not sufficient, pharmacotherapy using statins has been shown to be effective at lowering LDL-C, generally safe in short and medium- term studies, and may be beneficial at reducing ASCVD events. Other medications can be useful at lowering LDL-C in conjunction with statin therapy, although generally statins are sufficient in young patients. Homozygous FH is a rare disorder manifesting as extremely high LDL-C and ASCVD in childhood, requiring aggressive multimodal management. Overall, studies are needed to determine the optimal timing and intensity of statin therapy, and to better understand long-term safety and ASCVD outcomes in adulthood for lipid-lowering pharmacotherapy initiated in pediatric patients with heterozygous FH.

 

INTRODUCTION

 

Familial hypercholesterolemia (FH), as classically described, is the most common single gene disorder of lipoprotein metabolism and causes severely elevated low-density lipoprotein cholesterol (LDL-C) levels. The prevalence of FH is 1 in 200 to 1 in 300 individuals of different ethnicities (1,2), and it is strongly associated with premature coronary artery disease (CAD) (3). Data from observational studies suggest that untreated FH is associated with ~90-fold increase in mortality due to atherosclerotic cardiovascular disease (ASCVD) in young adults (4). Since early treatment may significantly reduce CAD-related morbidity and mortality in individuals with heterozygous FH (5), early identification and intervention during childhood may greatly improve outcomes in adulthood.

 

EPIDEMIOLOGY

 

The prevalence of FH varies substantially, depending upon the criteria used to define the disorder and the ancestry of the population.  Previously, the prevalence had been described as 1 in 500 individuals based on early work by Drs. Brown and Goldstein (6,7).  More recent analysis of white European populations, which tend to be less ethnically and racially diverse than the US, show higher prevalence rates (8,9).  An analysis using a modified version of the Dutch Lipid Clinic (DLC) criteria applied to participants in the 1999 to 2012 National Health and Education National Surveys (NHANES), a nationally representative survey of the US population, suggest FH affects 1 in 250 US adults (1). The prevalence of FH in US children and adolescents is not as well characterized, although presumably it is similar. Some estimate that as few as 10% of individuals with FH have been identified in the US.

 

PATHOPHYSIOLOGY AND GENETICS

 

FH was initially defined by Brown and Goldstein as a disorder or defect in the LDL receptor (LDL-R) (3). More recently, the description of FH has been expanded and used to describe any defects in LDL-C processing and/or signaling that may lead to a phenotype characteristic of FH (10). FH may be more common and complicated than previously thought, with many different genetic variants leading to pathogenesis. Overall, nine genes are causative for autosomal forms of FH, and up to 50 polymorphic loci contribute to polygenic susceptibility to elevated LDL- cholesterol levels (11).  Some etiologies for the FH phenotype include defects in apoB100 lipoprotein, the major atherogenic lipoprotein component of LDL-C, as well as gain of function mutations in proprotein convertase subtilisin/kexin type 9 (PCSK9), which promotes the degradation of the LDLR, resulting in reduced LDL-C clearance (7). Gene mutations in LDL-R, the apolipoprotein B gene, or in PCSK9 occur in approximately 93, 5, and 2 percent of individuals with a phenotype consistent with FH, respectively (12). The associated impairment in function of these receptors or proteins results in overall reduced clearance of LDL particles from the circulation and elevation in plasma LDL-C. There is also increased uptake of modified LDL by macrophage scavenger receptors, resulting in lipid accumulation in macrophages and foam cell formation, a precursor to atherosclerotic plaque development (13). Thus, the typical lipid profile of FH is characterized by elevated LDL-C (as high as 300 mg/dL) with subsequently increased total cholesterol (TC) levels; in general, triglycerides are normal, and high-density lipoprotein (HDL-C) can be low or normal. FH has an autosomal dominant inheritance pattern which results in hypercholesterolemia and early ASCVD events.

 

 

There are various genetic mutations that can cause FH, which can either be monogenic or polygenic in nature. See Table 1 below for a list of genes that are associated with FH.

 

Table 1. Genes Associated with FH

Monogenic

Autosomal Dominant

LDLR

APOB

PCSK9

APOE*

Autosomal Recessive

LDLRAP1

LIPA (Lysosomal acid lipase deficiency**)

ABCG5 and ABCG8 (Sitosterolemia**)

Polygenic

Genetic variants associated with FH which together can impact LDL-C levels

*Specific mutations or candidate regions associated with FH

**Refer to specific Endotext sections on lysosomal acid lipase deficiency and sitosterolemia for more information

 

In general, homozygotes for mutations in the LDL-R gene are more adversely affected than heterozygotes, reflecting a “gene dosing effect” of inheritance.  Unless there is consanguinity in a family in which heterozygous FH is present, homozygous FH is less prevalent and may affect as many as 1 in 160,000–300,000 individuals (14).  Additionally, the homozygous FH phenotype can be seen in compound heterozygotes which can occur in offspring of unrelated parents due to a different disease-causing mutation on each allele. The severity of the FH phenotype does not necessarily depend upon the presence of true homozygosity or compound heterozygosity inheritance; rather it is determined by the degree of disturbance in LDL metabolism.  For additional information on the genetics as well as pathophysiology, refer to “Familial Hypercholesterolemia: Genes and Beyond” by Warden et al., www.endotext.org (15).

 

FH PHENOTYPE

 

Clinical Symptoms

 

HOMOZYGOUS FH

 

Due to the excessively high plasma LDL-C levels in homozygous FH, cholesterol deposits are common in the tendons (xanthomas) and eyelids (xanthelasmas), and generally appear by one year of age. Tendon xanthomata are most common in the Achilles tendons and dorsum of the hands but can occur at other sites. Tuberous xanthomata typically occur over extensor surfaces such as the knee and elbow.  Planar xanthomas may occur on the palms of the hands and soles of the feet and are often painful.  Xanthelasmas are cholesterol-filled, soft, yellow plaques that usually appear on the medial aspects of the eyelids. Corneal arcus is a white or grey ring around the cornea (16).

 

HETEROZYGOUS FH

 

In addition to increased serum cholesterol and risk for premature coronary artery disease (see below), patients with heterozygous FH may have tendon xanthomas and corneal arcus that appear after the age of 20 years (16).

 

LDL-C Levels

 

In clinical practice, there is not a universal LDL-C threshold that determines a diagnosis of FH. Generally, the level of LDL-C that warrants further evaluation depends upon the age of the patient and whether additional family members have known hypercholesterolemia and/or early ASCVD. As suggested by recent guidelines (7,17), children and adolescents with a negative or unknown family history and LDL-C level persistently ≥190 mg/dL (4.9 mmol/L) suggests FH; in patients with a positive family history of hypercholesterolemia or early ASCVD, an LDL-C level of ≥160 mg/dL(4.1 mmol/L) is consistent with FH (see Table 2).

 

Table 2. Acceptable, Borderline, and High Lipid Levels for Children and Adolescents

Lipid

Low (mg/dl)

Acceptable (mg/dl

Borderline-High (mg/dl)

High (mg/dl)

TC

 

<170

170-199

>200

LDL-C

 

<110

110-129

>130

Non-HDL-C

 

<120

120-144

>145

Triglycerides

0-9 years

10-19 years

 

 

 

 

<75

<90

 

75-99

90-129

 

>100

>130

HDL-C

< 40

>45

 

 

Adapted from the NCEP expert panel on cholesterol levels in children (14)

 

Cardiovascular Disease

 

Cardiovascular risk in FH patients is determined by both the LDL-C concentration and by other traditional risk factors. Homozygotes have early-onset atherosclerosis, including myocardial infarction, in the first decade of life (reported as early as age two years), and are at increased risk for CAD-related mortality in the first and second decades (16).  Additionally, patients with homozygous FH can develop cholesterol and calcium deposits that can lead to aortic stenosis and occasionally to mitral regurgitation.

 

Heterozygotes are also at increased risk for early-onset CAD between the ages of 30-60 years (18). Children with heterozygous FH have thicker carotid intima-media thickness (cIMT), an anatomic measure of arterial thickness associated with atherosclerosis, compared to unaffected siblings and healthy controls (19,20). One study showed those treated with statin medications (HMG-CoA reductase inhibitors) at younger ages had less carotid atherosclerosis compared to the placebo group. Results from long-term studies of statins in children with FH are just emerging, and indicate that statin treatment during childhood may slow progression of cIMT and reduce the risk of CVD in adulthood (21).

 

SCREENING

 

Given the high prevalence of FH and the improved outcomes with early treatment, pediatric lipid screening has become very important for the detection of FH. However, the approach to lipid screening in childhood and adolescences has varied over the past decades and is somewhat controversial, with the US Preventative Services Task Force (USPSTF) recently concluding that that the current evidence is insufficient to assess the balance of benefits and harms of screening for lipid disorders in children and adolescents (22). Selective screening of young individuals with a family history of hypercholesterolemia and/or early CV events or patients at risk for atherosclerosis for other medical conditions has been recommended for several decades (23–25). However, screening individuals based only on family history may miss 30-50% of children with elevated LDL (24–26). Thus, the 2011 Expert Panel, supported by the more recent 2018 ACC/AHA Cholesterol Guidelines (27), recommended universal lipid screening, which involves screening in childhood at two time points, once between ages 9 and 11 years, and then again between ages 17 and 21 years (28). Universal screening is recommended in those not already selectively screened based on family history or personal risk factors (Table 3).

 

Selective screening for FH involves obtaining a fasting or non-fasting lipid panel in individuals ages 2 to 21 years with:

  1. Family history of early atherosclerosis or high cholesterol.
  2. Relatives of individuals with identified FH.

 

Lipid testing should also be performed in the presence of risk factors or medical diagnoses that increase risk for CVD (including hypertension, current cigarette smoking, body mass index ≥ 85th percentile, diabetes mellitus type I and II, chronic kidney disease/end-stage renal disease, chronic inflammatory diseases, human immunodeficiency virus infection, and nephrotic syndrome) (28).

 

Universal screening involves obtaining either a fasting lipid profile or a non-fasting non-HDL, (calculated by subtracting HDL from TC) in childhood at two time points:

  1. Between ages 9-11 years.
  2. Between ages 17-21 years.

 

Table 3. Screening for Hypercholesterolemia

Approach

Age in Years

Population

Selective

2-21

Family history of early atherosclerosis or high cholesterol

Presence of risk factors or medical conditions that increased early CVD risk*

Universal

9-11 and 17-21

All

*Selective screening is indicated in individuals with hypertension, current cigarette smoking, body mass index ≥ 85thpercentile, diabetes mellitus type I and II, chronic kidney disease/end-stage renal disease, chronic inflammatory diseases, human immunodeficiency virus infection, and nephrotic syndrome

 

DIAGNOSIS

 

The diagnosis of FH can be made clinically and through genetic testing; genotype needs to be interpreted in the context of phenotype. For heterozygous FH, the clinical diagnosis is made based on the presence of high levels of total and LDL cholesterol in combination with one or more of following (17):

 

  1. Family history of hypercholesterolemia (especially in children) or known FH.
  2. History of premature CAD in the patient or in family members.
  3. Physical examination findings of abnormal deposition of cholesterol in extravascular tissues (e.g., tendon xanthoma), although these rarely occur in childhood.

 

There are several clinical scoring systems used to diagnose FH, and these vary based on the weight given for each diagnostic criteria (11).  In general, clinical diagnosis of homozygous FH can be made in individuals with the following criteria (14):

 

  1. Untreated LDL-C >500mg/dL (>13 mmol/L) or treated LDL-C ≥300 mg/dL (>8 mmol/L), AND
  2. Cutaneous or tendon xanthoma before age 10 years, OR
  3. Elevated LDL-C levels consistent with heterozygous FH in both parents.

 

There are various disease states or other factors that can increase LDL-C levels and should be considered when diagnosing FH.  Some of these include the following:

 

  • Obesity
  • Hypothyroidism
  • Diabetes mellitus
  • Nephrotic syndrome
  • Chronic renal failure
  • Cholestasis
  • Biliary atresia
  • Hepatitis
  • Biliary cirrhosis
  • HIV infection/AIDS
  • Various drugs/medications
  • Alcohol
  • Pregnancy
  • Very low carbohydrate ketogenic diets

 

Genetic Testing

 

Identifiable gene defects in LDLR, APOB, or PCSK9 have been identified in 60 to 80% of individuals with a heterozygote FH phenotype. Genetic testing has not routinely been performed in the clinical setting due to concerns about cost to the patient and because it was not likely to alter management, given that treatment decisions were usually based on LDL-C levels. However, FH genetic testing has recently been recommended to become the standard of care for patients with definite or probable FH, and the rationale for such testing includes the following: 1) facilitation of definitive diagnosis of FH lowers the concern for other secondary causes of high LDL-C; 2) pathogenic variants correlate with higher cardiovascular risk, which indicates the potential need for more aggressive lipid lowering; 3) increase in initiation of and adherence to therapy; and 4) cascade testing of at-risk relatives (29). Overall, the clinical significance of normal or moderately elevated LDL-C levels in the setting of a genetic defect in the LDLR or other possibly pathogenic defects is unknown.

 

TREATMENT

 

The guidelines for initiating treatment in patients with the FH phenotype are based on age, severity of LDL-C elevation, as well as family and medical histories. Lifestyle therapy is recommended for all children and adolescents with LDL-C levels ≥ 130 mg/dL. If lifestyle intervention is insufficient, medications can be considered in children beginning at age 10 years, or as early as age 8 in high-risk patients and in the presence of a very high-risk family history. For healthy children and adolescents ages 10-21 years, lifestyle therapy should be provided to those with an LDL-C ≥ 130 mg/dL, and medication should be initiated if LDL-C remains ≥ 190 mg/dL despite 6 or more months of lifestyle modification. If there is a family history of early atherosclerotic disease, then medication should be started in individuals with LDL-C levels ≥ 160 mg/dL who do not respond sufficiently to lifestyle modification. If an individual has a high-risk medical condition, as noted above, medication can be considered for those with a persistently elevated LDL-C ≥ 130 mg/dL. In general, the goal of treatment is to maintain an LDL-C level ≤ 130 mg/dL or ≥ 50% reduction in LDL concentration; lower ranges may be considered in high-risk patients. Medications should be initiated in all patients with homozygous FH at the time of diagnosis, regardless of age, and additional treatments should also be considered.

 

Lifestyle Treatment

 

The mainstay of treatment for pediatric lipid disorders is lifestyle modification. A low saturated fat diet, without trans-fat, and high in fruits and vegetables, is the recommended diet for lowering LDL-C. This dietary approach has been shown to be both safe and beneficial in the general pediatric population (28,30,31).  Additionally, nutritional and physical activity interventions have been shown to lower LDL-C and improve CVD risk factors in children with obesity in meta-analyses (32). Despite this, in adults with FH, lifestyle modifications have been shown to only lower LDL-C modestly (33).  Furthermore, the effect of physical activity on LDL-C levels has not been well studied in children with FH.

 

Pharmacotherapy

 

STATINS  

 

The majority of patients with FH are treated with medications, and statins are the recommended first line pharmacotherapy. In a Cochrane meta-analysis of pediatric patients with FH, statins were shown to lower LDL-C by 32% (34). Furthermore, more intensive statin therapy in high doses has been shown to lower LDL-C even more significantly, by up to 50% (35,36).  Follow-up data from a statin trial in pediatric Dutch patients suggest efficacy and safety, as well as decreased atherosclerosis compared to the subjects’ parents (37). Most recently, the use of statins in children with FH have shown reduced CVD risk in adulthood for these patients compared to their untreated parents with FH, after a 20 year follow-up (21). Although this study was not a controlled or placebo study, it suggests that long-term statin use in childhood may prevent ASCVD compared to not treating.

 

Several different formulations of statin therapy are available and approved by the US Food and Drug Administration (FDA) for use in children. Treatment is initiated at a low dose (generally 5-20mg depending on the statin potency), which is given once a day, often at night. If needed, the dose is increased to meet the goals of therapy. Side effects with statins are rare, but include myopathy, new-onset type 2 diabetes mellitus (reported in adult primary prevention statin trials), and hepatic enzyme elevation. In pediatric clinical trials, rates of side effects with statin therapy were low and adherence to statin therapy was generally good (38). Side effects of statins are more likely at higher doses and in patients taking other medications, particularly cyclosporine, azole antifungal agents, and other medications and foods (such as grapefruit) that impact the cytochrome P450 system. Adolescent females should be counseled about the possibility of drug teratogenicity and appropriate contraceptive methods while receiving statin therapy. Additionally, providers should be aware that oral contraceptive pills can increase lipid levels.

 

The NHLBI guidelines recommend the following baseline laboratory evaluation when initiating statin therapy (28):

 

  • Fasting lipid profile.
  • Serum creatine kinase (CK).
  • Hepatic enzymes (i.e., serum alanine aminotransferase [ALT] and aspartate aminotransferase [AST]).

 

Screening for type 2 diabetes is also reasonable prior to starting statins. Fasting lipid profiles are repeated at four weeks after the initiation of statin therapy to titrate dose and are repeated every six months in patients on stable therapy. Liver function tests, CK, and hemoglobin A1C should be obtained if signs of adverse effects arise, and may be obtained at regular intervals, for example after dose changes based on best clinical judgement. Ongoing monitoring of growth, other measures of general and cardiovascular health, and review for the presence of additional ASCVD risk factors, such as smoking exposure, should also occur at each visit.

 

BILE ACID BINDING RESINS

 

Although bile acid binding resins or bile acid sequestrants have been shown to lower LDL-C by ~10-20% in pediatric trials (39,40), they are often difficult to tolerate given their unpalatability and associated adverse effects (such as bloating and constipation) (41). For these reasons, bile acid binding resins are used relatively infrequently. However, they may be useful in combination with a statin for patients who fail to meet target LDL-C levels (42). The sequestrants are not absorbed systemically, remain in the intestines, and are excreted along with bile containing cholesterol. Therefore, they are considered to be very safe. They can be used in patients who prefer to avoid statins, although they may not lower LDL-C sufficiently to achieve goal levels.

 

CHOLESTEROL ABSORPTION INHIBITORS (EZETIMIBE)

 

Ezetimibe is a lipid-lowering mediation that inhibits absorption of cholesterol and plant sterols in the intestines. This agent can be useful in pediatric patients with FH who are not able to reach LDL-C treatment goals on high-intensity statin therapy. Ezetimibe further lowers serum LDL-C and in adults has been shown to improve cardiovascular outcomes without altering the side effect profile (43–45). Specifically in the pediatric population, ezetimibe has been shown to be safe and effective at lowering LDL-C by up to almost 30%, even when used as monotherapy (44,46,47)

 

PCSK9 INHIBITORS

 

PCSK9 inhibitors (evolocumab and alirocumab) are human monoclonal antibodies that bind to PCSK9 and promote plasma LDL cholesterol clearance. In Europe, evolocumab is approved in adolescents (≥12 years old) with homozygous FH. In the US, alirocumab is approved only for use in adult patients, and evolocumab is approved for use in adults with heterozygous FH and in homozygous FH, ages 13 and older, who have not responded to other LDL-C lowering therapies. Overall, PCSK9 inhibitors appear to have a good safety and side effect profile in adults (48).  These medications have been shown to be very effective, reducing LDL-C by more than 15% in patients with homozygous FH and by 35% in patients with heterozygous FH (49–51). The main disadvantage of PCSK9 inhibitors is that they require injection for administration; cost is also a concern.

 

Inclisiran is another medical therapy that targets PCSK9 synthesis through a different mechanism.  It is a small interfering RNA molecule that triggers the breakdown of messenger RNA coding for the PCSK9 protein. This medication has been recently approved for clinical use and has been shown to be safe and effective in adult patients with heterozygous FH (49). Clinical trials of inclisiran are currently underway in adolescent patients with heterozygous and homozygous FH (50).

 

EMERGING MEDICAL THERAPIES

 

There are several promising therapies that aim to reduce LDL-C through different approaches.  These include Bempedoic Acid, Lomitapide, and Evinacumab.  Bempedoic acid blocks the cholesterol biosynthetic pathway upstream of HMG-CoA reductase through inhibition of adenosine triphosphate citrate lyase. This therapeutic agent has been shown to be effective in treating statin-resistant hypercholesterolemia and in reducing ASCVD in adults (51,52). Bempedoic acid is currently being studied in children with heterozygous FH.

 

Microsomal triglyceride transfer protein (MTP) plays an essential role in the formation of apoB-containing lipoproteins and has been shown to be inhibited by Lomitapide (53). This drug can reduce LDL-C by approximately 58% and has been approved for use in adults with homozygous FH.  So far, Lomitapide has been observed to be safe and effective in pediatric patients with homozygous FH (54).

 

Evinacumab is a human monoclonal antibody that targets angiopoietin-like 3 (ANGPTL3), which results in the reduction of LDL-C levels via an LDL-receptor independent mechanism (55–57).  This drug has been shown to significantly reduce LDL-C in patients with homozygous FH who show little to no LDL-receptor activity and who have had poor response to other treatments (58). Additionally, patients with heterozygous FH have also shown improvements in LDL-C by 50% reduction (59).  Evinacumab was approved in early 2021 by the FDA for the treatment of homozygous FH patients starting at age 12 years (60).

 

Lipoprotein Apheresis

 

Although statin therapy has been shown to be effective in reducing LDL and prolonging life expectancy in patients with homozygous FH (61), medical treatment alone may not be adequate to achieve recommended treatment goals. Therefore, it is suggested that lipoprotein apheresis (LA) be initiated in patients with homozygous FH as young as 2 years. The efficacy is dependent upon the type of apheresis but can reduce LDL-C by as much as 45 to 80%. Studies in pediatric patients are limited, but there is some evidence suggesting that LA therapy is safe and effective in children with homozygous FH (62).

 

SUMMARY

 

FH is an autosomal dominant disorder of LDL metabolism that affects 1 in 200 to 300 individuals. Screening involving lipid measurements, family and medical history, and physical examination is needed to identify affected individuals; cascade screening can be helpful. Lifestyle modification is the first-line therapy for hyperlipidemia in pediatric patients but is usually not sufficient to achieve goal LDL levels. Available evidence suggests that treatment with lipid-lowering pharmacotherapy, such as statins, is effective and generally safe in the short and medium-term.  However, further studies are needed to determine the long-term safety and efficacy in preventing ASCVD of lipid lowering medication in pediatric patients with FH.

 

REFERENCES

 

  1. de Ferranti SD, Rodday AM, Mendelson MM, Wong JB, Leslie LK, Sheldrick RC. Prevalence of Familial Hypercholesterolemia in the 1999 to 2012 United States National Health and Nutrition Examination Surveys (NHANES). Circulation. 2016 Mar 15;133(11):1067-72.
  2. Toft-Nielsen F, Emanuelsson F, Benn M. Familial Hypercholesterolemia Prevalence Among Ethnicities-Systematic Review and Meta-Analysis. Front Genet. 2022 Feb 3;13:840797.
  3. Goldstein JL, Schrott HG, Hazzard WR, Bierman EL, Motulsky AG. Hyperlipidemia in coronary heart disease. II. Genetic analysis of lipid levels in 176 families and delineation of a new inherited disorder, combined hyperlipidemia. J Clin Invest Internet. Published online July 1973. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=302426&tool=pmcentrez&rendertype=abstract
  4. Neil A, Cooper J, Betteridge J, Capps N, McDowell I, Durrington P. Reductions in all-cause, cancer, and coronary mortality in statin-treated patients with heterozygous familial hypercholesterolaemia: a prospective registry study. Eur Heart J Internet. Published online 2008. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2577142&tool=pmcentrez&rendertype=abstract
  5. Versmissen J, Oosterveer DM, Yazdanpanah M, Defesche JC, Basart DCG, Liem AH. Efficacy of statins in familial hypercholesterolaemia: a long term cohort study. BMJ Internet. Published online 2008. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2583391&tool=pmcentrez&rendertype=abstract
  6. Brown MS, Goldstein JL. Expression of the familial hypercholesterolemia gene in heterozygotes: mechanism for a dominant disorder in man. Sci Internet. Published online July 5, 1974. http://www.ncbi.nlm.nih.gov/pubmed/4366052
  7. Goldberg AC, Hopkins PN, Toth PP, Ballantyne CM, Rader DJ, Robinson JG. Familial hypercholesterolemia: screening, diagnosis and management of pediatric and adult patients: clinical guidance from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol Internet. Published online 2011. http://www.ncbi.nlm.nih.gov/pubmed/21600525
  8. Watts GF, Shaw JE, Pang J, Magliano DJ, Jennings GLR, Carrington MJ. Prevalence and treatment of familial hypercholesterolaemia in Australian communities. Int J Cardiol Internet. Published online April 15, 2015. http://www.ncbi.nlm.nih.gov/pubmed/25791093
  9. Austin MA, Hutter CM, Zimmern RL, Humphries SE. Familial hypercholesterolemia and coronary heart disease: a HuGE association review. Am J Epidemiol Internet. Published online September 1, 2004. http://www.ncbi.nlm.nih.gov/pubmed/15321838
  10. Hopkins PN, Toth PP, Ballantyne CM, Rader DJ. Familial hypercholesterolemias: prevalence, genetics, diagnosis and screening recommendations from the National Lipid Association Expert Panel on Familial Hypercholesterolemia. J Clin Lipidol Internet. Published online 2011. http://www.ncbi.nlm.nih.gov/pubmed/21600530
  11. Berberich AJ, Hegele RA. The complex molecular genetics of familial hypercholesterolaemia. Nat Rev Cardiol Internet. 2019;16(1):9-20. doi:10.1038/s41569-018-0052-6
  12. Talmud PJ, Shah S, Whittall R, Futema M, Howard P, Cooper JA. Use of low-density lipoprotein cholesterol gene score to distinguish patients with polygenic and monogenic familial hypercholesterolaemia: a case-control study. Lancet. http://www.ncbi.nlm.nih.gov/pubmed/23433573
  13. Epstein FH, Steinberg D, Parthasarathy S, Carew TE, Khoo JC, Witztum JL. Beyond Cholesterol. N Engl J Med Internet. Published online 1989. http://www.ncbi.nlm.nih.gov/pubmed/2648148
  14. Cuchel M, Bruckert E, Ginsberg HN, Raal FJ, Santos RD, Hegele RA. Homozygous familial hypercholesterolaemia: new insights and guidance for clinicians to improve detection and clinical management. A position paper from the Consensus Panel on Familial Hypercholesterolaemia of the European Atherosclerosis Society. Eur Heart J Internet. Published online August 21, 2014. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4139706&tool=pmcentrez&rendertype=abstract
  15. Warden BA, Fazio S, Shapiro MD, et al. Familial Hypercholesterolemia: Genes and Beyond. editors., ed. Endotext Internet South Dartm MA MDTextcom Inc. Published online 2018.
  16. Naoumova RP, Thompson GR, Soutar AK. Current management of severe homozygous hypercholesterolaemias. Curr Opin Lipidol Internet. Published online 2004. http://www.ncbi.nlm.nih.gov/pubmed/15243214
  17. Nordestgaard BG, Chapman MJ, Humphries SE, Ginsberg HN, Masana L, Descamps OS. Familial hypercholesterolaemia is underdiagnosed and undertreated in the general population: guidance for clinicians to prevent coronary heart disease: consensus statement of the European Atherosclerosis Society. Eur Heart J Internet. Published online 2013. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3844152&tool=pmcentrez&rendertype=abstract
  18. Ose L. Familial hypercholesterolemia from children to adults. http://www.ncbi.nlm.nih.gov/pubmed/12652097
  19. Wiegman A, Groot E, Hutten BA, Rodenburg J, Gort J, Bakker HD. Arterial intima-media thickness in children heterozygous for familial hypercholesterolaemia. Lancet. Internet. Published online January 31, 2004. http://www.ncbi.nlm.nih.gov/pubmed/15070569
  20. Tonstad S, Joakimsen O, Stensland-Bugge E, Leren TP, Ose L, Russell D. Risk factors related to carotid intima-media thickness and plaque in children with familial hypercholesterolemia and control subjects. Arter Thromb Vasc Biol Internet. Published online 1996. http://www.ncbi.nlm.nih.gov/pubmed/8696963
  21. Kusters DM, Ph D, Hof MH, Ph D. 20-Year Follow-up of Statins in Children with Familial Hypercholesterolemia. :2019 1547-56.
  22. Association AM. Lipid Screening in Childhood and Adolescence for Detection of Familial Hypercholesterolemia. Evid Rep Syst Rev US Prev Serv Task Force. 2019;98101(6):645-655.
  23. Daniels G SR, F.R. Lipid screening and cardiovascular health in childhood. Pediatr Internet. Published online 2008. http://www.ncbi.nlm.nih.gov/pubmed/18596007
  24. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III. JAMA Internet. Published online May 16, 2001. http://www.ncbi.nlm.nih.gov/pubmed/11368702
  25. National Cholesterol Education Program (NCEP): highlights of the report of the Expert Panel on Blood Cholesterol Levels in Children and Adolescents. Pediatr Internet. Published online 1992. http://www.ncbi.nlm.nih.gov/pubmed/1741227
  26. Dennison BA, Jenkins PL, Pearson TA. Challenges to implementing the current pediatric cholesterol screening guidelines into practice. Pediatr Internet. Published online 1994. http://www.ncbi.nlm.nih.gov/pubmed/8065853
  27. Grundy SM, Stone NJ, Bailey AL, Jones DW, Beam C, Lloyd-jones D. In: AHA / ACC / AACVPR / AAPA / ABC / ACPM / ADA / AGS / APhA / ASPC / NLA / PCNA Guideline on the Management of Blood Cholesterol A Report of the American College of Cardiology / American Heart Association Task Force on Clinical Practice Guidelines WRIT. ; 2018.
  28. Expert panel on integrated guidelines for cardiovascular health and risk reduction in children and adolescents: summary report. Pediatr Internet. Published online 2011. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4536582&tool=pmcentrez&rendertype=abstract
  29. Sturm AC, Knowles JW, Gidding SS, et al. Clinical Genetic Testing for Familial Hypercholesterolemia JACC Scienti fi c Expert Panel. Lipid Screen Child. 2019;1(10):1437-1438.
  30. Niinikoski H, Lagström H, Jokinen E, Siltala M, Rönnemaa T, Viikari J. Impact of repeated dietary counseling between infancy and 14 years of age on dietary intakes and serum lipids and lipoproteins: the STRIP study. Circ Internet. Published online August 28, 2007. http://www.ncbi.nlm.nih.gov/pubmed/17698729
  31. Efficacy and safety of lowering dietary intake of fat and cholesterol in children with elevated low-density lipoprotein cholesterol. The Dietary Intervention Study in Children (DISC). The Writing Group for the DISC Collaborative Research Group. JAMA Internet. Published online May 10, 1995. http://www.ncbi.nlm.nih.gov/pubmed/7723156
  32. Ho M, Garnett SP, Baur L, Burrows T, Stewart L, Neve M. Effectiveness of lifestyle interventions in child obesity: systematic review with meta-analysis. Pediatr Internet. Published online 2012. http://www.ncbi.nlm.nih.gov/pubmed/23166346
  33. Pt K, Se H. Statins for children with familial hypercholesterolemia (Review. Published online 2019.
  34. Vuorio A, Kuoppala J, Kovanen PT, Humphries SE, Tonstad S, Wiegman A. Statins for children with familial hypercholesterolemia. Cochrane Database Syst Rev Internet. Published online 2014. http://www.ncbi.nlm.nih.gov/pubmed/25054950
  35. McCrindle BW, Ose L, Marais AD. Efficacy and safety of atorvastatin in children and adolescents with familial hypercholesterolemia or severe hyperlipidemia: a multicenter, randomized, placebo-controlled trial. J Pediatr. 2003;143(1):74-80. doi:10.1016/S0022-3476(03)00186-0
  36. Avis HJ, Hutten BA, Gagné C, et al. Efficacy and safety of rosuvastatin therapy for children with familial hypercholesterolemia. J Am Coll Cardiol. 2010;55(11):1121-1126. doi:10.1016/j.jacc.2009.10.042
  37. Kusters DM, Avis HJ, Groot E, Wijburg FA, Kastelein JJP, Wiegman A. Ten-year follow-up after initiation of statin therapy in children with familial hypercholesterolemia. JAMA Internet. Published online September 10, 2014. http://www.ncbi.nlm.nih.gov/pubmed/25203086
  38. Braamskamp MJAM, Kusters DM, Avis HJ, Smets EMA, Wijburg FA, Kastelein JJP. Long-term statin treatment in children with familial hypercholesterolemia: more insight into tolerability and adherence. Paediatr Drugs Internet. Published online 2015. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4372689&tool=pmcentrez&rendertype=abstract
  39. Tonstad S, Knudtzon J, Sivertsen M, Refsum H, Ose L. Efficacy and safety of cholestyramine therapy in peripubertal and prepubertal children with familial hypercholesterolemia. J Pediatr Internet. Published online July 1996. http://www.ncbi.nlm.nih.gov/pubmed/8757561
  40. Liacouras CA, Coates PM, Gallagher PR, Cortner JA. Use of cholestyramine in the treatment of children with familial combined hyperlipidemia. J Pediatr Internet. Published online 1993. http://www.ncbi.nlm.nih.gov/pubmed/8441109
  41. McCrindle BW, O’Neill MB, Cullen-Dean G, Helden E. Acceptability and compliance with two forms of cholestyramine in the treatment of hypercholesterolemia in children: a randomized, crossover trial. J Pediatr Internet. Published online 1997. http://www.ncbi.nlm.nih.gov/pubmed/9042130
  42. McCrindle BW, Helden E, Cullen-Dean G, Conner WT. A randomized crossover trial of combination pharmacologic therapy in children with familial hyperlipidemia. http://www.ncbi.nlm.nih.gov/pubmed/12032266
  43. Ip C, Jin D, Gao J, Meng Z, Meng J, Tan Z. Effects of add-on lipid-modifying therapy on top of background statin treatment on major cardiovascular events: A meta-analysis of randomized controlled trials. Int J Cardiol Internet. Published online July 15, 2015. http://www.ncbi.nlm.nih.gov/pubmed/25965621
  44. Kusters DM, Caceres M, Coll M, Cuffie C, Gagné C, Jacobson MS. Efficacy and safety of ezetimibe monotherapy in children with heterozygous familial or nonfamilial hypercholesterolemia. J Pediatr Internet. Published online 2015. http://www.ncbi.nlm.nih.gov/pubmed/25841542
  45. van der Graaf A, Cuffie-Jackson C, Vissers MN, Trip MD, Gagné C, Shi G, Veltri E, Avis HJ, Kastelein JJ. Efficacy and safety of coadministration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia. J Am Coll Cardiol. 2008 Oct 21;52(17):1421-9
  46. Yeste D, Chacón P, Clemente M, Albisu MA, Gussinyé M, Carrascosa A. Ezetimibe as monotherapy in the treatment of hypercholesterolemia in children and adolescents. J Pediatr Endocrinol Metab JPEM. 2009;22(6):487-492. doi:10.1515/jpem.2009.22.6.487
  47. Clauss S, Wai KM, Kavey REW, Kuehl K. Ezetimibe treatment of pediatric patients with hypercholesterolemia. J Pediatr. 2009;154(6):869-872. doi:10.1016/j.jpeds.2008.12.044
  48. Zhang XL, Zhu QQ, Zhu L, Chen JZ, Chen QH, Li GN. Safety and efficacy of anti-PCSK9 antibodies: a meta-analysis of 25 randomized, controlled trials. BMC Med Internet. Published online 2015. http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=4477483&tool=pmcentrez&rendertype=abstract
  49. Raal FJ, Kallend D, Ray KK, et al. Inclisiran for the Treatment of Heterozygous Familial Hypercholesterolemia. N Engl J Med. 2020;382(16):1520-1530. doi:10.1056/NEJMoa1913805
  50. Reijman MD, Schweizer A, Peterson ALH, et al. Rationale and design of two trials assessing the efficacy, safety, and tolerability of inclisiran in adolescents with homozygous and heterozygous familial hypercholesterolaemia. Eur J Prev Cardiol. 2022;29(9):1361-1368. doi:10.1093/eurjpc/zwac025
  51. Goldberg AC, Leiter LA, Stroes ESG, et al. Effect of Bempedoic Acid vs Placebo Added to Maximally Tolerated Statins on Low-Density Lipoprotein Cholesterol in Patients at High Risk for Cardiovascular Disease: The CLEAR Wisdom Randomized Clinical Trial. JAMA. 2019;322(18):1780-1788. doi:10.1001/jama.2019.16585
  52. Nissen SE, Lincoff AM, Brennan D, et al. Bempedoic Acid and Cardiovascular Outcomes in Statin-Intolerant Patients. N Engl J Med. 2023;388(15):1353-1364. doi:10.1056/NEJMoa2215024
  53. Alonso R, Cuevas A, Mata P. Lomitapide: a review of its clinical use, efficacy, and tolerability. Core Evid. 2019;14:19-30. doi:10.2147/CE.S174169
  54. Ben-Omran T, Masana L, Kolovou G, et al. Real-World Outcomes with Lomitapide Use in Paediatric Patients with Homozygous Familial Hypercholesterolaemia. Adv Ther. 2019;36(7):1786-1811. doi:10.1007/s12325-019-00985-8
  55. Adam RC, Mintah IJ, Alexa-Braun CA, et al. Angiopoietin-like protein 3 governs LDL-cholesterol levels through endothelial lipase-dependent VLDL clearance. J Lipid Res. 2020;61(9):1271-1286. doi:10.1194/jlr.RA120000888
  56. Stitziel NO, Khera AV, Wang X, et al. ANGPTL3 Deficiency and Protection Against Coronary Artery Disease. J Am Coll Cardiol. 2017;69(16):2054-2063. doi:10.1016/j.jacc.2017.02.030
  57. Rhee JW, Wu JC. Dyslipidaemia: In vivo genome editing of ANGPTL3: a therapy for atherosclerosis? Nat Rev Cardiol. 2018;15(5):259-260. doi:10.1038/nrcardio.2018.38
  58. Raal FJ, Rosenson RS, Reeskamp LF, et al. Evinacumab for Homozygous Familial Hypercholesterolemia. N Engl J Med. 2020;383(8):711-720. doi:10.1056/NEJMoa2004215
  59. Rosenson RS, Burgess LJ, Ebenbichler CF, et al. Evinacumab in Patients with Refractory Hypercholesterolemia. N Engl J Med. 2020;383(24):2307-2319. doi:10.1056/NEJMoa2031049
  60. Markham A. Evinacumab: First Approval. Drugs. 2021;81(9):1101-1105. doi:10.1007/s40265-021-01516-y
  61. Raal FJ, Pilcher GJ, Panz VR, Deventer HE, Brice BC, Blom DJ. Reduction in mortality in subjects with homozygous familial hypercholesterolemia associated with advances in lipid-lowering therapy. Circulation. 2011;124(20):2202-2207.
  62. Luirink IK, Determeijer J, Hutten BA, Wiegman A, Bruckert E, Schmitt CP. Efficacy and safety of lipoprotein apheresis in children with homozygous familial hypercholesterolemia: A systematic review. J Clin Lipidol. 2019;13(1):31-39.

 

Use of Lipid Lowering Medications in Youth

ABSTRACT

 

The first comprehensive pediatric dyslipidemia guidelines were published by the National Cholesterol Education Program’s Expert Panel on Blood Cholesterol Levels in Children and Adolescents in 1992 and were updated by the American Academy of Pediatrics (AAP) in 1998. In 2008 the AAP issued an updated clinical report detailing recommendations for screening and evaluation of cholesterol levels in children and adolescents as well as prevention and treatment strategies. Since the publication of the first guidelines, rates of pediatric obesity have significantly increased, resulting in a concomitant increase in dyslipidemia. Recently, options for pharmacotherapeutic interventions in pediatric patients have expanded with new FDA approved indications of several lipid lowering medications, as well as additional safety and efficacy data. In 2011, The National Heart Lung and Blood Institute (NHLBI) published its comprehensive report Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents. As with previous guidelines, lifestyle modifications with an emphasis on a heart-healthy diet and daily moderate to vigorous exercise remain an integral part of treatment for pediatric lipid disorders; however, the recommendations for patients requiring management with pharmacotherapy have changed, and will be the focus of this discussion.

 

INTRODUCTION

 

The diagnosis, treatment, and monitoring of dyslipidemia in youth has undergone significant transformations in recent years.  As detailed by the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) and Bogalusa Heart studies, dyslipidemia plays a vital role in both the initiation, as well as the progression of atherosclerotic lesions in children and adolescents (1-3). Because of their role in premature cardiovascular disease, control of dyslipidemia provides clinicians with an opportunity for reducing morbidity and mortality.  Observational data from individuals with genetic mutations that lower atherogenic cholesterol, low-density lipoprotein cholesterol (LDL-C) and non-high-density lipoprotein cholesterol (non-HDL-C), over a lifetime are associated with fewer events and longer life expectancy (4, 5). While these observations are very encouraging, it is not known if achieving the same level of lipid lowering with medications over decades will offer the same protective effects as observed in individuals with life-long lower cholesterol secondary to a genetic mutation (6). Table 1 provides a comparison across the evolution of guideline recommendations for the initiation of pharmacologic intervention with the goal of balancing risk and benefit (7-11). Table 2 details the risk factors and risk conditions described in the NHLBI guidelines (9).

 

Table 1. Comparison of Recommendations for Treatment

Guidelines

NCEP, AAP – 1992 & 1998

AAP – 2008

NHLBI, AAP – 2011

Pharmacologic Treatment Initiation Parameters*

Age > 10 years with LDL-C

Age ≥ 8 years with LDL-C

Ages 10-21 years with LDL-C

 

o

≥ 190 mg/dL

 

o

≥ 190 mg/dL

 

o

≥ 190 mg/dL

 

o

> 160 mg/dL in addition to a positive family history of premature CVD or presence of at least 2 CVD risk factors in the child/adolescent

 

o

≥ 160 mg/dL in addition to a positive family history of premature CVD or presence of risk factors

 

o

160-189 mg/dL in addition to a positive family history of premature CVD or presence of 1 high level risk factor/condition or presence of 2 moderate level risk factors/conditions

 

 

 

 

 

 

 

 

 

 

 

o

≥ 130 mg/dL in addition to presence of diabetes mellitus

 

 

 

 

   

 

 

 

 

 

 

   

Age < 8 years with LDL-C:

 

o

130-159 mg/dL in addition to the presence of 2 high level risk factors/conditions or 1 high level and at least 2 moderate level risk factors/conditions

 

 

   

 

o

≥ 500 mg/dL

 

 

 

   

 

   

 

 

 

 

   

 

   

 

 

 

 

   

 

   

Age < 10 years with severe hyperlipidemia or high-risk conditions associated with serious morbidity

 

 

   

 

   

 

 

 

   

 

   

 

*After an adequate trial of diet and lifestyle management.

 

   

 

   

Ages 8-9 years with LDL-C levels consistently ≥ 190 mg/dL in addition to a positive family history OR presence of risk factors

 

   

 

   

 

 

   

 

   

 

 

 

 

 

 

 

 

Pharmacologic Medication Recommendations

Bile acid sequestrants

Bile acid sequestrants

Statins

 

   

Cholesterol absorption inhibitors

 

 

 

 

 

 

Statins

 

 

 

 

 

Table 2. NHLBI Risk Factors and Risk Conditions (9)

NHLBI, AAP 2011 Guidelines: Risk Factors and Risk Conditions

High Level Risk Factors

•       Hypertension requiring drug therapy

•       Tobacco use

•       BMI ≥ 97th percentile

•       High risk conditions

Moderate Level Risk Factors

•       Hypertension not requiring drug therapy

•       BMI ≥ 95th percentile, < 97th percentile

•       HDL < 40 mg/dL

•       Moderate risk conditions

High Risk Conditions

•       T1DM and T2DM

•       CKD, ESRD, post-renal transplant

•       Post-orthotopic heart transplant

•       Kawasaki disease with current aneurysms

Moderate Risk Conditions

•       Kawasaki disease with regressed aneurysms

•       Chronic inflammatory disease

•       HIV infection

•       Nephrotic syndrome

 

PHARMACOTHERAPEUTIC TREATMENT IN YOUTH

 

The treatment of youth with lipid lowering medications presents some unique challenges and consideration due to their developmental stage, the possibility of extended durations of treatment, and the potential use of concurrent medications that may be counter-productive by increasing lipid levels. As patients progress into adolescence it is particularly important for the patient to understand not only the need for their lipid lowering therapies, but also the consequences of non-compliance. Counseling regarding pharmacotherapy should begin at an early age with developmentally appropriate explanations and expand as patients mature. During adolescence when patients are developing their independence counseling that addresses how to integrate their therapy into their own social norms is important for achieving compliance to both pharmacotherapy as well as lifestyle modifications. Additionally, the cost of therapy significantly impacts compliance and should be factored into therapy decisions especially as youth transition into adulthood and may be faced with changes in insurance coverage. It is also critical to continually readdress the correct use of medications as patients are likely to be on multiple therapies and adolescents will begin some their own medication management as they mature. Regular monitoring for adverse events and side effects of therapy is essential as youth will have a greater lifetime exposure compared to adults and long-term data is generally limited.

 

As with all pharmacotherapy careful consideration should be given to potential drug interactions including those that may increase lipid levels. It is not uncommon for adolescent patients to be prescribed medications which have the potential to negatively impact lipid levels such as systemic steroids or oral contraceptive pills. Each patient case must be evaluated on an individual basis to determine the risk and benefit of prescribing medications which negatively alter lipid levels for patients also utilizing lipid lowering therapies. It should be noted there is significant risk for adolescent females should they become pregnant while taking lipid lowering medications as some have demonstrated a negative impact on fetal development. Adolescent females should be counselled regarding pregnancy and methods of contraception should be discussed. The US Medical Eligibility Criteria for Contraceptive Use compiled by the CDC details several contraceptive methods where the benefits generally outweigh any theoretical or proven risk for patients with hyperlipidemias (12).

 

HMG-CoA REDUCTASE INHIBITORS

 

HMG-CoA reductase inhibitors, or statins, are recommended as first line treatment of youth with severe dyslipidemia who fail non-pharmacologic interventions (i.e., diet and lifestyle modification) (8-11). Statins first debuted in clinical practice in 1987 with the FDA’s approval of lovastatin. At present, there are seven HMG-CoA reductase inhibitors with FDA approval, at varying dosages, for youth with heterozygous familial hypercholesterolemia. Lovastatin, simvastatin, atorvastatin and fluvastatin are approved for children 10 years of age and older. Pitavastatin and pravastatin are approved starting at 8 years of age and rosuvastatin is indicated in children as early as age 6 (13-21). Table 3 provides a summary of HMG-CoA reductase inhibitors, pediatric approval and indications, recommended dosing ranges, comments on dosing, and supporting clinical trials (13-21).

 

Table 3. HMG-CoA Reductase Inhibitors

Medication

Pediatric Approvals & Indications

Dosing

Comments

Supporting

Clinical Trials

Atorvastatin

Age 10-17
Heterozygous familial hypercholesterolemia

10-20 mg/day

May be titrated at ≥ 4-week intervals

McCrindle, et al (22)

Fluvastatin

Age 10-16
Heterozygous familial hypercholesterolemia

20-80 mg/day

May be titrated at ≥ 6-week intervals

van der Graaf, et al (23)

Lovastatin

Age 10-17
Heterozygous familial hypercholesterolemia

10-40 mg/day

Initiated at 20 mg/day for ≥20% LDL reduction, may be titrated at ≥ 4-week intervals

Clauss, et al (24)
Lambert, et al (25)
Stein, et al (26)

Pravastatin

Age 8 and older
Heterozygous familial hypercholesterolemia

20-40 mg/day

Age 8-13: 20 mg/day
Age 14-18: 40 mg/day

Knipscheer, et al (27)
Wiegman, et al (28)
Rodenburg, et al (29)

Rosuvastatin

Age 6 and older
Heterozygous familial hypercholesterolemia

5-20 mg/day

May be titrated at ≥ 4-week intervals

Avis, et al (30)

Simvastatin

Age 10-17
Heterozygous familial hypercholesterolemia

10-40 mg/day

May be titrated at ≥ 4-week intervals

de Jongh, et al (31)
de Jongh, et al (32)

Pitavastatin

Age 8 and older
Heterozygous familial hypercholesterolemia

1-4 mg/day

May be titrated at ≥ 4-week intervals

Ferrari, et al (20)

The above are approved as an adjunct to a diet that is low in cholesterol and saturated fat.  The above agents are approved for both males and females (females must be at least one-year post-menarche) if, despite an adequate diet and other non-pharmacologic measures, the following are present: LDL-C ≥ 190 mg/dL or LDL-C ≥ 160 mg/dL and the patient has a family history of premature cardiovascular disease or two or more cardiovascular disease risk factors.
Abbreviations: mg=milligrams, LDL=low density lipoprotein

 

As Table 4 outlines, statin therapies have demonstrated variable efficacy involving clinical trials of youth (21-32). With the longest half-life, rosuvastatin is the most potent statin, followed by atorvastatin (33). Given this knowledge, if pediatric patients who are initiated on statin therapy are having trouble meeting LDL-C goals, consideration should be given to switching to rosuvastatin or atorvastatin given potency prior to exploring second-line therapy options. Simvastatin is a moderately potent statin at clinically tolerable maximum doses of 40 mg/day (33-35). Lovastatin, pravastatin, and fluvastatin, respectively, are the least potent statins (34, 35). As many studies have demonstrated, reduced potency can be compensated by an increase in the amount of statin given; however, dose escalation is often associated with an increased occurrence of adverse events (21-35). As a result, selection of a specific statin therapy should be individualized and capable of reaching treatment goals. Equally important, consideration should be given to the prevalence and severity of reported side effects (36).

 

Table 4. Statin Therapy Results

Study

Medication

Dose

Results

LDL-C

HDL-C

TC

TG

McCrindle, et al (22)

Atorvastatin

10-20 mg/day

-40%

+6%

-30%

-13%

van der Graaf, et al (23)

Fluvastatin

80 mg/day

-34%

+5%

-27%

-5%

Clauss, et al (24)

Lovastatin

40 mg/day

-27%

+3%

-22%

-23%

Lambert, et al (25)

Lovastatin

10 mg/day

-21%

+9%

-17%

-18%

Lambert, et al (25)

Lovastatin

20 mg/day

-24%

+2%

-19%

+9%

Lambert, et al (25)

Lovastatin

30 mg/day

-27%

+11%

-21%

+3%

Lambert, et al (25)

Lovastatin

40 mg/day

-36%

+3%

-29%

-9%

Stein, et al (26)

Lovastatin

10 mg/day

-17%

+4%

-13%

+4%

Stein, et al (26)

Lovastatin

20 mg/day

-24%

+4%

-19%

+8%

Stein, et al (26)

Lovastatin

40 mg/day

-27%

+5%

-21%

+6%

Knipscheer, et al (27)

Pravastatin

5 mg/day

-23%

+4%

-18%

+2%

Knipscheer, et al (27)

Pravastatin

10 mg/day

-24%

+6%

-17%

+7%

Knipscheer, et al (27)

Pravastatin

20 mg/day

-33%

+11%

-25%

+3%

Rodenburg, et al (29)

Pravastatin

20 mg/day or 40 mg/day

-29%

+3%

-23%

-2%

Wiegman, et al (28)

Pravastatin

20-40 mg/day

-24%

+6%

-19%

-17%

Avis, et al (30)

Rosuvastatin

5 mg/day

-38%

+4%

-30%

-13%

Avis, et al (30)

Rosuvastatin

10 mg/day

-45%

+10%

-34%

-15%

Avis, et al (30)

Rosuvastatin

20 mg/day

-50%

+9%

-39%

-16%

de Jongh, et al (31)

Simvastatin

10-40 mg/day

-41%

+3%

-31%

-9%

de Jongh, et al (32)

Simvastatin

40 mg/day

-40%

+5%

-30%

-17%

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report.  Pediatrics.  2011;128(S5):S213-S256: Table 9-11.
Abbreviations: mg=milligrams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides.

 

Although long term studies evaluating the safety and efficacy of lipid-lowering medications in youth are lacking, results of short term observational and randomized controlled trials are encouraging.  For example, atorvastatin was found to be well tolerated with no statistically significant differences in adverse events reported for either the treatment or placebo groups (22). Additionally, the percentage of patients with abnormal laboratory results was similar for both groups; the only noted difference was an increased percentage of patients with elevated triglycerides in the placebo group. Treatment with atorvastatin resulted in no significant difference in sexual development as assessed by Tanner staging. Van der Graaf and colleagues found that in youth treated with fluvastatin, 58 (68.2%) reported non-serious adverse events. Only four were believed to be drug related. Treatment with fluvastatin resulted in no abnormalities in hormone levels or sexual maturation (23). 

 

In their study of lovastatin, Clauss and colleagues reported no clinically significant alterations in vital signs; growth; hormone levels including luteinizing hormone, follicle-stimulating hormone, dehydroepiandosterone sulfate, estradiol, and cortisol; menstrual cycle length; liver function tests; or muscle function tests (24). Lambert and colleagues also found lovastatin generally well tolerated with no serious clinical adverse effects noted (25). While increased, levels of aspartate aminotransferase did not exceed two times the upper limit of normal and alanine aminotransferase did not display significant changes in study participants. Creatine kinase was elevated, greater than three times the upper limit of normal, in three patients. All subjects remained asymptomatic and the elevated creatine kinase levels resolved spontaneously while no adjustment was need in their medication. Assessment of growth and sexual maturation, by Tanner staging and estimation of testicular volumes, in youth treated with lovastatin found no significant differences between the treatment groups and placebo at either 24 or 48 weeks (26). While the authors reported no significant change in serum hormone levels or biochemical parameters of nutrition, they noted that the study was under powered to detect statistically significant changes in these safety parameters.

 

Use of pravastatin has been shown to have minimal adverse events dispersed evenly between the active drug recipients and those who received placebo (27). Plasma thyroid-stimulating hormone, adrenocorticotropic hormone, cortisol, creatine phosphokinase, alanine aminotransferase, aspartate aminotransferase, total bilirubin, and alkaline phosphatase levels failed to show significant changes from baseline in all treatment groups. Rodenburg and colleagues also evaluated the safety of pravastatin based on annual or biannual evaluation of plasma creatine phosphokinase levels, liver enzymes, sex steroids, gonadotropins, and hormones of the pituitary-adrenal axis (29). Height, weight, age at menarche, Tanner staging, and testicular volume were recorded at baseline and either annually or biannually. Two subjects demonstrated elevated creatine phosphokinase levels which returned to normal without adjustments in therapy, and were presumed to be due to extreme physical exercise. No occurrence of myalgia was associated with elevation in levels of creatinine phosphokinase.  None of the subjects discontinued therapy due to adverse events or laboratory abnormalities.  Similarly, Wiegman and colleagues examined the safety of pravastatin evaluated at baseline, one year, and two years via multiple variables including: sex steroids, endocrine function parameters, height, weight, body surface area, Tanner staging, menarche or testicular volume, alanine aminotransferase, aspartate aminotransferase, and creatine phosphokinase (28). All safety parameters demonstrated no statistically significant differences between active drug recipients verses placebo in changes from baseline.

 

The safety of rosuvastatin was assessed by Avis and colleagues.  Laboratory monitoring including liver enzymes and creatine kinase levels, and markers of growth and development, such as Tanner staging (30). Two serious adverse events were reported including blurred vision in one patient in the placebo group and a vesicular rash progressing to cellulitis in one patient taking rosuvastatin 20 mg. Transaminase levels either remained normal or normalized without permanent discontinuation of treatment. While elevations in creatine kinase and reports of myalgia did occur, symptoms and creatine kinase levels normalized without permanent discontinuation of therapy. Normal progression of height, weight, and sexual development were observed. 

 

The safety of simvastatin during short term therapy has also been reported. Levels of alanine aminotransferase, aspartate aminotransferase, creatine kinase, and physical examination all demonstrated no significant differences between simvastatin treated and placebo participants (31). de Jongh and colleagues evaluated the safety of simvastatin in a second trial by monitoring adverse events as well as changes in alanine aminotransferase, aspartate aminotransferase, and creatine kinase levels (32). Of note, none of the differences reported in events or laboratory values between simvastatin recipients and placebo reached statistical significance. Additionally, there were no statistically significant differences documented for height, body mass index, cortisol levels, testicular size and testosterone levels, menstrual cycle and estradiol levels, and Tanner staging.  Dehydroepiandrosterone sulfate levels demonstrated a statistically significant decrease in the simvastatin group compared to placebo. 

 

Braamskamp and colleagues investigated the efficacy and safety of pitavastatin in pediatric patients diagnosed with hyperlipidemia. 106 patients were enrolled in the study, ages 6-17, for a 12-week period (37). Patients were randomly assigned to 4 different groups categorized by dose 1 mg, 2 mg, 4 mg, or placebo (37). The results showed a reduction in all 3 dose groups in comparison to placebo, the 1 mg group showed a 23.5% reduction in LDL-C, the 2 mg group showed a 30.1% reduction, and the 4 mg group showed a 39.3% reduction (37). There was also a 52-week extension period where patients assigned to the 1 mg group were up-titrated to a maximum dose of 4 mg to try and achieve an LDL-C level of >110 mg/dL (37). There were no safety issues of concern throughout the study. The results indicated that pitavastatin is safe and efficacious for use in pediatric patients, 6-17 years of age, and it was well-tolerated (37).

 

Long-term data regarding the impact of statin therapy on growth, development, and reduction of cardiac risk are limited, particularly for high intensity statin therapy. Recently, Kusters and colleagues evaluated the safety of statin therapy in children and adolescents with familial hypercholesterolemia after 10 years of treatment comparing laboratory safety markers as well as growth and maturation in untreated siblings (36). Only three patients discontinued therapy due to adverse events. Safety parameters such as aspartate aminotransferase, alanine aminotransferase, creatine kinase, estimated glomerular filtration rate, c-reactive protein, and age of menarche did not differ between treated patients and siblings, demonstrating safety over the 10-year treatment period. The authors do note; however, that the study was underpowered to detect the occurrence of rare events (36).

 

When initiating HMG-CoA reductase inhibitor therapy, as with any new medication therapy, it is imperative for clinicians to establish an accurate baseline, monitor for new symptoms, and counsel both patients and family members regarding potential adverse events. Females should be informed about the need to avoid pregnancy and breastfeeding while using statins. Statins may be taken with or without meals, but are commonly given with the evening meal or at bedtime as this has the potential to improve LDL-C reduction (38). As a major substrate of P450, such as CYP3A4, there are multiple drug interactions associated with statin therapy.  Grapefruit juice has gained considerable notoriety as a potential food interaction; however, it should be noted that more than a quart of grapefruit juice would have to be consumed to increase serum statin levels. Of more concern is the possible interaction of gemfibrozil and statins, which should either be avoided as it can increase the toxicity of HMG-CoA reductase inhibitors. Macrolides and antifungal azoles are classes of drugs commonly prescribed to children, and they should be avoided as often as possible as they increase serum statin levels leading to potentially enhanced myopathic effects. Additionally, patients should avoid herbal products and nutraceuticals, such as red yeast rice, which may further enhance adverse effects.

 

BILE ACID SEQUESTRANTS

 

Bile acid sequestrants, or bile acid binding resins, present an additional treatment option for youth with severe dyslipidemia. Bile acid sequestrants represent one of the oldest classes of medications available to treat dyslipidemia and were the only medication recommended in the 1992 NCEP Pediatric Panel Report, at a time when no data were available for statin use in youth (4). While no longer a recommended first-line therapy, bile acid sequestrants do have potential as a treatment option either alone or in combination with a statin (9, 10). At present, colesevelam is the only bile acid sequestrant with FDA approval for youth age 10 years and older with heterozygous familial hypercholesterolemia (39, 40). Despite the lack of FDA approval, both colestipol and cholestyramine have been studied in pediatric patients (41-49).

 

A number of clinical trials have evaluated the bile acid sequestrants in pediatric patients with heterozygous familial hypercholesterolemia and other forms of severe dyslipidemia. While palatability and tolerance remain potential barriers to effective therapy, in general, bile acid sequestrants have demonstrate significant reductions in both total cholesterol and LDL-cholesterol in study subjects (40, 43, 44, 48, 49). Table 5 provides a summary of bile acid sequestrants, pediatric approval and indications, recommended dosing ranges, comments on therapy, and supporting clinical trials. As outlined in Table 6, studies demonstrated some variability in efficacy for the available bile acid sequestrants (40, 43, 44, 48, 49).

 

Table 5. Bile Acid Sequestrants

Medication

Pediatric Approvals & Indications

Dosing

Comments

Clinical Trials

Colesevelam

Age 10-17
Heterozygous familial hypercholesterolemia

1.875 g twice daily or 3.75 g daily

May be used as monotherapy or in combination with a statin

Stein, et al (40)

Colestipol

(Note: Not FDA Approved)
Age 7-12
Primary hypercholesterolemia

5 g twice daily, 10 g daily, or
125-500 mg/kg/day

N/A

McCrindle, et al (43)
Tonstad, et al (44)

(Note: Not FDA Approved)
Age ≥12
Primary hypercholesterolemia

10-15 g/day

N/A

Cholestyramine

(Note: Not FDA Approved)
Age 6-12
Hypercholesterolemia adjunct

240 mg/kg/day divided three times daily before meals

Initiate at 2-4 g twice daily

McCrindle, et al (48)
Tonstad, et al (49)

(Note: Not FDA Approved)
Age ≥12

8 g/day divided twice daily before meals

N/A

Colesevelam is approved as an adjunct to a diet that is low in cholesterol and saturated fat.  Colesevelam is approved for both males and females (females must be at least one-year post-menarche) if, despite an adequate diet and other non-pharmacologic measures, the following are present: LDL-C ≥ 190 mg/dL or LDL-C ≥ 160 mg/dL and the patient has a family history of premature cardiovascular disease or two or more cardiovascular disease risk factors.
Abbreviations: g=grams, mg=milligrams, kg=kilograms, N/A=not applicable.

 

Table 6. Bile Acid Sequestrant Results

Study

Medication

Dose

Results

LDL-C

HDL-C

TC

TG

Stein, et al (40)

Colesevelam

1.875 g/day

-6%

+5%

-3%

+6%

Stein, et al (40)

Colesevelam

3.75 g/day

-13%

+8%

-7%

+5%

McCrindle, et al (43)

Colestipol

10 g/day

-10%

+2%

-7%

+12%

McCrindle, et al (43)

Colestipol & Pravastatin

Colestipol: 5 g/day
Pravastatin: 10 mg/day

-17%

+4%

-13%

+8%

Tonstad, et al (44)

Colestipol

2-12 g/day

-20%

-7%

-17%

-13%

McCrindle, et al (48)

Cholestyramine

8 g/day

-10% to
 -15%

+2% to +4%

-7% to
 -11%

+6% to +9%

Tonstad, et al (49)

Cholestyramine

8 g/day

-17%

+8%

-12%

N/A

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report.  Pediatrics. 2011;128(S5):S213-S256: Table 9-11.
Abbreviations: g=grams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides.

 

Stein and colleagues assessed the safety of colesevelam at weeks 8-26 during an open-label study from.  All subjects received colesevelam 3.75 grams per day in addition to a statin (40). Safety was measured via adverse events, vital signs and physical exam, laboratory monitoring, and Tanner staging.  The most common adverse events related to use of colesevelam were gastrointestinal, including diarrhea, nausea, vomiting, and abdominal pain. It is important to note that no choking or difficulty swallowing were reported with the use of colesevelam. Vital signs, physical exams, laboratory monitoring, and Tanner staging remained the same or progressed as expected throughout the study period. 

 

McCrindle and colleagues evaluated conventional high-dose colestipol versus a combination of low-dose colestipol plus pravastatin, but did not cite safety as an endpoint for their study (40). The researchers did conduct safety monitoring in the form of laboratory tests, physical evaluation, and adverse event reporting. Significant deviations from baseline were noted for alkaline phosphatase, alanine aminotransferase, and aspartate aminotransferase at various time intervals with different medication regimens; however, the authors noted that when compared to reference values, none of the laboratory results were considered abnormal. Study participants in the two medication regimens did not significantly vary in weight gain, height changes, or body mass index. While the majority of patients experienced no adverse events, gastrointestinal symptoms such as constipation, gas or bloating, or stomach ache were more commonly reported by the patients taking the high-dose colestipol. The authors found similar suboptimal compliance with both regimens as determined by medication counts at the end of each study period. Tonstad and colleagues also assessed the tolerability of colestipol granules by monitoring side effects as well as by having subjects’ complete subjective evaluations (44). Side effects associated with colestipol included constipation, dyspepsia, flatulence, nausea, reduction in appetite, and abdominal pain. The subjective evaluations indicated that only 21% of patients liked the taste of the colestipol; however, of those who had previously taken bile acid binding resin, 86% preferred the taste of the newer orange flavored granules. Thirty seven percent of subjects also reported that they frequently forgot to take the medication, while 44% intentionally eliminated the medication from their routine on special occasions or during trips. 

 

The acceptability and compliance of cholestyramine has been studied (48). Eighty two percent (82%) of children preferred the pill formulation of cholestyramine compared to 16% who preferred the powder. Two percent of children in the study preferred neither form of the medication. Compliance was significantly impacted by medication formulation with patients taking the pill form reporting 61% compliance while those on the powder formulation were only 50% compliant. Compliance increased by at least 25% for 42% of patients when they switched to the pill formulation. Tonstad and colleagues assessed the safety of cholestyramine by measuring height velocity, erythrocyte folate, total plasma homocysteine, serum fat-soluble vitamins, and side effects (49). Weight and mean height velocity standard deviation scores were not statistically significant between treatment and placebo groups during the study. The cholestyramine active treatment group demonstrated decreased vitamin D levels and increased homocysteine levels. Differences in erythrocyte folate were not significant between the active treatment and placebo groups. Reported adverse events included intestinal obstruction, abdominal pain, nausea, and loose stools. Unpalatability was a common reason participants withdrew from the study.

 

As demonstrated by the previous studies, while bile acid sequestrants do present an effective therapy option, their side effect profile, issues of tolerability and drug interactions with statins make their use clinically challenging. It is generally recommended that all concurrent medications be given either one hour before or four hours after bile acid sequestrants to prevent decreased absorption of the additional therapies (41, 46). Use of bile acid sequestrants is generally limited to patients optimized on statin therapy who require additional therapy to achieve goal or those that cannot tolerate statins.  Data on long-term safety, however, are generally lacking. It should also be noted that bile acid sequestrants can increase triglyceride levels and should not be used in patients with increased triglyceride levels.

 

FIBRIC ACID DERIVATIVES

 

Experience with fibric acid derivatives in youth is limited. Currently there are no fibric acid derivatives with FDA approval for use in pediatric patients. Both fenofibrate and gemfibrozil are available in the United Sates, but lack pediatric data on safety, efficacy, and dosing (50, 51).  While there is very limited information on the use of bezafibrate in youth, the product is not available in the United States (52). It should be noted that fibric acid derivatives have the potential to increase the incidence in adverse events, such as rhabdomyolysis, when used with statins (45, 50, 51). However, the use of fibrates should be considered and can be beneficial in pediatric patients who also have triglyceride abnormalities (TG levels > 500 mg/dL) (9, 53).

 

NIACIN

 

Niacin provides a potential adjunct therapeutic option for youth with severe dyslipidemia who have not achieved their lipid goal. Extended-release niacin is the only formulation that has FDA-approval for use in children > 16 years of age (54). Despite a lack of FDA approval for ages younger than 16, limited efficacy and safety data are published for the use of niacin in children 10 years of age as older as adjunct therapy (54). Table 7 summarizes data on recommended dosing ranges, comments on dose adjustments, and references supporting clinical trials. 

Colletti and colleagues conducted a retrospective review to evaluate the efficacy and adverse effect profile of niacin for children with severe hypercholesterolemia (54). The effects on serum lipid profiles are detailed in Table 8.  Adverse effects were common, affecting 76% of children, and similar to those reported for adults including: flushing, abdominal pain, vomiting, headache, and elevated liver enzymes. Due to the high prevalence of adverse effects, use of niacin should be limited to patients not achieving goal with other therapies or those who cannot tolerate alternative adjunctive options. As with fibrates, niacin can also be considered for the purposes of treating pediatric patients who are concurrently diagnosed with hypertriglyceridemia (9).

 

Table 7. Niacin

Medication

Pediatric Approvals & Indications

Dosing

Comments

Clinical Trials

Niacin

Extended release: >16 years of age

Note: Immediate release is not FDA Approved:
Age ≥ 10
Adjunct therapy

Initial: 100-250 mg/day
(Max: 10 mg/kg/day)
divided three times daily with meals

May titrate weekly by 100 mg/day or every 2-3 weeks by 250 mg/day

Colletti, et al (54)

Abbreviations: mg=milligrams, kg=kilograms.

 

Table 8. Niacin

Study

Medication

Dose

Results

Colletti, et al (54)

Niacin

500-2,250 mg/day

LDL-C

HDL-C

TC

TG

-17%

+4%

-13%

+13%

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report.  Pediatrics. 2011;128(S5):S213-S256: Table 9-11.
Abbreviations: mg=milligrams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides.

 

EZETIMIBE

 

Ezetimibe is FDA approved for adolescents 10 years of age and older with FH (53, 55), and it presents a potential therapy option either as monotherapy or when synergistically paired with an HMG-CoA reductase inhibitor (56-58). Due to its favorable tolerability, it has become the most frequently used second-line agent (59). Table 9 summarizes data on recommended dosing ranges and references supporting clinical trials while Table 10 details efficacy of therapy. 

Tolerability of ezetimibe was prospectively evaluated by Yeste and colleagues via a combination of biochemical markers and adverse event reports (56). No change was seen in hemogram, transaminases, creatinine, calcium, phosphorus, and vitamins A and E for any of the 17 patients. Additionally, there were no reports of adverse events during the study period. Clauss and colleagues retrospectively evaluated ezetimibe; therefore, safety parameters were less defined, but included intermittent measurement of liver enzymes, occasional CK levels, and adverse event reports (57). There were no reported abnormalities in liver enzymes for study participants. Ultimately, one patient was discontinued from ezetimibe therapy for asymptomatic elevated CK levels, later determined to be likely unrelated to therapy.

 

Table 9. Ezetimibe

Medication

Pediatric Approvals & Indications

Dosing

Comments

Clinical Trials

Ezetimibe

Age ≥10
Homozygous familial hypercholesterolemia

10 mg/day

N/A

Yeste, et al (56)
Clauss, et al (57)
van der Graaf, et al (58)

Abbreviations: mg=milligrams, N/A=not applicable.

 

Table 10. Ezetimibe

Study

Medication

Dose

Results

Yeste, et al (56)

Ezetimibe

10 mg/day

            LDL-C

HDL-C

TC

TG

PH

-42%

N/A

-31%

N/A

FH

-30%

-15%

-26%

N/A

Clauss, et al (57)

Ezetimibe

10 mg/day

            LDL-C

HDL-C

TC

TG

FH

-28%

N/A

-22%

N/A

FCHL

N/A

-13%

N/A

van der Graaf, et al (58)

Ezetimibe
&
Simvastatin

Ezetimibe: 10 mg/day
Simvastatin: 10-40 mg/day

            LDL-C

HDL-C

TC

TG

              -49%

+7%

-38%

-17%

                 

Adapted from National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report.  Pediatrics. 2011;128(S5):S213-S256: Table 9-11.
Abbreviations: mg=milligrams, LDL-C=low density lipoprotein cholesterol, HDL-C=high density lipoprotein cholesterol, TC=total cholesterol, TG=triglycerides, PH=polygenic hypercholesterolemia, FH=familial hypercholesterolemia, FCHL=familial combined hyperlipidemia.

 

Van der Graaf and colleagues assessed the safety of combination therapy with ezetimibe and simvastatin based on reported adverse events as well as laboratory monitoring and clinical examination (58). After 53 weeks, 71% of study participants reported some types of treatment-emergent adverse events. Of those events reported, only influenza, nasopharyngitis, and headache occurred in greater than 5% of participants. Consecutive transaminase elevations of at least three times the upper limit of normal were reported in 6 participants; however, all values resolved with interruption or discontinuation of therapy.  Elevations in creatine phosphokinase occurred infrequently and were not associated with myalgia at levels greater than three times the upper limit of normal.  Height, weight, and sexual maturation were not significantly impacted by therapy. Ezetimibe affords flexibility in administration time with the ability to administer it without regard to meals or time of day (45). HMG-CoA reductase inhibitors have the risk of increasing myopathy and elevation in hepatic transaminases, but are generally considered a safe combination with ezetimibe.

 

OMEGA-3 FISH OILS

 

Omega-3 fish oils are a class of therapy for which there is significantly limited data in youth. To date the FDA approved formulations of omega-3 fatty acid lack a pediatric indication. High dose omega-3 fatty acid supplementation was evaluated by de Ferranti and colleagues, but ultimately the authors found no statistically significant improvement when subjects were compared to placebo (60). Chahal and colleagues similarly found no significant impact on hypertriglyceridemia when treating pediatric patients with fish oil (61).

 

Khorshidi and associates performed a systematic review and meta-analysis of the effect of omega-3 supplementation on lipid profiles in children and adolescents. They found that omega-3 supplementation improved triglyceride levels in patients diagnosed with hypertriglyceridemia that were less than or equal to 13 years of age; however, there was no significant effect seen in HDL, LDL, or TC values (62). Omega-3 fish oils can be considered in those who have elevated triglyceride levels.

 

FAMILIAL HYPERCHOLESTEROLEMIA AND THERAPEUTIC ADVANCES

 

Familial hypercholesterolemia (FH) is a common, but often misdiagnosed inherited gene disorder (63). The most common gene mutation seen in FH is in the low-density lipoprotein receptor gene (LDLR) accounting for 85-90% of cases, followed by the apolipoprotein (ApoB) gene (5-15% of cases) and the proprotein convertase subtilisin kexin 9 (PCSK9) genes (1% of cases) (63, 64). Patients who are diagnosed with FH have abnormally elevated LDL levels from birth. FH is associated with a twenty-fold increased risk in premature cardiovascular disease and cardiovascular events (65). There are two different types of FH, heterozygous (HeFH) and homozygous (HoFH). Heterozygous patients have one mutated allele and are more commonly seen in practice, while homozygous patients have two mutated alleles and are very rare (66). Distinction between the two types is imperative because HoFH patients tend to be treatment resistant and carry a worse prognosis, if left untreated, these patients rarely make it past age 20 (66).

 

Lipid lowering in these patients can be quite challenging, especially HoFH patients. Most typical therapies for lipid lowering require functional LDL receptors, therefore given the gene mutations which often render the receptors inactive, modest reductions of 10-25% in HoFH patients are usually all that will be gained (67). HeFH patients tend to see higher rates of reduction (25-40%) (68). However, first line treatment for both forms of FH is still high-intensity statin therapy at moderate to high doses to be initiated as early as age 8 (69). All seven statins are FDA-approved for the treatment of FH and have been proven to slow down the progression of carotid intima-media thickness (63). Statins also reduce the incidence of cardiovascular events and cardiac death (63). Until recently, long term data on the use of statins in this patient population was not available, but in 2019, a 20-year follow-up study of statins in pediatric patients with FH was published (70). The results found that the incidence of cardiovascular events and death was much lower in patients treated with statins (70). If LDL-C goals are not being met with the use of statins alone, the next recommend agent is ezetimibe; however, this should not be used in patients younger than 10 years of age (59).

 

Ezetimibe is the second-line option to statins in these patients. It’s use in combination with statins has demonstrated a reduction of LDL-C levels below 135 in more than 90% of children with FH (71). In one of the only studies that assessed the coadministration of simvastatin and ezetimibe in children with HeFH, it was found to be safe, well tolerated and provided a higher LDL reduction (15%) compared with simvastatin alone in HeFH patients (72). However, when it was investigated as a monotherapy option for children with HeFH, it only produced LDL-C lowering of 27% (71, 73). It is more appropriate to use as an adjunct therapy in this population of patients.

 

Alternative medications that can be considered other second-line options are bile acid sequestrants (71). Colesevelam is safe to use, but limited to children >10 years of age (71). These drugs however have minimal LDL-C lowering effects, usually only seeing a 10-20% reduction, and more importantly are very poorly tolerated due to gastrointestinal side effects (71).

 

PCSK9 Inhibitors

 

Given the difficulty and importance of treating these patients, especially those with HoFH, there is a need for stronger lipid lowering options, which is where PCSK9 inhibitors come into play. These are a more recent class addition to the therapy options for managing FH, which help to reduce the degradation of LDL receptors and the removal of LDL-cholesterol (74). In a recent study assessing the efficacy/safety of lipid-lowering agents in patients with familial hypercholesterolemia, it was concluded that PCSK9 inhibitors were the most effective in lowering lipid levels (75). They have none of the same side effects as statins and produced similar CV benefits. Therefore, based on these conclusions, PCSK9 inhibitors are recommended as first-line agents in patients with hypercholesteremia that have intolerances or resistance to statins (75). There is currently one PCSK9 inhibitor approved for pediatric use. Repatha (evolocumab) was originally approved for use in patients 13 years of age and older (76), but the HAUSER-RCT study assessed the use of Repatha in patients ages 10-17 years of age for 24 weeks. It showed that the drug improved lipid levels (by approximately 38% in HeFH patients and 21-24% in HoFH patients) and was safe for use as the incidence of adverse events was similar in both the drug and placebo groups (71, 77). So now it is considered safe to use in pediatric patients 10 years of age and older. The HAUSER-RCT trial was then continued for another 80 weeks to further assess the safety and efficacy of Repatha (78). This new trial, HAUSER-OLE, further confirmed that the drug was safe and well-tolerated (78).

 

Praluent (Alirocumab) is another PCSK9 inhibitor that is available for treatment of FH, however it is not currently approved for use in pediatric patients (79). Nevertheless, there are studies currently assessing the safety and efficacy within this population. Bruckert and associates utilized Praluent and conducted an open-label phase 3 study specifically in pediatric patients (8-17 years of age) diagnosed with HoFH that were inadequately controlled (80). Patients received 75 or 150 mg of the drug based on weight (<50 or >50 kg, respectively) every 2 weeks for 12 weeks (80). The primary endpoint was percent change in LDL-C levels from week 0 to 12 (80). Interestingly, the results showed only a 4.1% decrease in LDL-C levels by week 12 (80). The secondary endpoints (assessing percent change LDL-C levels from baseline to weeks 24 and 48, changes in other lipid parameters from baseline to weeks 12, 24, 48, patients with a reduction of more than 15% in LDL-C levels at weeks 12, 24 and 48, and absolute change in LDL-C from baseline to weeks 12, 24, and 48) produced incredibly variable results (80). Overall, there were quite small changes in LDL-C levels observed in this study with mean reductions of LDL-C levels noted to range anywhere from ~33 to 52 mg/dL (80). More importantly, previous studies have shown that PCSK9 inhibitors are linked to a decrease in major coronary/vascular events and all-cause mortality, so although the results produced small values, the changes seen are still clinically significant based on these added benefits (80). It was also noted that this study produced similar results when compared to the ODYSSEY study which assessed the use of Praluent in adult patients with HoFH (80). The drug was deemed safe and there were no issues with tolerability (80). The study supports the use of Praluent as an adjunct therapy in HoFH patients already on first- and second-line therapies and not reaching their goal LDL-C levels (80).

 

Another study assessed the use of Praluent in pediatric patients diagnosed with HeFH. The ODYSSEY KIDS study was a phase 2 dose-finding study that enrolled pediatric patients anywhere from 8-17 years of age (81). Patients were split into 4 cohorts and dosed every 2 weeks. Dosing was determined by weight and the primary endpoint assessed percent change in LDL-C from baseline to week 8 (81). Praluent demonstrated the best reduction in LDL-C levels in the highest dosed cohorts and was well-tolerated. This study also supported the use of the drug (with further analysis) in patients who require adjunct therapy, there is a phase 3 trial planned to assess the doses from this study that resulted in the greatest reduction in LDL-C levels. Overall, it is important to note that HoFH patients are more likely to fail PCKS9 inhibitors (82). This is attributable to their mechanism of action.  This class of medication requires functional LDL receptors, and this is impaired or completely absent in HOFH patients (82). Therefore, effectiveness of PCSK9 inhibitors tends to be much higher in HeFH patients (82).

 

Leqvio (inclisiran) is also another PCSK9 inhibitor currently not approved for pediatric use (83). The mechanism of action of this drug differs from Repatha and Praluent. Leqvio is a small interfering RNA (siRNA) that utilizes the RNA interference mechanism to cause the catalytic breakdown of mRNA for PCSK9, thus stopping the translation of the protein (84). It also only requires administration twice yearly as opposed to biweekly (84). There are currently ongoing studies investigating the possibility of using Leqvio in pediatric patients. ORION-13 and ORION-16 are studies assessing the efficacy, safety and tolerability of Leqvio in pediatric patients diagnosed with HoFH and HeFH, respectively (84). They are two-part (1-year double blind, the other year open-label) phase 3 trials consisting of patients aged 12 to <18 years with FH (84). The primary endpoint is the percentage change in LDL-C from baseline to day 330 (84). Based on the results, this could be another drug option as adjunct therapy to consider for use.

 

Angiopoietin-Like Protein 3 (ANGPTL3)

 

Angiopoietin-like protein 3 (ANGPTL3) also presents a novel target of adjunctive therapy for patients with homozygous familial hypercholesterolemia that are not meeting LDL-C goals with first-line agents (85, 86).  Evkeeva (evinacumab-dgnb) is a monoclonal ANGPTL3 inhibitor that is FDA-approved specifically for the adjunctive treatment of homozygous familial hypercholesterolemia in patients 5 years of age and older (87).

 

Lomitapide

 

Lomitapide is another potential treatment option for patients with HoFH. This medication works differently from more conventional options. It binds to microsomal triglyceride transfer protein (MTP) and prevents the production of lipoproteins that contain apo-B (88). This causes a decrease in the production of very-low-density lipoprotein (VLDL) and chylomicrons. Since VLDL is converted into LDL, this mechanism ultimately causing a decrease in LDL-C levels (89). It is administered once daily at doses ranging from 5 to 60 mg (88). The side effect profile of lomitapide can be difficult for patients as it can cause severe gastrointestinal side effects (due to the decrease in absorption of fats in the intestines), most often diarrhea (89). But it is also associated with raised hepatic fats and enzymes (82). It is currently approved for adult use only, but it has become an option for use in pediatric patients through an expanded access program or a named patient basis (82). There was a case series done exploring the effect of lomitapide in 11 pediatric patients diagnosed with HoFH. It demonstrated that the drug was effective in reducing LDL-C with all 11 HoFH patients and showed a similar side effect profile to that seen in adult patients (82). GI complaints were moderated and did not cause any discontinuation of use (82). It also showed greater reduction in LDL-C levels at lower doses (82). The greatest benefit of lomitapide was associated with its ability to reduce or stop the need for lipoprotein apheresis in the patients incorporated in this case study (82). An interesting mention about lomitapide from the case series is that adult patient data shows that early intervention utilizing the drug showed a potential for increased life expectancy and a delay in the time to first major adverse cardiovascular event (82). There is also currently an ongoing phase 3, open label trial investigating the efficacy and safety of lomitapide in pediatric patients with HoFH, estimated completion date is April of 2024 (82).

 

Bempedoic Acid

 

Bempedoic acid is a new medication that exerts its effects very similarly to that of statins. It works in the same pathway as statins and targets cholesterol biosynthesis (90-92). It is administered however as a prodrug and converted to active drug only in the liver and not in the muscles (90-92). The other difference between the two classes is that bempedoic acid inhibits ATP-citrate lyase (ACL), while statins inhibit HMG CoA reductase (90-92). Due to the lack of activation in skeletal muscles, this drug is a promising alternative to patients unable to take statins due to muscle related symptoms (90-92). The medication is FDA approved for use in patients with HeFH and those with established cardiovascular disease (93). It has shown promising results in adult trials, but there are currently no published pediatric trials to date assessing the safety or efficacy of use of the drug (93). There does however appear to be a trial in development: “An Open-Label Study to Evaluate the Pharmacokinetics, Pharmacodynamics, and Safety of Bempedoic acid in Pediatric Patients with Heterozygous Familial Hypercholesterolemia.” The results are highly anticipated so that this can offer another promising drug class for use in patients intolerant or unable to meet their LDL-C goals.

 

FAMILIAL CHYLOMICRONEMIA SYNDROME

 

Familial chylomicronemia syndrome (FCS) is an incredibly rare autosomal recessive gene disorder (94). There is reduced or absent lipoprotein lipase activity causing disruption in chylomicron metabolism leading to severely elevated triglyceride levels resulting in acute recurrent pancreatitis (94). There is not however an increased risk of ASCVD with an FCS diagnosis (94). The best way to treat FCS is also often referred to as the most difficult as it requires patients to restrict dietary intake to <10-15% of daily calories (94). Other treatment options utilized are fibrates, omega-3 fatty acids and statins with variable responses, but the use of these medications is most commonly seen in patients who have multifactorial chylomicronemia syndrome (94). Given the difficulty of ensuring these patients maintain low levels of triglycerides, medications like volanesorsen are being examined (94, 95).

 

Volanesorsen

 

Volanesorsen is a second-generation 2’-O-methoxyethyl (2’-MOE) antisense therapeutic oligonucleotide. It works by inhibiting apoC3 thus lowering triglyceride plasma levels (94). When it binds to apoC3, this interrupts mRNA translation which consequently promotes triglyceride clearance/lowering of triglyceride plasma levels (94). The efficacy and safety of volanesorsen was assessed in the APPROACH study (96). It included 67 patients that were randomized to either weekly volanesorsen or placebo for 3 months (97). The results showed a 77% reduction in triglyceride plasma levels at the end of the study period and there was only 1 event of pancreatitis in the study group (97). The largest trial performed assessing the use of volanesorsen was the COMPASS trial (97). It included 114 patients who were randomized to either weekly injections of volanesorsen or placebo for a total of 26 weeks (97). The results showed that patients in the treatment group saw a reduction in triglyceride levels, chylomicron triglycerides, VLDL levels and apoC3 levels by more than 70% (97). There were also no occurrences reported of pancreatitis in any of the patients randomized to the volanesorsen group (93). In both trials, volanesorsen proved itself as a promising agent for treatment of hypertriglyceridemia in FCS patients (95-98). This drug is not approved for use in the US but is approved in other countries.

 

CONCLUSION

 

As noted in the 2011 NHLBI’s guidelines, available information regarding the treatment of youth with lipid disorders has greatly expanded. HMG-CoA reductase inhibitors, or statins, are now considered first-line pharmacologic treatment of children and adolescents with severe hypercholesterolemia who fail treatment with diet and exercise alone, although statins are only FDA approved for youth with familial hypercholesterolemia. Despite their ability to effectively reduced cholesterol levels, use of bile acid sequestrants continue to pose challenges for pediatric patients due to their unpalatability and are typically utilized as adjunctive therapy or for patients not able to tolerate statins. Fibric acid derivatives, as a class of medications, not only lack an FDA approved agent, but also continue to lack significant pediatric safety and efficacy data. Niacin, a potential adjunct therapy, lacks FDA approval for pediatric patients and is plagued by significant adverse effects, making it an unlikely therapeutic option for youth.  Ezetimibe provides clinicians with an alternative adjunct therapy option when synergistically paired with an HMG-CoA reductase inhibitor or used as monotherapy for patients intolerant to statins and bile acid sequestrants. Despite their inherit appeal and popularity amongst the lay public, omega-3 fish oils have failed to demonstrate statistically significant cholesterol lowering in pediatric and adolescent patients, but can be used to lower triglyceride levels. PCSK9 and ANGPTL3 inhibiting agents are promising novel treatment options in pediatric patients diagnosed with FH. While recent years have witnessed a dramatic increase in studies of lipid lowering medications in youth, the long-term safety and efficacy data continue to present an active focus of research.

 

REFERENCES

 

  1. McMahan CA, Gidding SS, Malcom GT, Tracy RE, Strong JP, McGill Jr HC.  Pathobiological determinants of atherosclerosis in youth risk scores are associated with early and advanced atherosclerosis. (PDAY) Pediatrics.  2006;118(4): 1447-1455.
  2. Newman III WP, Freedman DS, Voors AW, et al.  Relation of serum lipoprotein levels and systolic blood pressure to early atherosclerosis. (The Bogalusa Heart Study).  N Engl J Med. 1986;314(3):138-44.
  3. Berenson GS, Srinivasan SR, Bao W, Newman III WP, Tracy RE, Wattigney WA.  Association between multiple cardiovascular risk factors and atherosclerosis in children and young adults. (The Bogalusa Heart Study). N Engl J Med. 1998;338(23):1650-1656.
  4. National Cholesterol Education Program (NCEP): Highlights of the Report of the Expert Panel on Blood Cholesterol Levels in Children and Adolescents.  Pediatrics.  1992;89:495-501.
  5. American Academy of Pediatrics. Committee on Nutrition. Cholesterol in childhood. Pediatrics.  1998;101 (1 Pt 1): 141-7.
  6. Daniels SR, Greer FR. Lipid Screening and cardiovascular health in childhood. Pediatrics. 2008;122:198-208.
  7. National Center for Chronic Disease Prevention and Health Promotion, Division of Adolescent and School Health.  Youth Risk Behavior Sheet(YRBS): National Trends, 1991-2009: Obesity, Dietary Behaviors, and Weight Control Practices.  http://www.cdc.gov/healthyyouth/yrbs/pdf/us_obesity_trend_yrbs.pdf.  Updated October 5, 2011.  Accessed December 2011.
  8. McCrindle BW, Urbina EM, Dennison BA, Jacobson MS, Steinberger J, Rocchini AP, Hayman LL, Daniels SR. American Heart Association scientific statement: drug therapy of high-risk lipid abnormalities in children and adolescents. Circulation. 2007;115:1948-67.
  9. National Heart Lung and Blood Institute (NHLBI): Expert Panel on Integrated Guidelines for Cardiovascular Health and Risk Reduction in Children and Adolescents: Summary Report.  Pediatrics.  2011;128(S5):S213-S256.
  10. Kavey RW, Allada V, Daniels SR, Hayman LL, McCrindle BW, Newburger JW, Parekh    RS, Steinberger J. American Heart Association scientific statement: cardiovascular risk reduction in high-risk pediatric patients. Circulation. 2006;114:2710-38.
  11. Sibley C, Stone NJ. Familial hypercholesterolemia: a challenge of diagnosis and therapy. Cleve Clin J Med. 2006;73:57-64.
  12. Division of Reproductive Health, National Center for Chronic Disease Prevention and Health Promotion. US Medical Eligibility Criteria for Contraceptive Use, 2016 (US MEC). https://www.cdc.gov/reproductivehealth/contraception/mmwr/mec/summary.html. Updated March 27, 2023.Accessed November 2023.
  13. Atorvastatin product monograph. Pfizer. New York, NY. 2007.
  14. Fluvastatin product monograph. Novartis. East Hanover, NJ. 2006.
  15. Lovastatin product monograph. Merck & Co. Whitehouse Station, NJ. 2007.
  16. Pravastatin product monograph. Bristol-Myers Squibb Company. Princeton, NJ. 2007.
  17. Rosuvastatin product monograph. AstraZeneca. Wilmington, DE. 2011.
  18. Simvastatin product monograph. Merck & Co. Whitehouse Station, NJ. 2006.
  19. Pitavastatin product monograph. Kowa. Montgomery, AL. 2022.
  20. Ferrari F., Martins V.M., Rocha V.Z., Santos R.D. Advances with lipid-lowering drugs for pediatric patients with familial hypercholesterolemia. Expert Opin. Pharmacother. 2021;22:483–495. doi: 10.1080/14656566.2020.1832991.
  21. Haney EM, Huffman LH, Bougatsos C, Freeman M, Steiner RD, Nelson HD. Screening and treatment for lipid disorders in children and adolescents: systematic evidence review for the US preventive services task force. Pediatrics. 2007;120:e189-214.
  22. McCrindle BW, Ose L, Marais AD. Efficacy and safety of atorvastatin in children and adolescents with familial hypercholesterolemia or severe hyperlipidemia: a multi-center, randomized, placebo-controlled trial. J Pediatr. 2003;142:74-80.
  23. van der Graaf A, Nierman MC, Firth JC, Wolmarans KH, Marais AD, de Groot E.  Efficacy and safety of fluvastatin in children and adolescents with heterozygous familial hypercholesterolemia.  Acta Paediatr.  2006;95(11):1461-1466.
  24. Clauss SB, Holmes KW, Hopkins P, Stein E, Cho M, Tate A, Johnson-Levonas AO, Kwiterovich PO. Efficacy and safety of lovastatin therapy in adolescent girls with heterozygous familial hypercholesterolemia. Pediatrics. 2005;116(3):682-8.
  25. Lambert M, Lupien PJ, Gagné C, et al.  Treatment of familial hypercholesterolemia in children and adolescents: effect of lovastatin.  Pediatrics.  1996;97(5):619-628.
  26. Stein EA, Illingworth DR, Kwiterovich Jr PO, et al.  Efficacy and safety of lovastatin in adolescent males with heterozygous familial hypercholesterolemia.  JAMA.  1999;281(2):137-144.
  27. Knipscheer HC, Boelen CCA, Kastelein JJP, et al.  Short-term efficacy and safety of pravastatin in 72 children with familial hypercholesterolemia.  Pediatr Res.  1996;39(5):867-871.
  28. Wiegman A, Hutten BA, de Groot E, et al.Efficacy and safety of statin therapy in children with familial hypercholesterolemia: a randomized controlled trial. JAMA.  2004;292(3):331-337.
  29. Rodenburg J, Vissers MN, Wiegman A, et al.  Statin treatment in children with familial hypercholesterolemia: The younger, the better.  Circulation. 2007;116:664-668.
  30. Avis HJ, Hutten BA, Gagné C, et al.  Efficacy and safety of rosuvastatin therapy for children with familial hypercholesterolemia.  J Am Coll Cardiol.  2010;55(11):1121-1126.
  31. de Jongh S, Ose L, Szamosi T, et al.  Efficacy and safety of statin therapy in children with familial hypercholesterolemia: a randomized, double-blind, placebo-controlled trial with simvastatin. Circulation. 2002;106(17):2231-2237.
  32. de Jongh S, Lilien MR, op’t Roodt J, Stroes ES, Bakker HD, Kastelein JJ. Early statin therapy restores endothelial function in children with familial hypercholesterolemia. J Am Coll Cardiol 2002;40(12):2117-2121.
  33. Rosenson RS.  Rosuvastatin: a new inhibitor of HMG-CoA reductase for the treatment of dyslipidemia.  Expert Rev Cardiovasc Ther.  2003;1(4):495-505.
  34. Jones PH, Davidson MH, Stein EA, et al.  Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* trial).  Am J Cardiol.  2003;92(2):152-160.
  35. Brown AS, Bakker-Arkema RG, Yellen L, et al.  Treating patients with documented atherosclerosis to National Cholesterol Education Program- recommended low-density-lipoprotein cholesterol goals with atorvastatin, fluvastatin, lovastatin, and simvastatin.  J Am Coll Cariol.   1998;32(3):665-672.
  36. Kuster DM, Avis HJ, de Groot E, et al.  Ten-year follow-up after initiation of statin therapy in children with familial hypercholesterolemia.  JAMA.  2014;312(10):1055-1057.
  37. Braamskamp MJ, Stefanutti C, Langslet G, Drogari E, Wiegman A, Hounslow N, Kastelein JJ; PASCAL Study Group. Efficacy and Safety of Pitavastatin in Children and Adolescents at High Future Cardiovascular Risk. J Pediatr. 2015 Aug;167(2):338-43.e5.doi: 10.1016/j.jpeds.2015.05.006. Epub 2015 Jun 6. PMID: 26059337.
  38. Simvastatin: pediatric drug information.  Lexicomp Online, Pediatric and Neonatal Lexi-Drugs Online, Hudson, Ohio: Lexi-Comp Inc.; 2015.
  39. Colesevelam product monograph.  Daiichi Sankyo, Inc.  Parsippany, NJ. 2009.
  40. Stein EA, Marais D, Szamosi T, et al.  Colesevelam hydrochloride: efficacy and safety in pediatric subjects with heterozygous familial hypercholesterolemia.  J Pediatr.  2010; 156(2):231-236.e1-3.
  41. Gold Standard, Inc.  Colestipol. Clinical Pharmacology [database online].  Available at: http://www.clinicalpharmacology.com. Accessed 1/4/2008.
  42. Colestipol. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
  43. McCrindle BW, Helden E, Cullen-Dean G, Conner WT. A randomized crossover trial of combination pharmacologic therapy in children with familial hyperlipidemia. Pediatr Res. 2002:51(6):715-21.
  44. Tonstad S, Sivertsen M, Aksnes L, Ose L.  Low dose colestipol in adolescents with familial hypercholesterolaemia.  Arch Dis Child.  1996;74(2):157-160.
  45. Taketomo CK, Hodding JH, Kraus DM, eds.  Lexi-Comp’s Pediatric Dosing Handbook.  15th edition.  Hudson, OH: Lexi-Comp; 2008.
  46. Gold Standard, Inc.  Cholestyramine.  Clinical Pharmacology [database online].  Available at: http://www.clinicalpharmacology.com.  Accessed 1/4/2008.
  47. Cholesytramine. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
  48. McCrindle BW, O’Neill MB, Cullen-Dean G, Helden E.  Acceptability and compliance with two forms of cholestyramine in the treatment of hypercholesterolemia in children: A randomized, crossover trial.  J Pediatr. 1997;130(2):266-273.
  49. Tonstad S, Knudtzon J, Sivertsen M, Refsum H, Ose L.  Efficacy and safety of cholestyramine therapy in peripubertal and prepubertal children with familial hypercholesterolemia.  J Pediatr.  1996;129(1):42-49.
  50. Fenofibrate. In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
  51. Gemfibrozil.  In: DRUGDEX® System [intranet database]. Version 5.1. Greenwood Village, Colo: Thomson Healthcare. 2007.
  52. Wheeler KA, West RJ, Lloyd JK, Barley J. Double blind trial of bezafibrate in familial hypercholesterolaemia. Arch Dis Child. 1985;60(1):34-37.

 

  1. Fiorentino R, Chiarelli F. Treatment of Dyslipidaemia in Children. Biomedicines. 2021 Aug 24;9(9):1078. doi: 10.3390/biomedicines9091078. PMID: 34572264; PMCID: PMC8470054.
  2. Colletti RB, Roff NK, Neufeld EJ, Baker AL, Newburger JW, McAuliffe TL.  Niacin treatment of hypercholesterolemia in children.  Pediatrics.  1993;92(1):78-82.
  3. Ezetimibe product monograph. Merck & CO., Inc. Whitehouse Station, NJ. 2012.
  4. Yeste D, Chacon P, Clemente M, et al.  Ezetimibe as monotherapy in the treatment of hypercholesterolemia in children and adolescents.  J Pediatr Endocrinol Metab.  2009;22:487-492.
  5. Clauss S, Wai KM, Kavey REW, et al.  Ezetimibe treatment of pediatric patients with hypercholesterolemia.  J Pediatr.  2009;154:869-872.
  6. van der Graaf A, Cuffie-Jackson C, Vissers MN, et al.  Efficacy and safety of co-administration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia.  J Am Coll Cardiol.  2008;52(17):1421-1429.
  7. Hammersley D., Signy M. Ezetimibe: An update on its clinical usefulness in specific patient groups. Ther. Adv. Chronic Dis. 2017;8:4–11. doi: 10.1177/2040622316672544.
  8. de Ferranti SD, Milliren CE, Denhoff ER, et al.  Using high-dose omega-3 fatty acid supplements to lower triglyceride levels in 10- to 19-year-olds.  Clin Pediatr (Phila).  2014;53(5):428-438.
  9. Chahal N, Manlhoit C, Wong H, McCrindle BW.Effectiveness of omega-3 polysaturated fatty acids (fish oil) supplementation for treating hypertriglyceridemia in children and adolescents.  Clin Pediatr (Phila).  2014;53(7):645-657.
  10. Khorshidi, M., Hazaveh, Z.S., Alimohammadi-kamalabadi, M. et al.Effect of omega-3 supplementation on lipid profile in children and adolescents: a systematic review and meta-analysis of randomized clinical trials. Nutr J 22, 9 (2023). https://doi.org/10.1186/s12937-022-00826-5
  11. McGowan MP, Hosseini Dehkordi SH, Moriarty PM, Duell PB. Diagnosis and Treatment of Heterozygous Familial Hypercholesterolemia. J Am Heart Assoc. 2019 Dec 17;8(24):e013225. doi: 10.1161/JAHA.119.013225. Epub 2019 Dec 16. PMID: 31838973; PMCID: PMC6951065.
  12. Diagnosis and Treatment of Heterozygous Familial Hypercholesterolemia Mary P. McGowanMD mary.p.mcgowan@hitchcock.org , Seyed Hamed Hosseini Dehkordi MD , Patrick M. Moriarty MD , and P. Barton Duell MD
  13. Defesche JC, Gidding SS, Harada-Shiba M, Hegele RA, Santos RD, Wierzbicki AS. Familial hypercholesterolaemia. Nat Rev Dis Primers. 2017 Dec 7;3:17093. doi: 10.1038/nrdp.2017.93. PMID: 29219151.
  14. Alenizi MM, Almushir S, Sulimanaa I. Surgical Management and Outcomes of Homozygous Familial Hypercholesterolemia in Two Cousins: A Rare Case Report. Cureus. 2020 Nov 25;12(11):e11692. doi: 10.7759/cureus.11692. PMID: 33391926; PMCID: PMC7769830.
  15. German, C. Homozygous Familial Hypercholesteremia: Diagnosis and Emerging Therapies. 2022 Feb 18. American College of Cardiology.
  16. Avis HJ, Vissers MN, Stein EA, Wijburg FA, Trip MD, Kastelein JJ, Hutten BA. A systematic review and meta-analysis of statin therapy in children with familial hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2007 Aug;27(8):1803-10. doi: 10.1161/ATVBAHA.107.145151. Epub 2007 Jun 14. PMID: 17569881.
  17. Mirzaee S, Cameron JD. Familial hypercholesterolemia and cardiovascular risk stratification. J Clin Lipidol. 2018 Sep-Oct;12(5):1328-1329. doi: 10.1016/j.jacl.2018.07.002. Epub 2018 Jul 10. PMID: 30061010.
  18. Luirink IK, Wiegman A, Kusters DM, Hof MH, Groothoff JW, de Groot E, Kastelein JJP, Hutten BA. 20-Year Follow-up of Statins in Children with Familial Hypercholesterolemia. N Engl J Med. 2019 Oct 17;381(16):1547-1556. doi: 10.1056/NEJMoa1816454. PMID: 31618540.
  19. Mainieri F, Tagi VM, Chiarelli F. Recent Advances on Familial Hypercholesterolemia in Children and Adolescents. Biomedicines. 2022; 10(5):1043. https://doi.org/10.3390/biomedicines10051043
  20. van der Graaf A, Cuffie-Jackson C, Vissers MN, Trip MD, Gagné C, Shi G, Veltri E, Avis HJ, Kastelein JJ. Efficacy and safety of coadministration of ezetimibe and simvastatin in adolescents with heterozygous familial hypercholesterolemia. J Am Coll Cardiol. 2008 Oct 21;52(17):1421-9. doi: 10.1016/j.jacc.2008.09.002. PMID: 18940534
  21. Kusters DM, Caceres M, Coll M, Cuffie C, Gagné C, Jacobson MS, Kwiterovich PO, Lee R, Lowe RS, Massaad R, McCrindle BW, Musliner TA, Triscari J, Kastelein JJ. Efficacy and safety of ezetimibe monotherapy in children with heterozygous familial or nonfamilial hypercholesterolemia. J Pediatr. 2015 Jun;166(6):1377-84.e1-3. doi: 10.1016/j.jpeds.2015.02.043. Epub 2015 Apr 1. PMID: 25841542.
  22. Hajar R. PCSK 9 Inhibitors: A Short History and a New Era of Lipid-lowering Therapy. Heart Views. 2019 Apr-Jun;20(2):74-75. doi: 10.4103/HEARTVIEWS.HEARTVIEWS_59_19. PMID: 31462965; PMCID: PMC6686613.
  23. Zhao Z, Du S, Shen S, Luo P, Ding S, Wang G, Wang L. Comparative efficacy and safety of lipid-lowering agents in patients with hypercholesterolemia: A frequentist network meta-analysis. Medicine (Baltimore). 2019 Feb;98(6):e14400. doi: 10.1097/MD.0000000000014400. PMID: 30732185; PMCID: PMC6380691.
  24. Repatha product monograph. Amgen. Thousand Oaks, California. 2017.
  25. Santos RD, Ruzza A, Hovingh GK, Wiegman A, Mach F, Kurtz CE, Hamer A, Bridges I, Bartuli A, Bergeron J, Szamosi T, Santra S, Stefanutti C, Descamps OS, Greber-Platzer S, Luirink I, Kastelein JJP, Gaudet D; HAUSER-RCT Investigators. Evolocumab in Pediatric Heterozygous Familial Hypercholesterolemia. N Engl J Med. 2020 Oct 1;383(14):1317-1327. doi: 10.1056/NEJMoa2019910. Epub 2020 Aug 29. PMID: 32865373.
  26. Santos RD, Ruzza A, Hovingh GK, Stefanutti C, Mach F, Descamps OS, Bergeron J, Wang B, Bartuli A, Buonuomo PS, Greber-Platzer S, Luirink I, Bhatia AK, Raal FJ, Kastelein JJP, Wiegman A, Gaudet D. Paediatric patients with heterozygous familial hypercholesterolaemia treated with evolocumab for 80 weeks (HAUSER-OLE): a single-arm, multicentre, open-label extension of HAUSER-RCT. Lancet Diabetes Endocrinol. 2022 Oct;10(10):732-740. doi: 10.1016/S2213-8587(22)00221-2. Epub 2022 Sep 5. PMID: 36075246.
  27. Praluent product monograph. Regeneron. Tarrytown, NY. 2021.
  28. Bruckert E, Caprio S, Wiegman A, Charng MJ, Zárate-Morales CA, Baccara-Dinet MT, Manvelian G, Ourliac A, Scemama M, Daniels SR. Efficacy and Safety of Alirocumab in Children and Adolescents With Homozygous Familial Hypercholesterolemia: Phase 3, Multinational Open-Label Study. Arterioscler Thromb Vasc Biol. 2022 Dec;42(12):1447-1457. doi: 10.1161/ATVBAHA.122.317793. Epub 2022 Nov 3. PMID: 36325897; PMCID: PMC9750107.
  29. Daniels S, Caprio S, Chaudhari U, Manvelian G, Baccara-Dinet MT, Brunet A, Scemama M, Loizeau V, Bruckert E. PCSK9 inhibition with alirocumab in pediatric patients with heterozygous familial hypercholesterolemia: The ODYSSEY KIDS study. J Clin Lipidol. 2020 May-Jun;14(3):322-330.e5. doi: 10.1016/j.jacl.2020.03.001. Epub 2020 Mar 28. Erratum in: J Clin Lipidol. 2020 Sep - Oct;14(5):741. PMID: 32331936.
  30. Ben-Omran T, Masana L, Kolovou G, Ariceta G, Nóvoa FJ, Lund AM, Bogsrud MP, Araujo M, Hussein O, Ibarretxe D, Sanchez-Hernández RM, Santos RD. Real-World Outcomes with Lomitapide Use in Paediatric Patients with Homozygous Familial Hypercholesterolaemia. Adv Ther. 2019 Jul;36(7):1786-1811. doi: 10.1007/s12325-019-00985-8. Epub 2019 May 17. PMID: 31102204; PMCID: PMC6824397.
  31. Leqvio product monograph. Novartis. East Hanover, NJ. 2021.
  32. Reijman MD, Schweizer A, Peterson ALH, Bruckert E, Stratz C, Defesche JC, Hegele RA, Wiegman A. Rationale and design of two trials assessing the efficacy, safety, and tolerability of inclisiran in adolescents with homozygous and heterozygous familial hypercholesterolaemia. Eur J Prev Cardiol. 2022 Jul 20;29(9):1361-1368. doi: 10.1093/eurjpc/zwac025. PMID: 35175352.
  33. Kosmas CE, Bousvarou MD, Sourlas A, Papakonstantinou EJ, Genao EP, Uceta RE, Guzman E.Angiopoietin-like protein 3 (ANGPTL3) inhibitors in the management of refractory hypercholesterolemia.  Clin Pharmacol. 2022;14:49-59. doi: 10.2147/CPAA.S345072.  Epub 2022 Jul 16. PMID: 358l73366.
  34. Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, Ali S, Banerjee P, Chan K-C, Gipe DA, Khilla N, Pordy R, Weinreich DM, Yancopoulos GD, Zhang Y, Guadet D, ELIPSE HoFH Investigators. Evinacumab for Homozygous Familial Hypercholesterolemia.N Engl J Med. 2020 Aug 20;383(8):711-720. doi: 10.1056/NEJMoa2004215. PMID: 32813947.
  35. Evkeeza (Evinacumab-dgnb) product monograph. Regeneron. Tarrytown, NY. 2023.
  36. Rayan RA, Sharma S. Lomitapide. [Updated 2022 Jul 18]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK560849/
  37. Goulooze SC, Cohen AF, Rissmann R. Lomitapide. Br J Clin Pharmacol. 2015 Aug;80(2):179-81. doi: 10.1111/bcp.12612. Epub 2015 Jul 2. PMID: 25702706; PMCID: PMC4541964.
  38. Ray KK, Bays HE, Catapano AL, Lalwani ND, Bloedon LT, Sterling LR, Robinson PL, Ballantyne CM; CLEAR Harmony Trial. Safety and Efficacy of Bempedoic Acid to Reduce LDL Cholesterol. N Engl J Med. 2019 Mar 14;380(11):1022-1032. doi: 10.1056/NEJMoa1803917. PMID: 30865796.
  39. Ballantyne CM, Bays H, Catapano AL, Goldberg A, Ray KK, Saseen JJ. Role of Bempedoic Acid in Clinical Practice. Cardiovasc Drugs Ther. 2021 Aug;35(4):853-864. doi: 10.1007/s10557-021-07147-5. Epub 2021 Apr 5. Erratum in: Cardiovasc Drugs Ther. 2021 Apr 19;: PMID: 33818688; PMCID: PMC8266788.
  40. Bempedoic acid product monograph.Esperion. Ann Arbor, MI. 2022.
  41. Sunil B, Foster C, Wilson DP, Ashraf AP. Novel therapeutic targets and agents for pediatric dyslipidemia. Ther Adv Endocrinol Metab. 2021 Nov 24;12:20420188211058323. doi: 10.1177/20420188211058323. PMID: 34868544; PMCID: PMC8637781.
  42. Witztum JL, Gaudet D, Freedman SD, Alexander VJ, Digenio A, Williams KR, Yang Q, Hughes SG, Geary RS, Arca M, Stroes ESG, Bergeron J, Soran H, Civeira F, Hemphill L, Tsimikas S, Blom DJ, O'Dea L, Bruckert E. Volanesorsen and Triglyceride Levels in Familial Chylomicronemia Syndrome. N Engl J Med. 2019 Aug 8;381(6):531-542. doi: 10.1056/NEJMoa1715944. PMID: 31390500.
  43. Kolovou G, Kolovou V, Katsiki N. Volanesorsen: A New Era in the Treatment of Severe Hypertriglyceridemia. J Clin Med. 2022 Feb 13;11(4):982. doi: 10.3390/jcm11040982. PMID: 35207255; PMCID: PMC8880470.
  44. Esan O, Wierzbicki AS. Volanesorsen in the Treatment of Familial Chylomicronemia Syndrome or Hypertriglyceridaemia: Design, Development and Place in Therapy. Drug Des Devel Ther. 2020 Jul 6;14:2623-2636. doi: 10.2147/DDDT.S224771. PMID: 32753844; PMCID: PMC7351689.
  45. Gelrud A, Digenio A, Alexander V, et al. Treatment with Volanesorsen (VLN) Reduced Triglycerides and Pancreatitis in Patients with FCS and sHTG vs Placebo: results of the APPROACH and COMPASS J Clin Lipidol. 2018;12(2):537. doi: 10.1016/j.jacl.2018.03.032
  46. Kohan AB. Apolipoprotein C-III: a potent modulator of hypertriglyceridemia and cardiovascular disease. Curr Opin Endocrinol Diabetes Obes. 2015 Apr;22(2):119-25. doi: 10.1097/MED.0000000000000136. PMID: 25692924; PMCID: PMC4524519.

Ambiguous Genitalia in the Newborn

ABSTRACT 

 

Ambiguous genitalia in a newborn are the clinical sign of atypical sexual development of the external genitalia in utero. This condition is rare and can result from various underlying factors, including certain disorders with potentially severe consequences, such as cortisol deficiency due to congenital adrenal hyperplasia. Therefore, it is crucial to promptly determine etiology when ambiguity is observed. The formation of typical male or female external genitalia is a complex process involving a cascade of genetic and physiological events that begin with sex determination and progress through the differentiation of internal and external reproductive structures. When this process is disrupted and does not occur in the typical manner, it is referred to as a difference or disorder of sex development (DSD). Not all DSD cases present with ambiguous genitalia at birth; for example, complete androgen insensitivity syndrome does not, but all cases of ambiguous genitalia are the result of a DSD. This chapter focuses on genital ambiguity associated with DSD in newborns who have either a 46,XY or 46,XX chromosomal sex. However, DSD with genital ambiguity may also be observed in newborns with other combinations of sex chromosomes, such as 45,X/46,XY. The chapter offers a comprehensive overview of the evaluation and management of newborns with ambiguous genitalia. It emphasizes the importance of a structured medical assessment of the external genitalia to diagnose and determine the underlying cause of genital ambiguity. It includes tables for differential diagnosis and step-by-step workup algorithms to guide medical professionals in their evaluation. It also includes additional information on structured physical assessments of external genitalia and tables with normative values for hormonal measurements, which are recommended in the diagnostic process. As the etiology of genital ambiguity in newborns is diverse and can have significant implications for management, the authors stress that obtaining an accurate diagnosis through a professional medical workup is crucial. The chapter highlights the recommendation for newborns with ambiguous genitalia (DSD) to be cared for by highly specialized, interdisciplinary DSD teams. These teams are equipped with medical and psychosocial expertise, and specialized psychologists are available to support parents and caregivers. The chapter recognizes that having a child with ambiguous genitalia can be very stressful for parents. It underscores the importance of early education, access to expert care through DSD network teams, psychological support, shared decision-making, and promoting acceptance and inclusivity. By providing comprehensive support and guidance, families can better navigate the challenges and uncertainties they may encounter when caring for a newborn with ambiguous genitalia.

 

INTRODUCTION

 

Typical male and female development commences with the presence of the typical 46,XY or 46,XX chromosomes, which play a pivotal role in determining the indifferent gonads. Around the 6th week of gestation, these gonads follow pathways to develop into male-typical or female-typical gonads (Figure 1). This gonadal determination is intricately regulated by a complex interplay of multiple genes, which guide the male gonads to become testes and the female gonads to become ovaries.

 

Subsequently, the gonads initiate the production of various hormones, including anti-Müllerian hormone (AMH) and testosterone. These hormones are responsible for driving the differentiation of the embryologic Wolffian and Müllerian structures into sex-typical internal reproductive organs (Figure 1).

 

Figure 1. Typical male and female sex determination and differentiation occur during fetal life. The presence of the Y chromosome in the karyotype and the SRY gene (sex-determining region Y gene) are the primary determinants of male sex development. The SRY gene initiates a cascade that leads to the development of testes from bipotential gonads. Testosterone and Anti Mullerian Hormone (AMH) are secreted by testicular cells, which masculinize the Wolffian (mesonephric) duct and cause the regression of the Mullerian (paramesonephric) duct, respectively. Dihydrotestosterone (DHT), produced by the 5-alpha reduction of testosterone, plays a major role in male external genital development and prostate formation. The absence of the SRY gene on the Y chromosome is the primary factor for the induction of the female sex, and the development of the uterus, fallopian tubes, cervix and upper vagina from the Mullerian duct.

 

The development of external genitalia begins with a neutral anlage, including a genital tubercule, genital folds, and a urogenital sinus. These structures are subsequently differentiated into typical male external genitalia under the influence of androgens, particularly dihydrotestosterone (DHT). Conversely, for the development of typical female external genitalia, the absence of androgens is crucial to prevent virilization.

 

Therefore, ambiguous genitalia at birth can arise from either an excessive or insufficient androgen effect on the neutral external genital anlage in males or females, respectively (Figure 2).

Figure 2. External genital differentiation. The development of external genitalia begins with a bipotential anlage which includes genital tubercle, urethral fold, urethral groove, and genital swellings. In a typical 46, XY, androgen secretion from the testes triggers the fusion of urethral folds, allowing the enclosure of the urethral tube. This, together with the cells from the genital swelling, forms the shaft of the penis. Genital swellings fuse in the midline to allow formation of the scrotum and genital Tubercle expands to give rise to the glans penis. Female external genital development is ensured by the absence of testosterone, it is independent of ovarian endocrine activity. Urethral folds and genital swellings remain separate to form the labia minora and majora. Genital tubercle forms the clitoris.

 

CHARACTERISTICS OF TYPICAL MALE AND FEMALE EXTERNAL GENITALIA

 

A full-term male infant is typically expected to have bilateral testicles that are descended, complete formation of scrotal folds with midline fusion, and a typical-sized penis, which includes well-formed corporal bodies and a urethral meatus located at the tip. The average penile length for a full-term newborn is approximately 3.5 ± 0.4 cm, but this measurement can vary with gestational age (Table 1) (1, 2). If an infant presents with bilateral cryptorchidism, a bifid scrotum, hypospadias, or isolated penoscrotal hypospadias, further investigations for a DSD are recommended. However, it’s important to note that isolated micropenis, as long as both testes are descended and normal in size, is not typically considered a manifestation of ambiguous genitalia. Similarly, in males, distal hypospadias with no other atypical genital features usually does not indicate a DSD (3).

 

Table 1. Stretched Penile Length in Preterm and Term Newborns According to Gestational Age (2)

Gestational age (week)

Number of cases

Median (cm)

Mean (cm)

SD

Min (cm)

Max (cm)

26

30

1.9

1.9

0.32

1.1

2.5

27

31

2.1

2.0

0.28

1.1

2.6

28

32

2.0

1.9

0.31

1.3

2.9

29

29

2.3

2.3

0.39

1.3

2.9

30

31

2.4

2.4

0.32

1.6

3.0

31

33

2.4

2.4

0.38

1.8

3.3

32

32

2.9

2.6

0.44

2.0

3.2

33

28

2.9

2.8

0.45

1.9

3.4

34

30

2.9

2.8

0.43

2.0

3.6

35

29

3.1

2.9

0.61

2.0

4.1

36

31

3.1

3.0

0.50

2.2

4.0

37

29

3.1

3.0

0.47

2.2

4.1

38

44

3.1

3.1

0.54

2.1

4.5

39

82

3.2

3.2

0.55

2.0

4.2

40

65

3.5

3.4

0.53

2.2

4.4

41

29

3.6

3.5

0.47

2.6

4.3

 

A full-term female infant typically presents with bilateral separated labial folds, no palpable gonads, and distinct urethral and vaginal openings (Figure 2). The average clitoral length and width for a full-term infant girl are approximately 6.1 mm and 4.2 mm, respectively (1, 4). In cases where clitoromegaly (clitoral size ˃10 mm), labial fusion or palpable gonads are observed in otherwise typical female genitalia, further investigation for a DSD is warranted. It’s important to note that perceived clitoromegaly is not typically associated with an underlying DSD in the event that the newborn girl was born prematurely (1). Please be aware that perceived clitoromegaly or pseudo-clitoromegaly is often seen in preterm newborns, where the clitoral size falls within the normal range, but the labia majora cannot completely cover labia minora, creating a misleading impression of clitoromegaly. Genital edema may also be observed in sick preterm babies or cases of ovarian hyperstimulation, which can cause pseudo-clitoromegaly.

 

Table 2. Clitoral Length and Width in Preterm and Term Newborns According to Gestational Age (4)

Gestational age (week)

Number of cases

Measure

Mean (mm)

+1 SD (mm)

+2 SD (mm)

+3 SD (mm)

28-30

30

Width

3.28

3.87

4.46

5.05

Length

5.03

5.91

6.79

7.67

30-32

57

Width

3.32

3.68

4.04

4.4

Length

5.03

5.52

6.01

6.5

32-34

74

Width

3.41

3.77

4.13

4.49

Length

5.14

5.65

6.16

6.67

34-36

104

Width

3.69

4.17

4.65

5.13

Length

5.53

6.12

6.71

7.3

36-38

128

Width

4.08

4.5

4.92

5.34

Length

5.83

6.29

6.75

7.21

>38

187

Width

4.21

4.64

5.07

5.5

Length

6.11

6.49

6.87

7.25

 

GENERAL THOUGHTS ON AMBIGUOUS GENITALIA AT BIRTH

 

The reported incidence of ambiguous genitalia in newborns is approximately one in 4,500. It is essential that a newborn with ambiguous genitalia and their parents are promptly referred to a specialized center for evaluation. This evaluation should be carried out by a multi-disciplinary team specializing in DSD in accordance with international recommendations on diagnostic and therapeutic strategies. This team should provide holistic care and treatment guidance, collaborating closely with relevant subspecialists and peer support groups (5). Key subspecialties involved in DSD teams typically include psychology, pediatric endocrinology, urology, neonatology, gynecology, andrology, nursing, social work, genetics, and medical ethics. It's worth noting that certain aspects of DSD management remain contentious or uncertain, resulting in recent changes in clinical practice that are ongoing. Notably, genital surgery has been banned in several countries, and it is increasingly avoided in young children to protect their rights to an open future and the integrity of their bodies.

 

Consequently, we can anticipate a growing number of children growing up with atypical-looking genitalia, requiring a careful assessment of the psychological impact (6). Providing psychological support to affected families as a standard component of care from the outset is, therefore, of utmost importance. Issues related to gender that may arise in the context of ambiguous genitalia should be openly and thoughtfully discussed.

 

A comprehensive evaluation of ambiguous genitalia in a newborn is essential for understanding the underlying cause and potential consequences. On one hand, this evaluation can lead to the detection of concomitant, treatable disorders, such as adrenal insufficiency that requires cortisol replacement. Also, it is important to note that in up to 30% of children with a Difference/Disorder of Sex Development (DSD), other organ system disorders may be identified, necessitating medical care and qualifying as syndromic DSD (7, 8). On the other hand, identifying the underlying cause of ambiguous genitalia forms the basis for making an informed decision regarding sex registration at birth, if necessary and desired by the parents. Gender assignment/sex registration at birth should take into account long-term satisfaction with the sex of rearing, sexual function, and fertility potential (1, 9, 10). Ideally, it should result from a shared decision-making process involving the child's parents and the DSD network team. This decision should be based on the individual's phenotype/genotype, existing knowledge from literature, databases, and patient representative experience (11-13). This process may require time, as parents need to grasp the complexity of their child's unique biology in order to participate in this decision. As a first step, the overall health of the child should be assessed, and parents should be educated about typical sexual development before explanations regarding the underlying cause and its implications for their child's ambiguous genitalia are provided. Even if the exact details of the underlying cause remain unresolved, it is essential to communicate what is known and what remains uncertain to the parents and make plans for the family's future in the best interests of the child.

 

Several websites are available for patients and their families to exchange information, share coping strategies, and discuss decision-making processes that can enhance outcomes for newborns with ambiguous genitalia. Such resources can be found on websites supported by organizations like the Accord Alliance (www.accordalliance.org), DSD families (www.dsdfamilies.org), AIS DSD Support Group (www.aisdsd.org), and the CARES Foundation (www.caresfoundation.org). However, the most current information will always be available from the responsible DSD team, which will also be aware of country-specific support services.

 

GENETICS OF SEX DETERMINATION AND DIFFERENTIATION 

 

Sex development is a sequential process that involves the coordinated action of numerous genes and pathways. This process culminates in the development of functional gonads, differentiated internal sex organs, external genitalia, and typical secondary sexual characteristics that manifest after puberty. Prenatal sex development can be divided into two distinct processes: sex determination and sex differentiation (Figure 3). Sex determination is the initial step that occurs shortly after conception, approximately at 6-7 weeks gestation. During this phase, the undifferentiated gonads evolve into either testes or ovaries. By contrast, sex differentiation occurs subsequently and involves hormones produced after the formation of the sex-specific gonads. These hormones play a critical role in shaping the further development of the dependent embryonic structures, leading to the formation of male and female internal and external phenotypes.

 

Figure 3. The genes and transcription factors required for gonadal development. The formation of the bipotential gonad requires at least seven genes including NR5A1 (Nuclear Receptor Subfamily 5 Group A Member 1), WT1 (WilmsTumor1 Transcription Factor), LHX9 (LIM Homeobox 9), EMX2 (Empty Spiracles Homeobox 2), CBX2 (Chromobox 2), PBX1 (Pre-B-Cell Leukemia Transcription Factor 1) and GATA4 (GATA Binding Protein 4). Adapted from (14).

 

The Male Specific Pathway

 

In the male specific pathway, the expression of the SRY gene in pre-Sertoli cells serves as a pivotal switch that directs the fate of the gonads toward testicular development. SRY, in conjunction with NR5A1, activates the SOX9 signaling pathway, initiating testis development (Figure 3). Other important genes for the early testis development are NR0B1(Nuclear Receptor subfamily 0 group B member 1), AMH (Anti-Mullerian Hormone) and GATA4. AMH secreted by Sertoli cells, plays a crucial role in promoting the regression of Müllerian structures and supporting the development of Leydig cells in the testes. Subsequently, androgens produced by the testes facilitate the development of male internal organs (epididymis, vas deferens, seminal vesicle) and external genitalia (penis, scrotum). The production of sex steroids relies on an intact steroidogenic pathway, with all steroid hormones originating from cholesterol. The initial steps of sex steroid biosynthesis overlap with the synthesis of mineralo-corticosteroids and gluco-corticosteroids (Figure 4). It is important to note that certain steroid disorders, often referred to as congenital adrenal hyperplasia (CAH), can affect both adrenal and gonadal steroidogenesis or result in androgen excess or deficiency that may impact fetal genital development. INSL3, produced in Leydig cells, plays a role in guiding the descent of the testes from the abdomen to the scrotal folds.

 

The process of masculinization of initially sex-neutral, undifferentiated external genitalia begins around 8-9 weeks of gestation when the potent androgen DHT (dihydrotestosterone) is produced by the testes. DHT is crucial for the fusion of the urethral and labioscrotal folds, elongation of the genital tubercle, regression of the urogenital sinus, and the development of the prostate (15). The enzyme 5α-reductase is responsible for converting testosterone produced by fetal Leydig cells into DHT. Complete masculinization of the external genitalia through DHT is typically achieved by week 14 of gestation (16).

 

Figure 4. Steroidogenesis of the adrenals and gonads. Adrenal and gonadal steroidogenesis starting from cholesterol and showing key steroids and intermediates of the classical and alternative pathways is illustrated. Cortisol and androgen biosynthesis are under control of hypothalamic-pituitary-adrenal (HPA) axis in a negative feedback loop. Androgen synthesis occurs via classical or aberrant and alternative pathways (i.e., backdoor pathway and 11oxC19 androgen pathway). Aberrant or alternative pathways are particularly active whenever cortisol with or without aldosterone biosynthesis is impaired and androgenic precursors accumulate proximal to enzymatic block. Abbreviations: StAR; Steroidogenic Acute Regulatory Protein defect, CYP11A1; P450 side-chain cleavage deficiency, CYP21A2; 21α-hydroxylase, HSD3B2; 3β-hydroxy steroid dehydrogenase 2, CYP11B1; 11β-hydroxylase, CYP11B2; Aldosterone synthase, DOC; 11-deoxycorticosterone, DHEA; Dehydroepiandrosterone, A4; Androstenedione, T; Testosterone, CYP17A1; 17α-hydroxylase/17,20-lyase, b5; CYP19A1; Cytochrome P450 aromatase, Cytochrome b5, POR; P450 oxidoreductase, 17β-HSD; 17β-hydroxysteroid oxidoreductase or 17-ketosteroid reductase; SRDA1/2; Steroid 5α -reductase 1/2, 11oxC19; 11-oxygenated 19-carbon (11oxC19) steroids; Fdx; Ferredoxin, FdR; Ferredoxin reductase, 11βOHA; 11β-hydroxyandrostenedione, 11βOHT; 11β-hydroxytestosterone, Pdione; 5α-Pregnane- 17α-ol-3,20-dione, Pdiol; 5α-Pregnane- 3α,17α-diol 20-one, AKR1C2,4; Aldo-Keto Reductase Family 1 Member C2,4, DHT; Dihydrotestosterone, HSD17B5,3,6= 17β-hydroxysteroid dehydrogenase 5,3,6. Adapted from (17).

 

The Female Specific Pathway

 

By contrast, the female-specific pathway is initiated in the absence of SRY and requires the suppression of SOX9 (Figure 3). Activation of Rspo1/Wnt4/b-catenin and Foxl2 (Forkhead Box L2) signaling pathways leads to the formation of typical ovaries. The absence of AMH allows the Müllerian structures to proliferate, resulting in the formation of the typical fallopian tube, uterus, cervix and the upper part of the vagina. In the absence of both testosterone secretion and 5α-reductase activity, the sex-neutral external genitalia develop along the female pathway. Specifically, without DHT, the labioscrotal and urethral folds give rise to the labia majora and minora, respectively. The genital tubercle develops into a clitoris, and the urogenital sinus contributes to the formation of the urethral opening and anterior portion of the vagina (16).

 

Any disruption in the intricate genetic and hormonal processes involved can lead to atypical sex development and result in atypical external genitalia at birth. Figure 3 provides a summary of these processes and highlights some key genes and hormones essential for typical development.

 

DIFFERENCES/DISORDERS OF SEX DEVELOPMENT (DSD) 

 

The discordance between genetic, gonadal, or anatomic sex is commonly referred to as DSD. In addition to candidate gene testing, the new era of molecular diagnostic tools, including whole exome/genome sequencing has uncovered numerous novel molecular etiologies in recent years. Accurate molecular diagnosis aids in managing affected individuals and provides families with information concerning prognosis and the risk of recurrence.  This information is often sourced from literature and comprehensive data registries, such as the I-DSD (18-24). For clinical purposes, DSD (including ambiguous genitalia in newborns) is typically classified based on the affected individual's karyotype. The consensus statement addressing the approach and care of DSD (originally published in 2006 and updated in 2016) suggests the following broad classification for DSD: (A) Sex Chromosome DSD, (B) 46,XY DSD, and (C) 46,XX DSD (Table 3) (1, 20).

 

Table 3. DSD Classification According to the Chicago Consensus.

Sex Chromosome DSD

46,XY DSD

46,XX DSD

A. 45,X0 (Turner syndrome and variants)

A. Disorders of gonadal (testicular) development

Complete gonadal dysgenesis (Swyer syndrome) or partial gonadal dysgenesis (e.g., SRY,SOX9, NR5A1, WT1, DHHgene mutations etc)

Testicular regression (e.g., DHX37 gene mutation)

Ovo-Testicular DSD

A. Disorder of gonadal (ovarian) development

Ovo-Testicular DSD

Testicular DSD (SRY (+))

Gonadal dysgenesis

B. 47,XXY (Klinefelter syndrome and variants)

B. Disorders of testicular hormone production or action

Impaired testosterone production

LH receptor mutations

CAH (3βHSD2 deficiency, 17OHD, POR, StAR, CYP11A1 deficiency)

HSD17B3 deficiency

 

Impaired testosterone action

5α-reductase deficiency

Androgen insensitivity syndrome (Complete/partial)

 

Impaired AMH production or action

Persistent Mullerian Duct Syndrome (AMH and AMHR2gene mutations)

B. Androgen excess

Fetal

CAH (21-hydroxylase, 11β-hydroxylase, 3βHSD2 deficiency)

Glucocorticoid resistance

 

Feto-placental

Aromatase deficiency

POR deficiency

 

Maternal

Luteoma, hilar cell tumors, arrhenoblastoma, lipoid cell tumors, Krukenberg tumors, androgen producing adrenal tumors,

External androgen exposure

C. 45,X0/46,XY (Mixed gonadal dysgenesis, ovotesticular DSD)

C. Other

Syndromic

Smith-Lemli-Opitz, Cloacal anomaly, Aarskog, Robinow, Meckel, Joubert, Hand-Foot-Genital, popliteal pterygium, CHARGE, VATER/VACTERL, IMAGe etc.

 

Drugs

Flutamide, ketoconazole, progestagens

 

Endocrine disruptors

Phthalate, BPA, paraben

 

Cryptorchidism

INSL3, GREAT gene mutations etc.

 

Isolated hypospadias

MAMLD1, HOXA4 gene mutations etc.

C. Other

Syndromic

Cloacal anomaly etc.

 

Mullerian anomalies

Mayer-Rokitanski-Küster-Hauser, MURCS, McKusick-Kaufmnann syndrome and variants, MODY5

 

D. 46,XX/46,XY (chimeric, ovotesticular DSD)

 

 

Abbreviations: StAR; Steroidogenic Acute Regulatory Protein defect, CYP11A1; P450 side-chain cleavage deficiency, 3βHSD2; 3β-hydroxy steroid dehydrogenase 2, CYP17A1; 17α-hydroxylase/17,20-lyase deficiency, 17OHD; 17α-hydroxylase/17,20-lyase deficiency, POR; P450 oxidoreductase, HSD17B3; 17β-hydroxysteroid oxidoreductase or 17-ketosteroid reductase; SF1/NR5A1; steroidogenic factor 1.

 

The 46,XY Newborn With Ambiguous Genitalia

 

Ambiguous genitalia in a 46XY newborn can result from abnormal formation of the early fetal testes (testicular dysgenesis), reduced production of testosterone or dihydrotestosterone (5α-reductase deficiency), or the inability to respond to androgens (androgen insensitivity syndrome, or AIS) (Table 3). Depending on the extent of androgen production defect or resistance, affected newborns may exhibit a range of external genitalia phenotype, varying from those that appear typically female to those that appear typically male but with a small phallus, hypospadias, and a bifid scrotum, with or without palpable testes. Here we focus primarily on causes that lead to ambiguous genitalia. Broadly, four groups of underlying causes of 46, XY DSD with ambiguous genitalia can be identified: (a) partial gonadal dysgenesis, (b) partial testosterone biosynthetic defects, (c) partial 5α-reductase deficiency, and (d) partial androgen insensitivity syndrome or PAIS.

 

PARTIAL GONADAL DYSGENESIS

 

In cases of partial gonadal dysgenesis, it is presumed that a gene mutation leads to a partial abnormality in the development of the early urogenital ridge or gonadal anlage. This category of DSD can also involve mutations in genes such as SRY and SOX9, which are essential for the differentiation of bipotential gonads into testes (Figure 3). Partial gonadal dysgenesis is typically associated with incomplete masculinization of the external genitalia, along with varying degrees of maintenance of Wolffian ducts and inhibition of Müllerian ducts. For a summary of known genetic variations associated with gonadal dysgenesis, please refer to Table 4.

 

Leydig cell aplasia or hypoplasia represents a variation of gonadal dysgenesis, characterized by inadequate Leydig cell differentiation in the male gonad, resulting in impaired androgen production.  Mutations in the LH receptor gene (LHCGR) as well as polymorphisms have been associated with this condition (25). The phenotype associated with Leydig cell hypoplasia includes incomplete masculinization of the external genitalia, along with incomplete development and maintenance of the Wolffian ducts. 

 

 

Table 4. Monogenic Disorders of Gonadal Development/Differentiation and Associated DSD Presentations in Humans, Both 46,XY and 46,XX

Gene defect

Locus

Mode of transmission

Clinical presentation

Reference

 

 

 

DSD phenotype

Other

 

 

 

 

46, XX

46, XY

 

 

ARX

Xp22.13

XL

 

Testicular dysgenesis

Lissencephaly, epilepsy, intellectual disability temperature instability

(26)

ATRX

Xq13.3

XL

 

Testicular dysgenesis

Dysmorphic features, intellectual disability, α-thalassemia

(27)

CBX2

17q25

AR

Ovarian dysgenesis

Testicular dysgenesis

 

(28)

DHH

12q13.12

AR

 

Testicular dysgenesis

Minifascicular neuropathy

(29)

DMRT1/DMRT2                       

9p24.3

AD

Ovarian dysgenesis

Testicular dysgenesis

Hypotonia, developmental delay, impaired intellectual development

(30)

DHX37

12q24.31

AD

 

Testicular dysgenesis, testicular regression

 

(31)

EMX2

10q26.11  

 

Ovarian dysgenesis

Testicular dysgenesis

Intellectual disability, kidney agenesis

(32)

ESR2

14q23.2-q23.3

AD,?

Ovarian dysgenesis

Testicular dysgenesis

Dysmorphic features, eye abnormalities, anal atresia, rectovestibular fistula

(33)

FGFR2

10q26.13

AD

 

Testicular dysgenesis, ovotesticular

Craniosynostosis

(34)

GATA4 

8p23.1

AD

 

Testicular dysgenesis

Congenital heart defects (atrial septum defects, ventricular septum defects, tetralogy of Fallot), diaphragmatic hernia

(35)

HHAT

1q32.2

AR

 

Testicular dysgenesis

Nivelon-Nivelon-Mabille syndrome. Dwarfism, chondrodysplasia, narrow, bell-shaped thorax, micromelia, brachydactyly, microcephaly with cerebellar vermis hypoplasia, facial anomalies, hypoplastic irides and coloboma of the optic discs

(36)

LHX9

1q31.3

AD

 

Testicular dysgenesis

Limb anomalies

 

(37)

MAMLD1

Xq28

XL

 

Severe hypospadias

Myotubular myopathy

 

(38)

MAP3K1

5q11.2

AD

 

Testicular dysgenesis

 

(39)

NR0B1 (Duplications, inversion

and upstream deletion)

X21.3

XL

 

 

Testicular dysgenesis

Congenital adrenal hypoplasia, hypogonadotropic hypogonadism, cleft palate, intellectual disability

 

(40)

NR2F2

15q26.2

AD

Ovarian dysgenesis, ovotesticular DSD

 

Congenital heart disease, somatic anomalies including

blepharophimosis-ptosis-epicanthus

inversus syndrome

(41)

NR5A1

9q33

AD

Ovarian dysgenesis, ovotesticular DSD

Testicular dysgenesis

Adrenocortical insufficiency,

spermatogenic failure, primary ovarian insufficiency, asplenia, polysplenia

(42)

PPP2R3C

14q13.2

AR

 

Testicular dysgenesis

Dysmorphic facies, retinal dystrophy, and

myopathy.

 (43)

RSPO1

1p34.3

AR

Testicular/

ovotesticular DSD

 

Palmoplantar hyperkeratosis

 (44)

SART3

12q23.3

AR

 

Testicular dysgenesis

Intellectual disability, global developmental delay and a subset of brain anomalies

 

(45)

SOX3 (Upstream deletion and

Duplication)

Xq27.1

XL

 

Testicular/ovotesticular DSD

 

Intellectual disability, hypopituitarism

(46)

SOX8 (SNV, upstream

duplication, inversion)

16p13.3

AD

 

 

Testicular dysgenesis

Variants may contribute to male and female infertility, primary ovarian insufficiency.

(47)

SOX9 (SNV, upstream

duplication, inversion)

17q24.3

AD

 

Testicular/ovotesticular DSD

Testicular dysgenesis

Campomelic dysplasia, Cooks syndrome, Pierre Robin sequence

 

 (48)

SOX10 (Duplication)

22q13.1

AD

Testicular/ovotesticular DSD

 

Peripheral and central demyelination, Waardenburg syndrome type IV, and Hirschsprung disease

 (49)

SRY (SNV, deletion,

Translocation)

Yp11.3

XL, YL

Testicular/ovotesticular DSD

Testicular dysgenesis

 

 (50, 51)

 

TSPYL1

6q22.1

AR

 

Testicular dysgenesis

Sudden infant death syndrome

(52)

WT1

11p13

AD

Testicular/ovotesticular DSD

Testicular dysgenesis

Denys-Drash syndrome; Frasier syndrome; WAGR syndrome; Meacham syndrome, Nephrotic syndrome; Wilms tumor

(53, 54)

 

 

Abbreviations: AD; autosomal dominant, AR; autosomal recessive, XL; X-linked, YL; Y-linked

 

DEFICIENCY OF TESTOSTERONE BIOSYNTHESIS

 

The inability to produce testosterone stems from defects in the activity of any of the enzymes required for testosterone biosynthesis from cholesterol (Figure 4able 5). Identified defects encompass genetic variants in Steroidogenic Acute Regulatory Protein (StAR), P450 side-chain cleavage (CYP11A1), 3β-hydroxy steroid dehydrogenase 2 (HSD3B2), 17α-hydroxylase/17,20-lyase (CYP17A1), and P450 oxidoreductase (POR) (59). In addition, apparent lyase deficiency due to variants in cytochrome b5 (CYB5) and variants in AKR1C2/4 have been very rarely described (60-62). Moreover, 17β-hydroxysteroid dehydrogenase deficiency (also known as 17β–hydroxysteroid oxidoreductase or 17-ketosteroid reductase, HSD17B3) is another cause of 46,XY DSD (63). Furthermore, variants in steroidogenic factor 1 (SF1/NR5A1), which regulates the transcription of several genes involved in steroidogenesis, may lead to ambiguous genitalia. Notably, variants in HSD3B2, POR, and NR5A1 can result in ambiguous genitalia in both 46,XY and 46,XX newborns (64-66).

 

Similar to partial gonadal dysgenesis, a partial testosterone biosynthesis defect leads to ambiguous external genitalia and variable degrees of Wolffian duct development. However, unlike partial gonadal dysgenesis, Müllerian ducts are not retained in 46,XY newborns with partial testosterone biosynthesis defects due to intact Sertoli cell function and normal AMH production.

 

Table 5. Adrenal and/or Gonadal Causes of Impaired Sex Steroid Biosynthesis Associated with Ambiguous Genitalia in 46,XX and 46,XY Individuals

Disorder

Gene/ OMIM

Adrenal Insufficiency

46,XY Gonadal Phenotype

(Testosterone Deficiency)

46,XX Gonadal Phenotype

(E2 Deficiency)

Fertility

Other Features

 

Lipoid congenital adrenal hyperplasia (LCAH)

StAR

201710

YES

Classic form: 46,XY DSD, gonadal insufficiency; ambiguous genitalia

Non-classic form: normal or NK

Classic: primary or secondary ovarian insufficiency (POI)

Non-classic: NK or normal

Classic:Absent in 46,XY; variable in 46,XX

 

P450 side chain cleavage syndrome (CAH)

CYP11A1

118485

YES

Classic form: 46,XY DSD, gonadal insufficiency; ambiguous genitalia

Non-classic form: normal

Classic: primary or secondary ovarian insufficiency (POI)

Non-classic: NK or normal

Reported in 46,XX

 

3β-hydroxysteroid dehydrogenase II deficiency (CAH)

HSD3B2

201810

YES

46,XY DSD, gonadal insufficiency; ambiguous genitalia

Non-classic form: normal, but premature adrenarche

46,XX DSD with atypical genital development; gonadal insufficiency; ambiguous genitalia

Non-classic form: normal, but premature adrenarche

Absent in 46,XY; reported in 46,XX

 

21-hydroxylase deficiency (CAH)

CYP21A2

201910

YES

Classic form: normal

Non-classic form: normal

46,XX DSD with atypical genital development;

Non-classic form: premature adrenarche, virilization, PCO

Normal in both 46,XX and 46,XY, if treated

Cave: Testicular adrenal rest tumor (m>>f)

CAH-X (when combined with Ehlers-Danlos syndrome with contiguous gene variants)

11-hydroxylase deficiency (CAH)

CYP11B1

202010

YES

Classic form: normal

Non-classic form: normal

46,XX DSD with atypical genital development;

Non-classic form: premature adrenarche, virilization, PCO

Normal in both 46,XX and 46,XY, if treated

Hypertension

Combined 17-hydroxylase, 17,20 lyase deficiency (CAH)

CYP17A1

202110

Rare

46,XY DSD, gonadal insufficiency; atypical genital development

Lack of pubertal development, POI

Possible in 46,XX with assisted fertility measures

Hypertension and hypokalemic alkalosis (not seen with isolated lyase deficiency)

P450 oxidoreductase deficiency (CAH)

POR

124015

201750

Variable

Mild to severe 46,XY DSD, gonadal insufficiency; atypical genital development

46,XX DSD with atypical genital development or

premature adrenarche, virilisation, POI, PCO

Reported

Maternal virilization during pregnancy; Antley-Bixler skeletal malformation syndrome; changes in drug metabolism

Cytochrome b5 deficiency

CYB5A

613218

NO

46,XY DSD; atypical genital development

NK

NK

Methemoglobinemia

17β-hydroxysteroid dehydrogenase III deficiency / 17-ketosteroid reductase deficiency

HSD17B3

264300

NO

46,XY DSD; atypical genital development; progressive virilization and gynecomastia at puberty

Normal

Decreased or absent in 46,XY

 

5α-reductase II deficiency

SRD5A2

607306

NO

46,XY DSD; atypical genital development; progressive virilisation and gynecomastia at puberty

Normal

Impaired in 46,XY

 

3α-hydroxysteroid dehydrogenase deficiency

AKR1C2/4

600450

600451

NO

46,XY DSD; gonadal insufficiency; atypical genital development

Normal

NK

 

Aromatase deficiency

CYP19A1

107910

NO

Normal

46,XX DSD with variable degree of virilisation at birth (ambiguous genitalia), gonadal insufficiency, POI

Impaired in 46,XX

Overgrowth and metabolic anomalies in males

Steroidogenic factor 1

NR5A1/

SF1

184757

Rare

Mild to severe 46,XY DSD; gonadal insufficiency – very variable; ambiguous genitalia

POI or normal; rarely 46,XX DSD with atypical genital development

Mostly impaired in 46,XY; variable in 46,XX

 

Abbreviations: NK; not known, POI; primary ovarian insufficiency, PCO; polycystic ovary.

 

5-ALPHA-REDUCTASE-2 DEFICIENCY (SRD5A2 VARIANTS)

 

Deficiency of the 5α-reductase enzyme arises from variants in the steroid 5α-reductase type 2 (SRD5A2) gene, which can show single point mutations to complete gene deletions. Newborns affected by this condition possess functioning Leydig and Sertoli cells, but due to their inability to convert testosterone to DHT, they may exhibit varying degrees of under-masculinized external genitalia, including genital ambiguity at birth in some cases (16, 67-69). The Müllerian ducts in affected individuals regress as expected, due to normal Sertoli cell function and AMH production. During puberty, significant virilization becomes possible in affected individuals, and the testes are capable of supporting spermatogenesis, as DHT is not required for germ cell maturation. Therefore, fertility is attainable in less severely affected individuals (70-72) or with the use of intrauterine insemination. Recent evidence also suggests that SRD5A2 activity may be even influenced by genetic polymorphisms of SRD5A2 (50).

 

GENETIC DEFECTS OF ANDROGEN ACTION

 

Complete or partial androgen insensitivity syndromes (CAIS/PAIS) are caused by genetic mutations affecting androgen receptor (AR) function (73, 74), and serum testosterone levels are typically elevated. Internationally, incidence of AIS is reported in 1 in 20,400 live born 46,XY infants, with CAIS occurring at a higher rate than PAIS (75).

 

The human androgen receptor (AR) is encoded by a single gene (AR) composed of 8 exons located in the q11-12 region of the X chromosome. The AR gene mutation database includes hundreds of AR variants that lead in varying degrees of atypical sex differentiation of 46,XY fetuses, also classified as AIS type I (76-78). A small number of complete AR gene deletions have been reported, as well as deletions that start at exons 2, 3 or 4 and extend to the terminus of the gene. In addition, a limited number of mutations resulting from premature terminations, base deletions and terminations have been identified. However, the most common type of AR gene mutation results from base substitutions (79). When mutations result in partial inactivation of AR activity (PAIS), the phenotypic variability in under-masculinization can be substantial and depends on the residual activity of the AR. In some cases, this can lead to ambiguous genitalia in newborns with a 46,XY chromosomal sex.

 

Unlike in CAIS, the underlying genetic defect in PAIS is only found in the AR gene in about 40% of cases (80). However, the clinical and biochemical phenotype is similar, and in vitro functional tests demonstrate androgen resistance in all of them. Thus, individuals with AIS without AR mutations are classified as AIS type II (73). In cases of AIS type II without AR variants, largely unidentified regulators or cofactors of the AR are responsible for the impaired AR signaling, as revealed by an AR-dependent bioassay using genital skin fibroblasts and the targeted apolipoprotein D as a biomarker (81). Given that AR activity can be regulated at various levels, the potential mechanisms of AIS type II are diverse.Recently, altered DNA methylation of the AR promoter has been observed in some individuals with PAIS (82). Additionally, mutations in the Disheveled associated activator of morphogenesis 2 gene (DAAM2) that impair nuclear actin assembly at AR have been associated with AIS type II (83).

 

Overall, individuals with PAIS experience variable degrees of end-organ unresponsiveness to androgens, resulting in varying degrees of Wolffian duct development and external genital ambiguity. Within families with the same AR mutation, phenotypic variability can occur due to variable degrees of insensitivity to androgens (84, 85). Consequently, solely relying on genetic information, it is challenging to predict whether a newborn with PAIS will identify as male, intersex, or female in later life and respond to future testosterone therapy. Similarly, genetic counselling is challenging.

 

The 46,XX Newborn With Ambiguous Genitalia

 

Ambiguous genitalia in a chromosomal 46,XX fetus invariably results from androgen excess during fetal development, and the degree of virilization (Figure 2) often provides clues about the timing and severity of the underlying disorder.

 

Similar to a 46,XY fetus, a 46,XX fetus may experience partial gonadal dysgenesis or develop testicular or ovo-testicular DSD due to pathogenic variants in genes involved in early sex determination (Table 3 and 4). Ovo-testicular DSD occurs when both ovarian and testicular tissue develop in the same individual (1). Most newborns with ovo-testicular DSD possess 46,XX chromosomes and present with ambiguous genitalia, although some affected individuals have 46,XY chromosomes or 46,XX/46,XY mosaicism. Just like in 46,XY DSD, the degree of testicular development determines the extent of masculinization/virilization of the external genitalia, Wolffian duct development, and Müllerian duct regression in affected newborns (79).  A summary of known genetic variations is given in Table 4.

 

In the majority of cases, ambiguous genitalia in a 46,XX fetus with typical ovarian organogenesis can be attributed to excessive androgen production/exposure early in utero, originating either from the fetus itself (Table 3 and Table 5) or from the mother or environment.

 

ABNORMAL FETAL ANDROGEN PRODUCTION BY CONGENITAL ADRENAL HYPERPLASIA (CAH)

 

The term congenital adrenal hyperplasia (CAH) encompasses several adrenal disorders, each linked to a mutation in one of the enzymes necessary for the biosynthesis of cortisol from cholesterol (86) (Figure 4 and Table 5). These abnormalities lead to increased ACTH secretion by the pituitary gland, which, in turn, results in the increased secretion of cortisol precursors, including adrenal androgens.

 

Deficiency of steroid acute regulatory protein (StAR) causes congenital lipoid adrenal hyperplasia, characterized by salt loss and a lack of cortisol, androgen, and estrogen secretion. Genetic 46,XX infants affected by StAR deficiency exhibit typical female external genitalia, while genetic 46,XY are born with ambiguous genitalia or may experience sex reversal in cases of severe loss-of-function variants. A similar phenotype is also observed with variants in CYP11A1.

 

Deficiency in 17α-hydroxylase/17,20-lyase leads to hypertension due to the hypersecretion of corticosterone and impaired androgen secretion. Similar to congenital lipoid adrenal hyperplasia, 46,XX fetus affected by 17α-hydroxylase/17, 20-lyase deficiency (CYP17A1) develop typical female external genitalia, while 46,XY fetus present with ambiguity.

 

Deficiency of 3β-hydroxysteroid dehydrogenase (HSD3B2) results in salt loss and impaired androgen synthesis. Affected 46,XX females may or may not exhibit minimal genital masculinization, whereas all genetic 46,XY males are variably under-masculinized (87).

 

CAH due to 11β-hydroxylase deficiency (CYP11B1) and 21-hydroxylase deficiency (CYP21A2) results in the most significant masculinization/virilization of external genitalia in 46,XX fetus compared to all other types of CAH. Additionally, 11β-hydroxylase deficiency can lead to hypertension in individuals of either sex in later life (88, 89).

 

21-hydroxylase deficiency CAH, caused by variants in CYP21A2, represents the most common form (accounting for more than 90% of cases) of CAH and is the most frequent genetic cause of DSD with ambiguous genitalia at birth in 46,XX newborns (59, 63). Cortisol deficiency necessitates replacement therapy in all classic forms of CAH. In the milder (simple-virilizing) presentation of classical 21-hydroxylase deficiency, salt loss is typically not an issue, whereas in the more severe (salt-wasting) form of classical 21-hydroxylase deficiency, salt-loss occurs and may pose a risk to the newborn shortly after birth if not treated with both mineralocorticoids and glucocorticoids (86, 90, 91).

 

The pathological mechanisms of 21-hydroxylase deficiency leading to 46,XX virilization in utero have been well described (92). It involves androgen excess due to lack of 21-hydroxylase activity during the critical time window around 6-12 weeks of gestation when the external genitalia are formed, and adrenal androgen production should be redirected to cortisol production to protect the female external genital anlage. In 46,XX CAH (Table 5), the resulting adrenal androgen excess exceeds the androgen metabolizing capacities of the fetal-placental unit, and alternative pathways of androgen production (Figure 4) are also activated, contributing to the overall androgen excess. Figure 5 illustrates in a scheme how the fetal adrenals, liver and placenta cooperate under normal conditions to metabolize fetal adrenal androgens into estrogens.

 

Figure 5. Steroid metabolic pathways in the fetal-placental-maternal unit, also known as the fetal-placental unit. The main function of the FZ of the fetal adrenal is the production of DHEA-S from cholesterol, which is transported to the placenta, desulfated to DHEA, and sequentially metabolized by placental 3β-hydroxysteroid dehydrogenase type 1 (3βHSD1), 17β-hydroxysteroid dehydrogenases (17βHSD) and cytochrome P450 aromatase (CYP19A1), to androstenedione (A4), testosterone (T) and estradiol (E2), respectively. E2 is then transported from the placenta into the maternal circulation. A4 is additionally also a substrate for placental CYP19A1, producing estrone (E1). Fetal adrenal DHEA-S is also metabolized by the fetal liver to 16α-hydroxy-DHEA-S by cytochrome P450 family 3 subfamily A member 7 (CYP3A7), which is metabolized to estriol (E3), the estrogen marker of pregnancy. 
Abbreviations: PREG, pregnenolone; 17OHPREG, 17α-hydroxypregnenolone; PROG, progesterone; DHEA, dehydroepiandrosterone; DHEA-S, dehydroepiandrosterone-sulfate; CYP11A1, cytochrome P450 cholesterol side chain cleavage; StAR, steroidogenic acute regulatory protein; CYP17A1, cytochrome P450 17α-hydroxylase/17,20-lyase; SULT2A1, sulfotransferase; CYP3A7, cytochrome P450 family 3 subfamily A member 7; CYP19A1, cytochrome P450 aromatase; HSD17B, 17β-hydroxysteroid dehydrogenase; HSD3B1, 3β-hydroxysteroid dehydrogenase type 1; HSD11B2, 11β-hydroxysteroid dehydrogenase type 2; STS, sulfatase.

Figure 5. Steroid metabolic pathways in the fetal-placental-maternal unit, also known as the fetal-placental unit. The main function of the FZ of the fetal adrenal is the production of DHEA-S from cholesterol, which is transported to the placenta, desulfated to DHEA, and sequentially metabolized by placental 3β-hydroxysteroid dehydrogenase type 1 (3βHSD1), 17β-hydroxysteroid dehydrogenases (17βHSD) and cytochrome P450 aromatase (CYP19A1), to androstenedione (A4), testosterone (T) and estradiol (E2), respectively. E2 is then transported from the placenta into the maternal circulation. A4 is additionally also a substrate for placental CYP19A1, producing estrone (E1). Fetal adrenal DHEA-S is also metabolized by the fetal liver to 16α-hydroxy-DHEA-S by cytochrome P450 family 3 subfamily A member 7 (CYP3A7), which is metabolized to estriol (E3), the estrogen marker of pregnancy. Abbreviations: PREG, pregnenolone; 17OHPREG, 17α-hydroxypregnenolone; PROG, progesterone; DHEA, dehydroepiandrosterone; DHEA-S, dehydroepiandrosterone-sulfate; CYP11A1, cytochrome P450 cholesterol side chain cleavage; StAR, steroidogenic acute regulatory protein; CYP17A1, cytochrome P450 17α-hydroxylase/17,20-lyase; SULT2A1, sulfotransferase; CYP3A7, cytochrome P450 family 3 subfamily A member 7; CYP19A1, cytochrome P450 aromatase; HSD17B, 17β-hydroxysteroid dehydrogenase; HSD3B1, 3β-hydroxysteroid dehydrogenase type 1; HSD11B2, 11β-hydroxysteroid dehydrogenase type 2; STS, sulfatase.

 

FETAL AND PLACENTAL AROMATASE DEFICIENCY

 

During fetal development, the adrenals produce substantial amounts of DHEA/S. These are either converted to 16-hydroxy-DHEA/S in the fetal liver and then transported or directly transferred to the placenta, where they are converted into estrogens (Figure 5). In case of an aromatase enzyme deficiency in the fetus (caused by variants in the CYP19A1 gene), androgen precursors accumulate and lead to virilization of 46,XX fetuses, typically leading to ambiguous genitalia at birth (93). Androgen excess due to fetal aromatase deficiency may also induce maternal virilization during pregnancy, as indicated by a deepening of the voice, oily skin and hair, and excessive hair growth (hirsutism).

 

EXCESS MATERNAL ANDROGEN PRODUCTION

 

Excessive androgen production can negatively impact fertility due to anovulation, therefore cases of maternal androgen production during pregnancy are exceedingly rare. Nonetheless, androgens originating from the mother can travers the placenta and lead to the masculinization of a female fetus. These maternal androgens usually originate from the ovaries or adrenal glands. Androgen-producing ovarian tumors include hilar cell tumors, arrhenoblastomas/androblastomas, lipoid cell tumors, and Krukenberg tumors. In contrast, pregnancy luteomas, rare non-neoplastic ovarian lesions occurring during pregnancy, are believed to be caused by hormonal effects related to pregnancy. Although exceptionally rare, androgen-secreting tumors of the adrenals can also occur during pregnancy.

 

DRUGS ADMINISTERED TO THE MOTHER DURING GESTATION

 

In 1958, Wilkins et al. reported that certain synthetic progestins administered to pregnant women, such as 17α-ethinyl-19-nortestosterone, can masculinize the external genitalia of female fetuses (94). Similarly, diethylstilbesterol (DHE), a nonsteroidal synthetic estrogen, has been linked with urogenital anomalies in both 46,XX and 46,XY fetuses (95, 96). More recently, numerous studies have indicated that fetal exposure to various endocrine disrupting chemicals (EDCs) such as pesticides, fungicides, herbicides, and plasticizers may adversely affect genital development (97-99).

 

Syndromes Associated with Ambiguous Genitalia

 

As previously mentioned in sections of 46,XX and 46,XY DSD, ambiguous genitalia can be associated with syndromes characterized by multiple congenital malformations, as summarized in Table 4. These associations arise because many of the transcription factors involved in sex development and differentiation also have roles in other aspects of development (Table 4).

 

For example, mutations of the WT-1 gene (11p3) can lead to several syndromes, including WAGR, Denys-Drash or Frasier syndromes (70-72). Mutations in SOX-9 (17q24-25) can result in campomelic dysplasia (100).

 

Mutations in DMRT1/DMRT2 (9p24.3), EMX2 (10q25.3q26.13), ATRX (Xq13.3) and WNT-4 (1p35) are often associated with developmental delay (101, 102). SF-1 (9q33) and DAX-1 (Xp21.3) play roles in the early formation of the adrenals, anterior pituitary, and parts of the hypothalamus.

 

Consequently, mutations can lead to abnormalities of these organs, in addition to those of the urogenital system. Abnormal development of the lower abdominal wall with pubic diastasis can result in urogenital anomalies observed in the VA(C)TER(L) and CHARGE syndromes, as well as bladder and cloacal exstrophy (103).

 

These examples underscore the complexities of the anomalies that may be associated with ambiguous genitalia in newborns and emphasize the importance of conducting a thorough physical examination and involving an experienced DSD team when faced with a child presenting ambiguous genitalia.

 

WORKUP OF NEWBORNS WITH AMBIGUOUS GENITALIA

 

The etiology of genital ambiguity in newborns is diverse and can have significant implications for management. Therefore, obtaining an accurate diagnosis through a medical workup is crucial. Some investigations are urgent, such as ruling out or confirming and treating potentially life-threatening adrenal insufficiency with mineralocorticoid and glucocorticoid replacement therapy in children with underlying CAH. Additionally, associated organ anomalies may require immediate attention to ensure the child’s survival, such as cardiac, pulmonary, or kidney malfunctions.

 

When the newborn is shown to be capable of excreting urine and stool without problems, and adrenal and other organ functions are found to be intact, investigations to understand genital ambiguity become primarily a psychosocial “emergency” for the family and caregivers. As a result, psychological support and holistic care by a DSD team as early as possible are essential to guide the parents and make decisions in the child’s best long-term interest. Obtaining an accurate diagnosis of the underlying cause of genital ambiguity early on is necessary for decision-making processes regarded to sex registration, hormonal and surgical treatments, and outcomes such as potential fertility and risk of gonadal malignancy. Therefore, it is recommended practice that a thorough workup should be performed for every child born with ambiguous genitalia, and guidelines for such assessments are available (22, 104-107).

 

Overall Assessment of the Child with Ambiguous Genitalia

 

Figure 6 provides an overview of the initial workup for a newborn with ambiguous genitalia. Several aspects are considered, including pregnancy and family history, as well as postnatal adaptation (such as feeding, weight gain, alertness, excretion).  In some cases, suspicion of genital ambiguity may arise during pregnancy through ultrasound surveillance. In such cases, first contact and information through a DSD team member (e.g., neonatologist, geneticist, pediatric endocrinologist) together with the DSD psychologist is recommended to guide the parents through pregnancy and alert the future care team early. Prenatal workup of ambiguous genitalia is generally not necessary if the fetus is otherwise developing normally without other organ anomalies that require attention. It is important to note that a fetus with CAH does not suffer from mineralocorticoid and cortisol deficiency in utero, as these hormones become essential only after birth (Figure 4). Genetic workup of a child with ambiguous genitalia during the prenatal period may only be recommended when there is a previous index case with a diagnosis, or when parental genetic data inform an abnormality, and when genetic counseling of the parents has led to a personalized action plan.

 

When taking the medical history of a newborn with ambiguous genitalia, asking specific questions can help guide the diagnostic workup and management. Figure 6 provides a list of important questions to consider. For example, androgenic drugs used in pregnancy may explain the virilization of a 46, XX newborns. Maternal virilization during pregnancy, for example, voice change and hirsutism may be important for the diagnoses of maternal virilizing tumors, P450 oxidoreductase (POR) deficiency, or placental aromatase deficiency. Questions about parental consanguinity are important, because many of the causes of ambiguous genitalia are recessively inherited. History of previously unexplained neonatal deaths, ambiguous genitalia, infertility or genital surgery in the family is important for identifying potential genetic etiologies.

 

In addition to the medical history, every newborn with ambiguous genitalia should undergo a comprehensive physical examination, with a special focus on signs of adrenal insufficiency (dehydration, lethargy, poor feeding) and any dysmorphic features (Figure 6). This examination may guide the diagnostic process.

 

Figure 6. Algorithm for initial workup of the child with ambiguous genitalia at birth.
Abbreviations: SDSST; Standard dose synthetic ACTH (synacthen) stimulation test, HCG; Human chorionic gonadotropin, WES; Whole exome sequencing, WGS; Whole genome sequencing, EMS; External masculinization score, EGS; External genital score, QF-PCR; quantitative fluorescence polymerase chain reaction, SRY FISH; SRY fluorescent in situ hybridization.

 

Physical Examination of the External Genital Area

 

Systematic assessment of the external genital area in newborns with ambiguous genitalia is crucial, and it should be carefully documented. Over the years, different scales and scoring systems have been used to evaluate the external genitalia. These assessments help provide a standardized way to document the physical characteristics, which is essential for comparing data over time and between different healthcare providers and patients. The use of these scales and scoring systems aids in follow-up and tracking of changes in the external genitalia.

 

Historically, the Prader or Quigley scales were commonly used for this purpose (108, 109) (Figure 7). Later, the external masculinization score (EMS) was introduced as a more comprehensive assessment tool (104) (Table 6). Nowadays, the external genital score (EGS) is recommended for evaluating and documenting the external genitalia (110) (Table 7). These tools provide a structured way to assess and record the physical findings, making it easier to monitor and communicate about the patient's condition.

 

The careful examination of the external genitalia in newborns with ambiguous genitalia provides valuable non-invasive, indirect information about prenatal exposure and response to DHT, and about the timing of this exposure. For instance, androgen exposure must have occurred in the first trimester to cause labial fusion.

 

During the genital exam, it’s important to measure the stretched phallic length, assess the quality of the corpora (erectile tissues), inspect of the labia, labio-scrotal folds, or scrotum, and document the position of the urethral opening (and vaginal opening, if applicable). Furthermore, the presence and location of palpable gonads should also be recorded. Any asymmetry in the gonads and external genitalia should be noted. Asymmetry in internal and/or external genital structures may suggest mixed gonadal dysgenesis or ovo-testicular DSD. All these observations can be documented using the external genital score (EGS), which provides a standardized way to assess and record the physical characteristics of the external genitalia (Table 7). In addition, the anogenital distance (AGD), the distance between the anus and the base of the genitalia, can be helpful in determining the extent of androgen exposure during fetal development, as a higher ratio is generally consistent with increased androgen exposure (111) (Table 8). In older infants, the anogenital index (AGI), which is the body weight standardized index of AGD [AGI/weight (mm/kg)], may be used for assessments to account for changes of the external genitalia through growth and postnatal development (111).

 

Overall, a thorough examination and documentation of the external genitalia provide important clinical information that can guide the diagnosis and management of newborns with ambiguous genitalia.

 

Figure 7. Prader and Quigley Scales for visual grading of external genital virilization. In Stage 3 a single urogenital orifice can be observed.

 

 

Table 6. External Masculinization Score (EMS)

External Masculinization Score (EMS).

Scrotal fusion

Micropenis

Urethral meatus

Right gonad

Left gonad

3

Yes

No

Normal

 

 

2

 

 

Distal

 

 

1.5

 

 

 

Labioscrotal

Labioscrotal

1

 

 

Mid

Inguinal

Inguinal

0.5

 

 

 

Abdominal

Abdominal

0

No

Yes

Proximal

Absent

Absent

An EMS score <11 out of 12 needs further clinical investigation for DSD in 46,XY.

 

Table 7. External Genital Score (EGS)

External genital score (EGS)

Labio-scrotal fusion

Genital tubercle size (mm)

Urethral meatus

Right gonad

Left gonad

3

Fused

≥31

Typical male

 

 

2.5

 

26-30

Coronal glandular

 

 

2

 

 

 Penile

 

 

1.5

Posterior fusion

21-25

Peno-scrotal

Labio-scrotal

Labio-scrotal

1

 

10-20

Scrotal

 Inguino-scrotal

Inguino-scrotal

0.5

   

Perineal

Inguinal

Inguinal

0

Unfused

≤ 10

Typical female

Impalpable

Impalpable

Median EGS (10th to 90th centile) in males < 28 weeks gestation is 10 (8.6-11.5); in males 28-32 weeks 11.5 (9.2-12); in males 33-36 weeks 11.5 (10.5-12) and in full-term males 12 (10.5-12). In all female babies, EGS is 0 (0-0).

 

Table 8. Reference Ranges for Anogenital Distance and Anogenital Index in Term Neonates

 

5th

10th

25th

50th

75th

90th

95th

Males

 

 

 

 

 

 

 

AGD (mm)

18.47

19.44

21.00

22.68

25.20

27.80

29.08

AGI (mm/kg)

5.59

5.83

6.30

6.93

7.62

8.22

8.57

Females

 

 

 

 

 

 

 

AGD (mm)

9.40

9.85

10.55

11.65

12.60

13.45

14.10

AGI (mm/kg)

3.04

3.15

3.38

3.59

3.80

4.00

4.19

Anogenital index (AGI): the body weight standardized index of AGD [AGI=AGD/weight (mm/kg)] and found that using AGI as parameter has a better correlation with age.

 

Basic Auxiliary Investigations in the First Days

 

In the first few days of life, basic investigations are essential to evaluate adrenal and gonadal development and function in otherwise healthy newborns with ambiguous genitalia. Some key aspects to address include serum electrolytes and glucose levels. They should be monitored daily as cortisol deficiency can manifest as hypoglycemia in newborns affected by CAH. Body weight and feeding characteristics should also be monitored as excessive weight loss may indicate dehydration and electrolyte disturbances.

 

KARYOTYPE

 

Besides careful clinical evaluation, first-line investigation of an individual with DSD involves confirming the chromosomal sex using quantitative fluorescence polymerase chain reaction (QF-PCR) and karyotype (22). QF-PCR detects a series of markers on the sex chromosomes and has a turn-around time of about 1–2 days. This technique has largely replaced fluorescence in situ hybridization (FISH) methods. The analysis of karyotype is a cytogenetic technique (involving G-banding) and is crucial for the initial classification of a newborn with ambiguous genitalia (Table 3). Some centers have replaced karyotyping by array-comparative genomic hybridization (aCGH) or SNP array, with faster turn-around times of 5–10 days. But these techniques will not detect structural chromosomal rearrangements and may be less effective at detecting sex chromosome mosaicism.

 

HORMONE STUDIES 

 

Detailed hormone studies may be indicated including serum gonadotropins (LH, FSH), androgens and androgen precursors (17-hydroxypregnenolone, 17-hydroxyprogesterone, androstenedione, testosterone, dihydrotestosterone), adrenal steroids and hormones (cortisol, aldosterone, and their precursors; ACTH, renin), and anti-Müllerian hormone (AMH) (1, 24).

 

Care must be taken when interpreting the results in premature babies, in whom these studies may need to be repeated at a later age and special normative value may apply. Even in term babies the timepoint and the clinical context (mode of delivery, drug effects, actual stress level etc.) need to be considered when evaluating hormonal data immediately after birth. Hormonal data change massively during delivery (e.g., surge) and within days thereafter. Additionally, it's essential to be aware that normative values for hormone levels can vary depending on the laboratory and the methods used for testing. The interpretation of hormonal data should be done by experienced healthcare providers and, if necessary, in consultation with a specialist in DSD.

 

IMAGING STUDIES

 

Ultrasound imaging of the abdominal-pelvic organs is a valuable diagnostic tool in the evaluation of newborns with ambiguous genitalia. It provides important insights into the internal reproductive and urinary structures, helping to determine the presence and characteristics of gonads, Müllerian structures (e.g., the uterus), and any associated abnormalities of the urinary tract. It can be performed at the first visit of a child with ambiguous genitalia. Of course, this investigation is not easy and may only be performed by an experienced ultra-sonographer to reveal meaningful results. Ultrasound can also detect abnormalities of the urinary tract (kidney, ureters, bladder) that sometimes occur in conjunction with genital ambiguity. Interestingly, data show that a child with a uterus and a female internal system that presents with ambiguous genitalia without gonads in the genital folds and has a 46,XX karyotype, will have in 80% of cases a diagnosis of a 46,XX CAH (most likely 21-hydroxylase deficiency). The information needed to come to this conclusion in a newborn with ambiguous genitalia can be available within 48 hours in institutions with an experienced DSD team.

 

In a second step, usually after months or years, additional imaging studies with MRI can be helpful in identifying both the type and extent of internal sex organ development in more details. An MRI may reveal further details on Müllerian structures (uterus remanent, fallopian tubes, upper portion of the vagina) than an ultrasound and can also help for localizing abdominal gonads. MRI may also be used for surveillance of the gonad at risk for cancer.

 

A genitogram had been used in the past (often early) to visualize the urinary tract, and to determine its position in relation to the vagina or vagino-utricular pouch but is no longer performed in most institutions.

 

However, in follow-up, cystoscopy and vaginoscopy may be used. These investigations are invasive procedures requiring general anesthesia, and therefore need to have a clear indication. Often, they are performed after complications such as urinary tract infections, to better understand the anatomy and its consequences, and in preparation for a surgical procedure (112).

 

Overall, the choice of imaging studies and procedures should be guided by the individual's clinical presentation, diagnosis, and the specific clinical questions that need to be addressed. In many cases, a multi-disciplinary DSD team collaborates to determine the most appropriate imaging approach to provide a comprehensive evaluation and guide treatment decisions.

 

GENETIC STUDIES

 

The rapid advancement of genetic technologies, particularly massive parallel sequencing methods like DSD panels, whole exome sequencing (WES), and whole genome sequencing (WGS), has significantly expanded our understanding of the genes involved in sex development. As a result, genetic workup has become an integral part of the recommended routine evaluation for individuals with DSD and thus ambiguous genitalia at birth (8, 22, 24, 113, 114).

 

While making a precise genetic diagnosis is still a challenge in some cases, the use of advanced genetic techniques has improved the diagnostic yield. Currently, a clear molecular-level diagnosis can be established in approximately half of individuals with 46,XY DSD who undergo genetic workup. However, as genetic technologies continue to evolve, and our understanding of sex development genes expands, it is likely that more cases, including those involving individuals with ambiguous genitalia at birth, will receive accurate and valid diagnoses. Therefore, re-review of genetically unsolved cases by a DSD team and genetic re-analysis by DSD-specialized teams is recommended to achieve a final diagnosis in as many as possible.

 

In summary, the integration of genetic workup, in combination with ongoing advancements in technology and genetic knowledge, is expected to enhance our ability to provide individuals with DSD, including those with ambiguous genitalia at birth, with more precise and comprehensive diagnoses in the future. This progress will lead to better-informed clinical decisions and management strategies for newborns with ambiguous genitalia.

 

DIFFERENTIAL DIAGNOSIS

 

Generally, the result from the karyotype plays a major role in the differential diagnosis and workup of DSD and thus ambiguous genitalia (Table 3). In most cases, the chromosomes in newborns with ambiguous genitalia will be either 46,XX or 46,XY. In rare instances, chromosomal aberration such as 45,X/46,XY or 46,XX/46,XY mosaicism may be the underlying cause explaining the phenotype. Additionally, measurements of specific hormones, such as serum luteinizing hormone (LH), follicle-stimulating hormone (FSH), sex steroids, AMH, inhibin B at birth and at follow-up, in the so-called mini-puberty of infancy period, can provide valuable information for the differential diagnosis of DSD and ambiguous genitalia.

 

In the following we provide two flowcharts and give some general information for the step-by-step workup of newborns with ambiguous genitalia and either 46,XX or 46,XY chromosomes.

 

46, XX DSD

 

A 46, XX karyotype in a newborn with ambiguous genitalia indicates that the child is a genetic female who was exposed to excessive amounts of androgens during fetal life. A flowchart for differential diagnosis is given in Figure 8.

 

Marked elevation of plasma 17-hydroxypregnenolone, 17-hydroxyprogesterone, 21-deoxycortisol and androstenedione, along with male-typical levels of testosterone, are characteristic of 21-hydroxylase deficiency (CYP21A2). High values of corticosterone and 11-deoxycortisol, along with elevated androgens, indicate 11β-hydroxylase deficiency (biallelic mutations in CYP11B1). In 3beta-HSD deficiency (biallelic mutations in HSD3B2), calculation of ratios of delta-5 (pregnenolone, 17OH-pregnenolone, DHEA) over delta-4 (progesterone, 17OH-progesterone, androstenedione) steroid metabolites (see Figure 4) will point towards the genetic defect.

 

Overall, comprehensive steroid profiling from blood or urine using chromatographic, mass spectrometric methods allow the diagnosis of most steroidogenic defects causing androgen excess CAH in 46,XX newborns (115-117) (see also Table 5). In addition, they have led to the description of novel alternate pathways to produce active androgens in the intermediate metabolism through the backdoor or 11-oxygenated pathways, especially in disorders such as 21-hydroxylase or POR deficiencies, thereby enhancing our understanding and diagnostic possibilities (115, 117, 118)(Figure 4). However, these methods are not (yet) widely available, and the interpretation of these data is not trivial. In addition, instruments and methods vary from lab to lab. Thus, method validation is tedious and comparison between labs difficult so far (119-121).

 

When excess maternal androgen production is the underlying cause for masculinization of a female fetus, the source of these steroids is eliminated postnatally. Thus, the various steroids studied in affected newborns will be in the female-typical range despite masculinization of the external genitalia. Then, detailed examination of the mother may provide the explanation.

 

Likewise, in cases of mild POR deficiency or aromatase deficiency, in which the feto-placental unit was not able to metabolize the fetal androgens and led to ambiguous genitalia, the problem will not (always) be visible in the newborn’s steroid metabolome, and the diagnosis may only be made by genetic testing. If not, only in later life, when aromatase activity is needed for estrogen production in puberty, or when ovarian cysts hint an underlying problem, the diagnosis may be made (Figure 8).

 

In individuals with ovo-testicular DSD and 46,XX chromosomes, masculinization arises from androgens secreted by the testicular portion of the differentiated gonads. Androgen production is similar to that produced by testes in unaffected males except that the amount is usually smaller. The degree of masculinization of the genitalia is thus linked to the amount of functioning testicular tissue, which can be determined by a serum AMH measurement (Table 9) (122).

 

Very low values of AMH and female-typical values for LH/FSH (Tables 9 and 10) are expected in 46,XX newborns with masculinized/virilized genitalia that is attributed to CAH or excess maternal androgen production during gestation (122, 123). AMH is higher in newborns with ovo-testicular DSD, due to Sertoli cell development in the testicular portion of their gonads where AMH is mainly produced.

 

On occasion, translocation of the pseudo-autosomal part of the Y chromosome along with a mutated SRY gene to an X chromosome occurs. The result is partial masculinization of the genitalia in a 46,XX newborn. With maturity, the phenotype of affected individuals closely resembles that of boys and men with Klinefelter syndrome.

 

Figure 8. 46,XX DSD decision tree diagram.
Abbreviations: USG: Ultrasound; 17OHP: 17OH-Progesterone; DSD: Differences in sex development; CAH: Congenital adrenal hyperplasia; 21OHD: 21α-Hydroxylase deficiency; 11β-OHD: 11β-Hydroxylase deficiency; 3β-HSD2: 3β-Hydroxysteroid dehydrogenase type 2; PORD: P450 oxidoreductase deficiency.

 

 

Table 9. Normal Values for AMH (ng/mL)* in Fetuses, Newborns and Infants Under 1 Year (122)

 

Female

Male

 

n

Median

5-95th percentile

n

Median

5-95th percentile

Fetal blood

25

<0.4

<0.4-0.5

23

44.4

13.4-86.2

Cord blood

53

<0.4

<0.4-1.1

48

30.7

7.4-610.7

1-30 days

70

<0.4

<0.4-1.3

73

57.2

23.8-124.0

31-120 days

31

0.8

<0.4-6.4

39

88.4

46.8-173.0

4-12 months

13

1.0

<0.4-2.7

53

124.7

67.4-197.0

* AMH/MIS enzyme immunoassay kit cat 2368, Immunotech-Beckman Coulter, France

 

Table 10. LH and FSH Values (means ± SD) From Female and Male Newborns. Modified from (123)

 

 

Female

Male

Age (days)

Hormone

n

Median

Mean±SD

n

Median

Mean±SD

1-5

FSH (IU/L)

31

1.80

2.00 ± 1.37

30

0.85

0.96 ± 0.60

LH (IU/L)

0.20

0.48 ± 0.66

0.20

0.39 ± 0.48

6-10

FSH (IU/L)

17

1.40

2.44 ± 2.52

15

1.40

2.91 ± 4.38

LH (IU/L)

0.30

0.45 ± 0.33

0.30

0.45 ± 0.33

11-15

FSH (IU/L)

8

8.95

8.16 ± 4.27

17

3.00

3.71 ± 2.69

LH (IU/L)

1.60

1.58 ± 1.28

2.90

3.55 ± 2.84

16-20

FSH (IU/L)

6

1.90

1.62 ± 1.05

14

2.15

2.63 ± 1.45

LH (IU/L)

0.35

1.03 ± 1.39

3.65

4.13 ± 2.76

21-25

FSH (IU/L)

3

3.90

7.07 ± 5.92

7

2.10

2.50 ± 1.51

LH (IU/L)

0.50

0.46 ± 0.25

2.70

2.86 ± 1.51

26-28

FSH (IU/L)

8

6.15

9.74 ± 9.89

8

2.40

2.25 ± 0.81

LH (IU/L)

2.80

2.75 ± 2.39

1.40

2.22 ± 2.37

Abbreviations: LH, luteinizing hormone; FSH, follicle-stimulating hormone.

 

46, XY DSD

 

A 46, XY karyotype reveals that one is dealing with a genetic male who was under-masculinized during fetal development. A flowchart for differential diagnosis is given in Figure 9.

 

Laboratory findings of normal or elevated testosterone and DHT with normal AMH (Figure 9) indicate a diagnosis of AIS. If testosterone levels are normal but DHT levels are low, a diagnosis of steroid 5α-reductase deficiency can be made. Low levels of testosterone and DHT, along with marked elevation of some androgen precursors, indicate a deficiency of one of the enzymes required for androgen biosynthesis (Figure 9). For example, if the elevated precursors include androstenedione and 17-hydroxyprogesterone, then the defective enzyme is 17-ketosteroid reductase (HSD17B3). In all cases of testosterone biosynthetic defects, AMH levels are similar to those observed in healthy 46,XY male infants with male-typical external genitalia (Figure 9). With early defects of steroidogenesis that affect both adrenal and gonadal function, it is though important to evaluate adrenal function, as it might be impaired with deficiencies of STAR, CYP11A1, HSD3B2, CYP17A1 and POR (Table 5, Figure 9).

 

Finally, when all androgens and their precursors are below normal levels, one is dealing with gonadal dysgenesis or 46,XY ovo-testicular DSD. In these cases, AMH values should also be low. In contrast, for babies affected by Leydig cell hypoplasia, androgens and their precursors are low, while AMH values should be in the normal male range.

 

Figure 9. 46,XY DSD decision tree diagram.
Abbreviations: USG: Ultrasound; 17OHP: 17OH-Progesterone; DSD: Differences in sex development; AMH: Anti-Müllerian hormone; T: Testosterone; N: Normal; PMDS: Persistent Müllerian duct syndrome; AIS: Androgen insensitivity syndrome; SRD5A2: 5α-reductase A2; HCG: Human chorionic gonadotropin; DHT: Dihydrotestosterone; 3β-HSD2: 3β-Hydroxysteroid dehydrogenase type 2; CYP17A1: 17α-Hydroxylase/17-20 lyase; POR: P450 oxidoreductase; StAR: Steroidogenic acute regulatory protein; CYP11A1; Cholesterol side-chain cleavage enzyme; 17β-HSD3: 17β-Hydroxysteroid dehydrogenase type 3; AKR1C2/4: Aldo-keto reductase family 1 member C2/4; CYB5: Cytochrome b5.

 

Testing Early Postnatal Gonadal Function and Mini-Puberty

 

In cases of ambiguous genitalia where a diagnosis may not be reached through first line investigations, and where a gonadal malfunction is suspected, functional testing and/or re-analysis of gonadal function at the timepoint of mini-puberty may be helpful, especially also with respect to having information when making considerations towards sex of registration (e.g., in utero effect of sex hormones on the brain) and prediction of later pubertal development and fertility potential.

 

Mini-puberty is the time window at day 30-100 after birth, when the hypothalamic-pituitary-gonadal (HPG) axis is re-activated for a short time frame in infancy, before going to inactivity until puberty. Little is known about the purpose of mini-puberty, but as the HPG axis is active and stimulates the gonads, it is a time window of opportunity to assess gonadal function without the necessity to stimulate with human chorionic gonadotrophin (hCG).

 

As a principle, in mini-puberty, the typical ovaries of a 46,XX newborn show little functional activity, while the typical testes of a 46,XY newborn are quite (re)active in testosterone (T), DHT, AMH and inhibin B production. While androgens are produced in the Leydig cells of the testes, AMH and inhibin B originate from the Sertoli cells. Table 11 gives an overview of the hormonal values observed during normal mini-puberty in healthy 46,XX and 46,XY infants (124).

 

Table 11. Evaluation of Reproductive Hormones During Mini-Puberty in Infants. Normative Values of Different Serum Hormones in 46,XX Female and 46,XY Male Infants. Modified from  (124)

 

 

Females

Males

 

Post-natal time (months)

n

GMean

2.5-97.5th percentile

n

GMean

2.5-97.5th percentile

LH (IU/L)

2-3.5

432

<LOD

<LOD-0.98

581

1.71

0.62-4.08

3.5-5

110

<LOD

<LOD-1.25

166

1.40

0.54-3.32

FSH (IU/L)

2-3.5

435

3.98

1.23-17.4

578

1.19

0.41- 3.02

3.5-5

111

3.93

1.30-17.7

165

1.11

0.42-2.68

Testosterone (RIA,nmol/L)

2-3.5

74

<LOD

<LOD-0.40

592

3.04

0.69-7.60

3.5-5

14

<LOD

<LOD-NA

168

1.97

<LOD-7.01

Testosterone

(LC-MS/MS, nmol/L)

2-3.5

165

<LOD

<LOD-0.21

251

4.75

1.35- 11.3

3.5-5

125

<LOD

<LOD-0.17

175

2.25

0.32- 9.65

Estradiol (pmol/L)

2-3.5

455

29

<LOD-79

571

<LOD

<LOD-47

3.5-5

113

31

<LOD-98

160

<LOD

<LOD-49

SHBG (nmol/L)

2-3.5

427

133

67-264

579

135

66-268

3.5-5

110

141

72-282

162

143

71-262

Inhibin B (pg/mL)

2-3.5

423

62

<LOD-184

571

379

229-631

3.5-5

106

67

<LOD-174

158

379

222-662

AMH (pmol/L)

2-3.5

339

11

<LOD-49

48

1013

425- 1810

3.5-5

67

15

2-46

12

1183

797-NA

Abbreviations: LOD, Limit of detection; GMean, geometric mean; LH, luteinizing hormone; FSH, follicle-stimulating hormone; T, testosterone; RIA, radioimmunoassay; SHBG, sex-hormone binding globulin; AMH, anti-Müllerian hormone; LC-MS/MS, liquid chromatography-tandem mass spectrometry; NA, not available.

 

HCG Testing of Gonadal Function in a DSD Newborn

 

The human chorionic gonadotrophin (hCG) test can assess whether in a newborn with ambiguous genitalia, functioning Leydig cells are present. The test may therefore be useful for diagnosing testosterone biosynthesis defects such as HSD17B3 or SRD5A2 deficiency (Table 12). Several protocols for the hCG test exist, but all baseline serum samples are taken for T, DHT and precursors. Then, one to three intramuscular injections of high dose hCG (500-1500 IU) are given at 24 hours interval, and finally repeat stimulated samples are taken for androgen measurements after 72 or 24 hours, respectively, after the last injection. The following Table 12 shows how results of the test may be interpreted in various DSD conditions that may lead to ambiguous genitalia (125, 126).  

 

Table 12. Interpretation of hCG Stimulation Test Results (125, 126)

Indication

Consideration

Abnormal result

Testosterone biosynthesis defects

T before and after hCG administration

T increase < 100-150 ng/dL (3.5-5 nmol/L)

5α-Reductase deficiency

T/DHT ratio after hCG administration

≥ 8.5 for minipuberty,

≥ 10 for prepuberty,

≥ 17 for puberty,

> 20 in genetically proven cases using LC-MS/MS

17β-HSD3 deficiency

T/A4 ratio after hCG administration

< 0.8

AIS

T, SHBG before and after hCG administration

T increases but SHBG does not change significantly

Abbreviations: T, testosterone; DHT, dihydrotestosterone; HCG, human chorionic gonadotropin; A4, androstenedione; SHBG, sex-hormone binding globulin; AMH, anti-Müllerian hormone; LC-MS/MS, liquid chromatography-tandem mass spectrometry; 17β-HSD3 deficiency, 17β-hydroxysteroid dehydrogenase type 3 deficiency; AIS, androgen insensitivity syndrome

 

RISK OF MALIGNANCY OF THE DSD GONAD

 

Individuals with certain disorders of sex development (DSD), especially those involving Y chromosome material, have an increased risk of developing gonadal tumors. These tumors are mainly gonadal germ cell tumors (GGCTs), which occur in about 15 % of all DSD cases, with the risk varying depending on the age and specific underlying diagnosis. Gonadal stromal tumors and smooth muscle leiomyomas/hamartomas are less common and are found in only about 0.9% of DSD cases (127). Risk factors for development of gonadal malignancy in DSD are shown in Figure 10. Several factors contribute to the increased risk of gonadal malignancy in individuals with DSD, including genetic factors, the presence of Y chromosome material, and the type of DSD. Therefore, ongoing monitoring and appropriate management are essential to minimize this risk. While gonadectomy (surgical removal of the gonads) is not recommended for every case of 46,XY DSD, it's crucial to consider the risk of malignant tumors in the decision-making process (1, 69). The consensus statement on the management of DSD provides guidelines and recommendations regarding the timing of gonadectomy for specific patients. These recommendations aim to balance the potential benefits of gonadectomy with the associated risks and individual patient needs.

 

In individuals with DSDs, germ cells typically undergo a process of apoptosis (programmed cell death) as the body grows and develops. This process eliminates germ cells that are not needed for reproduction. However, in some cases, certain germ cells may not undergo the normal maturation process and remain in an immature state. These immature germ cells can continue to express embryonic markers such as octamer-binding transcription factor 3/4 (OCT3/4) encoded by POU5F1; stem cell factor (SCF), also known as c-KIT ligand (encoded by KITLG); and placental alkaline phosphatase (PLAP). These cells are susceptible to developing into cancerous lesions, starting as pre-invasive lesions (gonado-blastoma) or as precursors to invasive GGCT (dysgerminoma) (Figure 10).

 

DSD patients who have Y chromosome material in their gonadal karyotype are at risk of GGCTs regardless of the degree of testicular differentiation. Y chromosome material associated with gonadal tumors is GBY locus which is around the centromere of the Y chromosome and includes the gene for ¨testis-specific protein on the Y chromosome¨ (TSPY, located in Yp11.2). Other potentially related genes are SRY and DYZ3.

 

The risk of developing gonadal tumors in gonadal dysgenesis may also vary according to the underlying molecular etiology. Patients with genetic defects that lead to early blockage of gonadal development such as mutation or deletions of SRY or WT1 have a higher risk. The risk of developing gonadal tumors ranges from 40% to 60% in gonadal dysgenesis due to WT1 gene mutations (Frasier syndrome and Denys-Drash syndrome). Early bilateral gonadectomy is therefore recommended. Similarly, early bilateral gonadectomy is recommended in patients with complete or partial gonadal dysgenesis due to SRY gene mutations due to the 20% to 52.5% risk of gonadal tumor development. Gonadal dysgenesis due to MAP3K1 gene mutations is another indication for early gonadectomy (127). Gonadal tumor risk is also present, though somewhat lower in gonadal dysgenesis due to 45,X0/46,XY mosaicism. The exact time of malignancy development and the prevalence of tumors in other molecular etiologies require further research for a comprehensive understanding.

 

Histologically the risk of gonadal tumor development increases with gonadal immaturity. Fully developed gonads such as the testes, ovary or ovotestis have lower risk of malignancy than streak or dysgenetic testes (Figure 10). In ovo-testicular DSD, the risk of germ cell tumors is low (3%) (1). Newborns with PAIS are at high risk (50%) for developing gonadal tumors, and bilateral orchiopexy or gonadectomy is recommended at the time of diagnosis, if the testes are located in the abdomen (1, 128).

 

The risk of developing germ cell malignancy in newborns with CYP11A1, HSD3B2 or CYP17A1 deficiency is unknown. Newborns with 17-ketosteroid reductase deficiency have a fairly low risk for malignancy (1). Tumor risk in newborns with Leydig cell hypoplasia is unclear, and thus the recommendation for gonadectomy is not well established (1, 25, 129, 130).

 

Overall, it remains challenging to predict the personal risk of gonadal neoplasia (mostly GGCT) for a DSD individual, because it depends on many other factors besides genetics. Stimulating gonadotropins and gonadal hormones during mini-puberty and after puberty onset are also regarded as risk factors increasing gonadal tumor risk in patients with DSD. Similarly, gonads located intra-abdominal carry a higher tumor risk. The management of gonadal tumor risk in individuals with DSD is a complex issue that requires careful consideration and a personalized approach, taking into account the specific DSD subtype, genetic factors, and patient preferences. Regular monitoring and discussions with a healthcare team experienced in DSD management are essential to make informed decisions about gonadectomy. If gonads are located in the scrotum, then periodic examination of the testes may be advised as an alternative to gonadectomy, while an intra-abdominal gonad at risk may only be monitored by imaging methods such as ultrasound or MRI.

 

Figure 10. Risk of malignancy development in the DSD gonad.
Abbreviations: (OCT3/4); Octamer-binding transcription factor 3/4, TSPY; testis-specific protein on the Y chromosome, c-KITL; c-KIT ligand also called stem cell factor (SCF).

 

SEX REGISTRATION OF A NEWBORN

 

Assigning or registering the sex of a newborn with atypical external genitalia is a complex and important decision that requires careful consideration, consultation, and a patient-centered approach. The parents and DSD network team should not rush but take the time needed to come to a consensus in the best interest of the child in its environment. Not knowing whether the newborn is a girl or boy causes mostly a lot of distress and one is tempted to call it an emergency, but if there are no medical reasons to make it an emergency (like e.g., suspected adrenal insufficiency or additional organ anomalies), it is important to declare it accordingly and inform that the newborn is not at risk and actually does not care whether it is a male or female or intersex person at this moment. Thus, the stress of ambiguity of the newborn is actually at this moment “only” an issue of the parents and the care team who mostly wish to know ASAP whether the child can be assigned/registered a “correct” sex that might also fit later in life.

 

To consider sex registration, not only is it important to take into account the underlying cause and knowledge on the possible outcome of the specific, often unique condition, but also the family, cultural, and societal preconditions and aspects. Most societies have a binary sex registration obligation by legislation, but some societies have more recently introduced a third sex category besides male and female. Although this seems to make things easier, it has been discussed that such categorization in a third or other or unclassified group might also stigmatize a person in a society that is mostly binary. Thus, in real life practice, there is rumor that a third sex category is rarely chosen for a newborn with ambiguous genitalia by the parents and the DSD team. Overall, a personalized approach for every newborn with ambiguous genitalia involving the parents and the DSD network team is recommended following the shared-decision-making process (12, 13).

 

Most important is that the team around the newborn with ambiguous genitalia works together in the best interest of the child with the latest knowledge of the literature and expertise when taking the decision of sex registration, and still remains humble and open minded for the child’s future that may follow a different path leading to sex reassignment later in life. Therefore, continuous support of the family and the child by a DSD psychologist is extremely helpful to recognize early signs of gender dysphoria, inform the child about the condition in an age-appropriate manner and follow its will, if case sex reassignment is desired.

 

Generally, people believe that the sex chromosomes indicate a person’s “true sex” and  laws exist supporting this idea, to the detriment of some individuals affected by DSD (131). Scientifically speaking, it is clear that the majority of genes on the X chromosome do not influence sex development and differentiation, although the AR gene is necessary for phenotypic masculinization. Concerning the Y chromosome, only the SRY gene contributes to testicular formation. In fact, most of the genes required for sex development and differentiation are found on the autosomes (1). Therefore, chromosomes do not dictate the sex of rearing in newborns affected by a DSD and are only one piece of a big puzzle (Figure 3).

 

Although there is no overall recommendation for sex registration at birth for a child with ambiguous genitalia, some helpful information is available from larger patients’ cohorts with a specific DSD diagnosis in the literature.

 

Newborns presenting with 46,XY chromosomes and female external genitalia at birth (thus actually not with ambiguous genitalia) due to complete androgen insensitivity syndromes (CAIS), complete gonadal dysgenesis (Swyer syndrome), or other complete loss of testosterone biosynthesis live successfully when assigned a female sex of rearing. Female assignment in such cases is widely accepted by patients throughout their lives despite challenges that they may experience regarding sexual dysfunction and infertility (1, 132-134).

 

For newborns with ambiguous genitalia affected by partial gonadal dysgenesis, no general recommendation can be made. Fewer procedures were historically required for surgical feminization compared to surgical masculinization of the genitalia. However, the functional outcome in individuals who received feminizing or masculinizing procedures is less than optimal (135, 136). Additionally, rates of satisfaction with sex of rearing are similar for affected individuals whether raised male or female (1, 20, 137). Thus, the decision should not be made on the basis of “surgical possibilities”, and overall, such surgical procedures should be executed with reservation. Clearly, further studies are needed to elucidate why some individuals with partial gonadal dysgenesis experience gender dysphoria, while others do not.

 

Likewise, no general recommendation can be made for patients with ambiguous genitalia due to a partial androgen biosynthesis defect, (e.g., partial 3βHSD2, CYP17A1, POR, StAR, CYP11A1 deficiencies). But some studies suggest that the degree of masculinization of the external genitalia may reflect the degree of virilization of the brain and thus the gender preference of this person (138). This might be true for 46,XY newborns with ambiguous genitalia that are manifesting with severe hypospadias, but not without exceptions. For patients with variants in the SRD5A2 or HSD17B3genes recognized with ambiguous genitalia at birth (or even missed because manifesting with a female-typical external genitalia), there is quite a strong body of data recommending male sex registration at birth as this is the preferred sex of these persons when asked in later life (139, 140). Normal virilization at puberty, coupled with intact fertility potential, are strong factors for this recommendation at birth. If registered as female at birth and diagnosed later, gender dysphoria and desire of sex reassignment later in life is very likely.

 

Similar to partial gonadal dysgenesis, consensus regarding an optimal sex of registration for newborns affected by PAIS does not exist. Although earlier data showed that fewer procedures were usually required for surgical feminization, compared to surgical masculinization, in a person with ambiguous external genitalia in the past, when larger series of genital surgery were analyzed, it is unclear if the functional outcome was better among newborns who received feminizing procedures compared to those who received masculinizing procedures. Rates of satisfaction with sex of rearing are similar for individuals with PAIS raised female or male (132, 141). While reports exist of impaired sexual function in people with PAIS raised male, it is highly suspected that sexual functional outcomes in affected people raised female are similarly poor (137). Reports of sexual satisfaction for people with PAIS who have not received genitoplasty in infancy, or at a later age, are limited. However, more information on outcome of newborns with ambiguous genitalia who had no genital surgery, at least not in infancy and childhood, will hopefully soon be added to the medical literature as current developments recommend abstaining from genital surgery.

 

By contrast, in 46,XX newborns with ambiguous genitalia due to CAH, it is generally recommended to register them a priori in the female sex, even if they are severely virilized (142-144). Masculinization due to 21-hydroxylase deficiency does not impair the development of the ovaries or the Müllerian ducts and thus fertility potential. Limited information is available about the medical and psychosexual outcome of 46,XX newborns affected by 21-hydroxylase deficiency and raised male (145, 146), although these patients do tend to have an increased risk of gender dysphoria compared to their female counterparts (147). Thus, a female sex of rearing is typically the decision for 46,XX newborns affected by 21-hydroxylase deficiency (145). Female sex of rearing in 46,XX newborns with ambiguous genitalia is also recommended for conditions resulting from maternal overexposure to androgens and placental aromatase deficiency.

 

FINAL REMARKS TO OVERALL MANAGEMENT

 

Only recently has the understanding of, and reaction to, having a child with ambiguous genitalia received systematic study. For some parents and caregivers, feelings of isolation and concern over what the future may hold for their affected child in terms of stigmatization and sexual dysfunction are paramount (148). One potential approach to ameliorate this issue is to provide information and management options to the family early in pregnancy or as soon as the child is born. Guidance by a DSD network team is therefore extremely important.

 

Some research has been done to examine the effects of prenatal diagnosis and treatment. In females with 21-hydroxylase deficiency CAH, one of the most common causes of ambiguous genitalia, dexamethasone treatment of the pregnant mother has been reported to reduce virilization of an affected female fetus by 80-85% (149), but this approach is no longer offered or recommended in guidelines because of potential adverse effects of dexamethasone on the developing fetus. Before the discovery of methods to measure fetal DNA in maternal blood, which enabled specific diagnosis of sex and whether the fetus inherited and was affected by the mutation, controversies existed because 7 unaffected fetuses would be treated unnecessarily (1:4 chance of inherited mutation and 1:2 chance of male vs female), So, on the one hand 7 unaffected fetuses and the mother would be exposed to an unnecessary drug, while on the other hand, even in affected fetuses long-term outcome showed adverse effects on neurocognitive parameters (150). Today, noninvasive prenatal diagnosis using massive parallel sequencing of cell-free fetal DNA can be used to determine a specific diagnosis, such as CAH, as early as 6 weeks of gestation (149, 151). Even preimplantation diagnostics is available (152). These techniques could be used to look at other autosomal dominant or X-linked conditions that cause ambiguous genitalia, especially when the fetus is known to be at risk (151). Further studies need to be done on this subject, but for families who already have an affected child or know that they are carriers, this may be an option that needs to be discussed with a DSD geneticist.

 

Patients for whom prenatal diagnosis and/or treatment is not an option, prognosis and management has improved significantly over time, but many open questions and controversies still exist. Our studies of parents of children with DSD reveal that the appearance of atypical genitalia can result in significant stress and maladaptive parenting strategies. For example, mothers of children with ambiguous genitalia experience greater stress if their child has not received “corrective” genital surgery (153) and some parents believe such surgeries eliminate stigmatization that may arise due to their child’s ambiguous genitalia However, as their children mature past infancy, some parents realize that their child’s DSD has not been ameliorated by genitoplasty, and concerns for their son or daughter resurface (148). To illustrate, among parents studied by our group, caregivers of adolescents with DSD experienced increased stress as their child matured despite the fact that many had received earlier surgery (154). Additionally, parents of children reared male reported the most perceived child vulnerability for their sons (155),and their reported levels of depression were directly associated with more atypical genital appearance for their sons (156). Overall, parents report that while early genitoplasty seems to “fix” some of their concerns for their child with DSD, this “fix” is not long-lasting. Instead, what needs to be emphasized to families is how to support their child who is different, but not damaged. Family centered, interdisciplinary care, with open and clear communication between patients, parents, and caregivers has been shown to be essential for optimal quality of life (157). In addition, networks of families who have the personal experience of parenting children with ambiguous genitalia can be an invaluable resource (158). Reservation towards early interventions needs to be considered,keeping in mind to preserve the personal right of the child to take his/her own decision towards body integrity. Any intervention that can be delayed without harming the newborn with ambiguous genitalia should be postponed until personal consent can be given. Finally, referring parents to support groups and introducing them to other caregivers of children born with ambiguous genitalia is extremely important to optimize parents’ understanding and acceptance of their child’s condition. With increased understanding and acceptance, optimal growth and development for children born with ambiguous genitalia may be obtained.

 

In conclusion, this Chapter give some insight into the current diagnostic evaluation and care of the newborn with ambiguous genitalia. We underscore the importance of a multidisciplinary and compassionate approach to the care of newborns with ambiguous genitalia, acknowledging the complex medical, psychological, and emotional aspects involved in such cases. We highlight the critical role of specialized teams and support in ensuring the well-being of both the child and his/her family.

 

REFERENCES

 

  1. Hughes IA, Houk C, Ahmed SF, Lee PA. Consensus statement on management of intersex disorders. Arch Dis Child. 2006;91(7):554-63.
  2. Halil H, Oguz SS. Establishment of normative data for stretched penile length in Turkish preterm and term newborns. Turk J Pediatr. 2017;59(3):269-73.
  3. Holmes NM, Miller WL, Baskin LS. Lack of defects in androgen production in children with hypospadias. J Clin Endocrinol Metab. 2004;89(6):2811-6.
  4. Alaei M, Rohani F, Norouzi E, Hematian Boroujeni N, Tafreshi RI, Salehiniya H, Soheilipour F. The Nomogram of Clitoral Length and Width in Iranian Term and Preterm Neonates. Front Endocrinol (Lausanne). 2020;11:297.
  5. Cools M, Nordenström A, Robeva R, Hall J, Westerveld P, Flück C, et al. Caring for individuals with a difference of sex development (DSD): a Consensus Statement. Nat Rev Endocrinol. 2018;14(7):415-29.
  6. Gorduza DB, Quigley CA, Caldamone AA, Mouriquand PDE. Surgery of Anomalies of Gonadal and Genital Development in the "Post-Truth Era". Urol Clin North Am. 2018;45(4):659-69.
  7. McElreavey K, Bashamboo A. Monogenic Forms of DSD: An Update. Horm Res Paediatr. 2023;96(2):144-68.
  8. O'Connell MA, Atlas G, Ayers K, Sinclair A. Establishing a Molecular Genetic Diagnosis in Children with Differences of Sex Development: A Clinical Approach. Horm Res Paediatr. 2023;96(2):128-43.
  9. Brown J, Warne G. Practical management of the intersex infant. J Pediatr Endocrinol Metab. 2005;18(1):3-23.
  10. Dayner JE, Lee PA, Houk CP. Medical treatment of intersex: parental perspectives. J Urol. 2004;172(4 Pt 2):1762-5; discussion 5.
  11. Lightfoot S, Carley M, Brinkman W, Gardner MD, Gruppen LD, Liang N, et al. Co-creating a suite of patient decision aids for parents of an infant or young child with differences of sex development: A methods roadmap. Front Urol. 2023;2.
  12. Légaré F, Witteman HO. Shared decision making: examining key elements and barriers to adoption into routine clinical practice. Health Aff (Millwood). 2013;32(2):276-84.
  13. Sandberg DE, Gardner M, Kopec K, Urbanski M, Callens N, Keegan CE, et al. Development of a decision support tool in pediatric Differences/Disorders of Sex Development. Semin Pediatr Surg. 2019;28(5):150838.
  14. Martinez de LaPiscina I, Fluck CE. Genetics of human sexual development and related disorders. Curr Opin Pediatr. 2021;33(6):556-63.
  15. Siiteri PK, Wilson JD. Testosterone formation and metabolism during male sexual differentiation in the human embryo. J Clin Endocrinol Metab. 1974;38(1):113-25.
  16. Wilson JD, Griffin JE, Russell DW. Steroid 5 alpha-reductase 2 deficiency. Endocr Rev. 1993;14(5):577-93.
  17. Güran T. Latest Insights on the Etiology and Management of Primary Adrenal Insufficiency in Children. J Clin Res Pediatr Endocrinol. 2017;9(Suppl 2):9-22.
  18. Ali SR, Lucas-Herald A, Bryce J, Ahmed SF. The Role of International Databases in Understanding the Aetiology and Consequences of Differences/Disorders of Sex Development. Int J Mol Sci. 2019;20(18).
  19. Wiesemann C, Ude-Koeller S, Sinnecker GH, Thyen U. Ethical principles and recommendations for the medical management of differences of sex development (DSD)/intersex in children and adolescents. Eur J Pediatr. 2010;169(6):671-9.
  20. Lee PA, Nordenström A, Houk CP, Ahmed SF, Auchus R, Baratz A, et al. Global Disorders of Sex Development Update since 2006: Perceptions, Approach and Care. Horm Res Paediatr. 2016;85(3):158-80.
  21. Krishnan S, Wisniewski AB. Ambiguous genitalia in newborns. Genetic Steroid Disorders. 2014:87-97.
  22. Audi L, Ahmed SF, Krone N, Cools M, McElreavey K, Holterhus PM, et al. GENETICS IN ENDOCRINOLOGY: Approaches to molecular genetic diagnosis in the management of differences/disorders of sex development (DSD): position paper of EU COST Action BM 1303 ‘DSDnet’. Eur J Endocrinol. 2018;179(4):R197-r206.
  23. Pyle LC, Nathanson KL. A practical guide for evaluating gonadal germ cell tumor predisposition in differences of sex development. Am J Med Genet C Semin Med Genet. 2017;175(2):304-14.
  24. Yatsenko SA, Witchel SF. Genetic approach to ambiguous genitalia and disorders of sex development: What clinicians need to know. Semin Perinatol. 2017;41(4):232-43.
  25. Zenteno JC, Canto P, Kofman-Alfaro S, Mendez JP. Evidence for genetic heterogeneity in male pseudohermaphroditism due to Leydig cell hypoplasia. J Clin Endocrinol Metab. 1999;84(10):3803-6.
  26. Ogata T, Matsuo N, Hiraoka N, Hata JI. X-linked lissencephaly with ambiguous genitalia: delineation of further case. Am J Med Genet. 2000;94(2):174-6.
  27. Reardon W, Gibbons RJ, Winter RM, Baraitser M. Male pseudohermaphroditism in sibs with the alpha-thalassemia/mental retardation (ATR-X) syndrome. Am J Med Genet. 1995;55(3):285-7.
  28. Biason-Lauber A, Konrad D, Meyer M, DeBeaufort C, Schoenle EJ. Ovaries and female phenotype in a girl with 46,XY karyotype and mutations in the CBX2 gene. Am J Hum Genet. 2009;84(5):658-63.
  29. Umehara F, Tate G, Itoh K, Yamaguchi N, Douchi T, Mitsuya T, Osame M. A novel mutation of desert hedgehog in a patient with 46,XY partial gonadal dysgenesis accompanied by minifascicular neuropathy. Am J Hum Genet. 2000;67(5):1302-5.
  30. Quinonez SC, Park JM, Rabah R, Owens KM, Yashar BM, Glover TW, Keegan CE. 9p partial monosomy and disorders of sex development: review and postulation of a pathogenetic mechanism. Am J Med Genet A. 2013;161a(8):1882-96.
  31. da Silva TE, Gomes NL, Lerário AM, Keegan CE, Nishi MY, Carvalho FM, et al. Genetic Evidence of the Association of DEAH-Box Helicase 37 Defects With 46,XY Gonadal Dysgenesis Spectrum. J Clin Endocrinol Metab. 2019;104(12):5923-34.
  32. Pellegrini M, Pantano S, Lucchini F, Fumi M, Forabosco A. Emx2 developmental expression in the primordia of the reproductive and excretory systems. Anat Embryol (Berl). 1997;196(6):427-33.
  33. Lang-Muritano M, Sproll P, Wyss S, Kolly A, Hürlimann R, Konrad D, Biason-Lauber A. Early-Onset Complete Ovarian Failure and Lack of Puberty in a Woman With Mutated Estrogen Receptor β (ESR2). J Clin Endocrinol Metab. 2018;103(10):3748-56.
  34. Bagheri-Fam S, Ono M, Li L, Zhao L, Ryan J, Lai R, et al. FGFR2 mutation in 46,XY sex reversal with craniosynostosis. Hum Mol Genet. 2015;24(23):6699-710.
  35. Lourenço D, Brauner R, Rybczynska M, Nihoul-Fékété C, McElreavey K, Bashamboo A. Loss-of-function mutation in GATA4 causes anomalies of human testicular development. Proc Natl Acad Sci U S A. 2011;108(4):1597-602.
  36. Callier P, Calvel P, Matevossian A, Makrythanasis P, Bernard P, Kurosaka H, et al. Loss of function mutation in the palmitoyl-transferase HHAT leads to syndromic 46,XY disorder of sex development by impeding Hedgehog protein palmitoylation and signaling. PLoS Genet. 2014;10(5):e1004340.
  37. Kunitomo M, Khokhar A, Kresge C, Edobor-Osula F, Pletcher BA. 46,XY DSD and limb abnormalities in a female with a de novo LHX9 missense mutation. Am J Med Genet A. 2020;182(12):2887-90.
  38. Fukami M, Wada Y, Miyabayashi K, Nishino I, Hasegawa T, Nordenskjöld A, et al. CXorf6 is a causative gene for hypospadias. Nat Genet. 2006;38(12):1369-71.
  39. Pearlman A, Loke J, Le Caignec C, White S, Chin L, Friedman A, et al. Mutations in MAP3K1 cause 46,XY disorders of sex development and implicate a common signal transduction pathway in human testis determination. Am J Hum Genet. 2010;87(6):898-904.
  40. Bardoni B, Zanaria E, Guioli S, Floridia G, Worley KC, Tonini G, et al. A dosage sensitive locus at chromosome Xp21 is involved in male to female sex reversal. Nat Genet. 1994;7(4):497-501.
  41. Bashamboo A, Eozenou C, Jorgensen A, Bignon-Topalovic J, Siffroi JP, Hyon C, et al. Loss of Function of the Nuclear Receptor NR2F2, Encoding COUP-TF2, Causes Testis Development and Cardiac Defects in 46,XX Children. Am J Hum Genet. 2018;102(3):487-93.
  42. Achermann JC, Ito M, Ito M, Hindmarsh PC, Jameson JL. A mutation in the gene encoding steroidogenic factor-1 causes XY sex reversal and adrenal failure in humans. Nat Genet. 1999;22(2):125-6.
  43. Guran T, Yesil G, Turan S, Atay Z, Bozkurtlar E, Aghayev A, et al. PPP2R3C gene variants cause syndromic 46,XY gonadal dysgenesis and impaired spermatogenesis in humans. Eur J Endocrinol. 2019;180(5):291-309.
  44. Parma P, Radi O, Vidal V, Chaboissier MC, Dellambra E, Valentini S, et al. R-spondin1 is essential in sex determination, skin differentiation and malignancy. Nat Genet. 2006;38(11):1304-9.
  45. Ayers KL, Eggers S, Rollo BN, Smith KR, Davidson NM, Siddall NA, et al. Variants in SART3 cause a spliceosomopathy characterised by failure of testis development and neuronal defects. Nat Commun. 2023;14(1):3403.
  46. Sutton E, Hughes J, White S, Sekido R, Tan J, Arboleda V, et al. Identification of SOX3 as an XX male sex reversal gene in mice and humans. J Clin Invest. 2011;121(1):328-41.
  47. Portnoi MF, Dumargne MC, Rojo S, Witchel SF, Duncan AJ, Eozenou C, et al. Mutations involving the SRY-related gene SOX8 are associated with a spectrum of human reproductive anomalies. Hum Mol Genet. 2018;27(7):1228-40.
  48. Croft B, Ohnesorg T, Hewitt J, Bowles J, Quinn A, Tan J, et al. Human sex reversal is caused by duplication or deletion of core enhancers upstream of SOX9. Nat Commun. 2018;9(1):5319.
  49. Falah N, Posey JE, Thorson W, Benke P, Tekin M, Tarshish B, et al. 22q11.2q13 duplication including SOX10 causes sex-reversal and peripheral demyelinating neuropathy, central dysmyelinating leukodystrophy, Waardenburg syndrome, and Hirschsprung disease. Am J Med Genet A. 2017;173(4):1066-70.
  50. Ahmad A, Siddiqui MA, Goyal A, Wangnoo SK. Is 46XX karyotype always a female? BMJ Case Rep. 2012;2012.
  51. Jäger RJ, Anvret M, Hall K, Scherer G. A human XY female with a frame shift mutation in the candidate testis-determining gene SRY. Nature. 1990;348(6300):452-4.
  52. Puffenberger EG, Hu-Lince D, Parod JM, Craig DW, Dobrin SE, Conway AR, et al. Mapping of sudden infant death with dysgenesis of the testes syndrome (SIDDT) by a SNP genome scan and identification of TSPYL loss of function. Proc Natl Acad Sci U S A. 2004;101(32):11689-94.
  53. Eozenou C, Gonen N, Touzon MS, Jorgensen A, Yatsenko SA, Fusee L, et al. Testis formation in XX individuals resulting from novel pathogenic variants in Wilms' tumor 1 (WT1) gene. Proc Natl Acad Sci U S A. 2020;117(24):13680-8.
  54. Hammes A, Guo JK, Lutsch G, Leheste JR, Landrock D, Ziegler U, et al. Two splice variants of the Wilms' tumor 1 gene have distinct functions during sex determination and nephron formation. Cell. 2001;106(3):319-29.
  55. Mandel H, Shemer R, Borochowitz ZU, Okopnik M, Knopf C, Indelman M, et al. SERKAL syndrome: an autosomal-recessive disorder caused by a loss-of-function mutation in WNT4. Am J Hum Genet. 2008;82(1):39-47.
  56. Biason-Lauber A, Konrad D, Navratil F, Schoenle EJ. A WNT4 mutation associated with Müllerian-duct regression and virilization in a 46,XX woman. N Engl J Med. 2004;351(8):792-8.
  57. Bashamboo A, Brauner R, Bignon-Topalovic J, Lortat-Jacob S, Karageorgou V, Lourenco D, et al. Mutations in the FOG2/ZFPM2 gene are associated with anomalies of human testis determination. Hum Mol Genet. 2014;23(14):3657-65.
  58. Harris A, Siggers P, Corrochano S, Warr N, Sagar D, Grimes DT, et al. ZNRF3 functions in mammalian sex determination by inhibiting canonical WNT signaling. Proc Natl Acad Sci U S A. 2018;115(21):5474-9.
  59. Miller WL. MECHANISMS IN ENDOCRINOLOGY: Rare defects in adrenal steroidogenesis. Eur J Endocrinol. 2018;179(3):R125-r41.
  60. Flück CE, Meyer-Böni M, Pandey AV, Kempná P, Miller WL, Schoenle EJ, Biason-Lauber A. Why boys will be boys: two pathways of fetal testicular androgen biosynthesis are needed for male sexual differentiation. Am J Hum Genet. 2011;89(2):201-18.
  61. Flück CE, Pandey AV. Steroidogenesis of the testis -- new genes and pathways. Ann Endocrinol (Paris). 2014;75(2):40-7.
  62. Kok RC, Timmerman MA, Wolffenbuttel KP, Drop SL, de Jong FH. Isolated 17,20-lyase deficiency due to the cytochrome b5 mutation W27X. J Clin Endocrinol Metab. 2010;95(3):994-9.
  63. Khattab A, Yuen T, Yau M, Domenice S, Frade Costa EM, Diya K, et al. Pitfalls in hormonal diagnosis of 17-beta hydroxysteroid dehydrogenase III deficiency. J Pediatr Endocrinol Metab. 2015;28(5-6):623-8.
  64. Burkhard FZ, Parween S, Udhane SS, Flück CE, Pandey AV. P450 Oxidoreductase deficiency: Analysis of mutations and polymorphisms. J Steroid Biochem Mol Biol. 2017;165(Pt A):38-50.
  65. Miller WL, Huang N, Pandey AV, Flück CE, Agrawal V. P450 oxidoreductase deficiency: a new disorder of steroidogenesis. Ann N Y Acad Sci. 2005;1061:100-8.
  66. Miller WL, Huang N, Flück CE, Pandey AV. P450 oxidoreductase deficiency. Lancet. 2004;364(9446):1663.
  67. Walsh PC, Madden JD, Harrod MJ, Goldstein JL, MacDonald PC, Wilson JD. Familial incomplete male pseudohermaphroditism, type 2. Decreased dihydrotestosterone formation in pseudovaginal perineoscrotal hypospadias. N Engl J Med. 1974;291(18):944-9.
  68. Imperato-McGinley J, Zhu YS. Androgens and male physiology the syndrome of 5alpha-reductase-2 deficiency. Mol Cell Endocrinol. 2002;198(1-2):51-9.
  69. Kang HJ, Imperato-McGinley J, Zhu YS, Rosenwaks Z. The effect of 5α-reductase-2 deficiency on human fertility. Fertil Steril. 2014;101(2):310-6.
  70. Nordenskjöld A, Ivarsson SA. Molecular characterization of 5 alpha-reductase type 2 deficiency and fertility in a Swedish family. J Clin Endocrinol Metab. 1998;83(9):3236-8.
  71. Bertelloni S, Baldinotti F, Baroncelli GI, Caligo MA, Peroni D. Paternity in 5α-Reductase-2 Deficiency: Report of Two Brothers with Spontaneous or Assisted Fertility and Literature Review. Sex Dev. 2019;13(2):55-9.
  72. Mendonca BB, Batista RL, Domenice S, Costa EM, Arnhold IJ, Russell DW, Wilson JD. Steroid 5α-reductase 2 deficiency. J Steroid Biochem Mol Biol. 2016;163:206-11.
  73. Hornig NC, Holterhus PM. Molecular basis of androgen insensitivity syndromes. Mol Cell Endocrinol. 2021;523:111146.
  74. Batista RL, Costa EMF, Rodrigues AS, Gomes NL, Faria JA, Jr., Nishi MY, et al. Androgen insensitivity syndrome: a review. Arch Endocrinol Metab. 2018;62(2):227-35.
  75. Shukla GC, Plaga AR, Shankar E, Gupta S. Androgen receptor-related diseases: what do we know? Andrology. 2016;4(3):366-81.
  76. Zhou ZX, Wong CI, Sar M, Wilson EM. The androgen receptor: an overview. Recent Prog Horm Res. 1994;49:249-74.
  77. The androgen receptor gene mutation world wide web server[Available from: http://androgendb.mcgill.ca/.
  78. Gottlieb B TM. Androgen Insensitivity Syndrome. 1999 Mar 24 [Updated 2017 May 11]In: GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993-2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK1429/.
  79. Gottlieb B, Beitel LK, Nadarajah A, Paliouras M, Trifiro M. The androgen receptor gene mutations database: 2012 update. Hum Mutat. 2012;33(5):887-94.
  80. Ahmed SF, Bashamboo A, Lucas-Herald A, McElreavey K. Understanding the genetic aetiology in patients with XY DSD. Br Med Bull. 2013;106:67-89.
  81. Hornig NC, Ukat M, Schweikert HU, Hiort O, Werner R, Drop SL, et al. Identification of an AR Mutation-Negative Class of Androgen Insensitivity by Determining Endogenous AR Activity. J Clin Endocrinol Metab. 2016;101(11):4468-77.
  82. Hornig NC, Rodens P, Dörr H, Hubner NC, Kulle AE, Schweikert HU, et al. Epigenetic Repression of Androgen Receptor Transcription in Mutation-Negative Androgen Insensitivity Syndrome (AIS Type II). J Clin Endocrinol Metab. 2018;103(12):4617-27.
  83. Knerr J, Werner R, Schwan C, Wang H, Gebhardt P, Grötsch H, et al. Formin-mediated nuclear actin at androgen receptors promotes transcription. Nature. 2023;617(7961):616-22.
  84. Mongan NP, Tadokoro-Cuccaro R, Bunch T, Hughes IA. Androgen insensitivity syndrome. Best Pract Res Clin Endocrinol Metab. 2015;29(4):569-80.
  85. Hughes IA, Werner R, Bunch T, Hiort O. Androgen insensitivity syndrome. Semin Reprod Med. 2012;30(5):432-42.
  86. El-Maouche D, Arlt W, Merke DP. Congenital adrenal hyperplasia. Lancet. 2017;390(10108):2194-210.
  87. Morel Y, Roucher F, Plotton I, Simard J, Coll M. 3β-hydroxysteroid dehydrogenase deficiency.Genetic steroid disorders: Elsevier; 2014. p. 99-110.
  88. White PC. Genetic Steroid Disorders: Chapter 3D. Steroid 11β-Hydroxylase Deficiency and Related Disorders: Elsevier Inc. Chapters; 2013.
  89. Khattab A, Haider S, Kumar A, Dhawan S, Alam D, Romero R, et al. Clinical, genetic, and structural basis of congenital adrenal hyperplasia due to 11β-hydroxylase deficiency. Proc Natl Acad Sci U S A. 2017;114(10):E1933-e40.
  90. Khattab A, Marshall I. Management of congenital adrenal hyperplasia: beyond conventional glucocorticoid therapy. Curr Opin Pediatr. 2019;31(4):550-4.
  91. Yau M, Gujral J, New MI. Congenital Adrenal Hyperplasia: Diagnosis and Emergency Treatment. In: Feingold KR, Anawalt B, Blackman MR, Boyce A, Chrousos G, Corpas E, et al., editors. Endotext. South Dartmouth (MA): MDText.com, Inc. Copyright © 2000-2023, MDText.com, Inc.; 2000.
  92. Goto M, Piper Hanley K, Marcos J, Wood PJ, Wright S, Postle AD, et al. In humans, early cortisol biosynthesis provides a mechanism to safeguard female sexual development. J Clin Invest. 2006;116(4):953-60.
  93. Harada N. Genetic analysis of human placental aromatase deficiency. J Steroid Biochem Mol Biol. 1993;44(4-6):331-40.
  94. Wilkins L, Jones HW, Jr., Holman GH, Stempfel RS, Jr. Masculinization of the female fetus associated with administration of oral and intramuscular progestins during gestation: non-adrenal female pseudohermaphrodism. J Clin Endocrinol Metab. 1958;18(6):559-85.
  95. Troisi R, Hatch EE, Palmer JR, Titus L, Robboy SJ, Strohsnitter WC, et al. Prenatal diethylstilbestrol exposure and high-grade squamous cell neoplasia of the lower genital tract. Am J Obstet Gynecol. 2016;215(3):322.e1-8.
  96. Stillman RJ. In utero exposure to diethylstilbestrol: adverse effects on the reproductive tract and reproductive performance and male and female offspring. Am J Obstet Gynecol. 1982;142(7):905-21.
  97. Kalfa N, Philibert P, Baskin LS, Sultan C. Hypospadias: interactions between environment and genetics. Mol Cell Endocrinol. 2011;335(2):89-95.
  98. Jorgensen A, Svingen T, Miles H, Chetty T, Stukenborg JB, Mitchell RT. Environmental Impacts on Male Reproductive Development: Lessons from Experimental Models. Horm Res Paediatr. 2023;96(2):190-206.
  99. Hurtado-Gonzalez P, Mitchell RT. Analgesic use in pregnancy and male reproductive development. Curr Opin Endocrinol Diabetes Obes. 2017;24(3):225-32.
  100. Giordano J, Prior HM, Bamforth JS, Walter MA. Genetic study of SOX9 in a case of campomelic dysplasia. Am J Med Genet. 2001;98(2):176-81.
  101. Granata T, Freri E, Caccia C, Setola V, Taroni F, Battaglia G. Schizencephaly: clinical spectrum, epilepsy, and pathogenesis. J Child Neurol. 2005;20(4):313-8.
  102. Kremen J, Chan YM. Genetic evaluation of disorders of sex development: current practice and novel gene discovery. Curr Opin Endocrinol Diabetes Obes. 2019;26(1):54-9.
  103. Sofatzis JA, Alexacos L, Skouteli HN, Tiniakos G, Padiatellis C. Malformed female genitalia in newborns with the VATER association. Acta Paediatr Scand. 1983;72(6):923-4.
  104. Ahmed SF, Achermann JC, Arlt W, Balen A, Conway G, Edwards Z, et al. Society for Endocrinology UK guidance on the initial evaluation of an infant or an adolescent with a suspected disorder of sex development (Revised 2015). Clin Endocrinol (Oxf). 2016;84(5):771-88.
  105. Davies JH, Cheetham T. Recognition and assessment of atypical and ambiguous genitalia in the newborn. Arch Dis Child. 2017;102(10):968-74.
  106. Baidya A, Basu AK, Bhattacharjee R, Biswas D, Biswas K, Chakraborty PP, et al. Diagnostic approach in 46, XY DSD: an endocrine society of bengal (ESB) consensus statement. J Pediatr Endocrinol Metab. 2023;36(1):4-18.
  107. Bever YV, Brüggenwirth HT, Wolffenbuttel KP, Dessens AB, Groenenberg IAL, Knapen M, et al. Under-reported aspects of diagnosis and treatment addressed in the Dutch-Flemish guideline for comprehensive diagnostics in disorders/differences of sex development. J Med Genet. 2020;57(9):581-9.
  108. Prader A. [Genital findings in the female pseudo-hermaphroditism of the congenital adrenogenital syndrome; morphology, frequency, development and heredity of the different genital forms]. Helv Paediatr Acta. 1954;9(3):231-48.
  109. Quigley CA, De Bellis A, Marschke KB, el-Awady MK, Wilson EM, French FS. Androgen receptor defects: historical, clinical, and molecular perspectives. Endocr Rev. 1995;16(3):271-321.
  110. van der Straaten S, Springer A, Zecic A, Hebenstreit D, Tonnhofer U, Gawlik A, et al. The External Genitalia Score (EGS): A European Multicenter Validation Study. J Clin Endocrinol Metab. 2020;105(3).
  111. Cao W, Ding X, Dong Z, Tang H. Reference Values for and Correlation Analysis of the Anogenital Distance of Chinese Han Full-Term Singleton Neonates. Front Pediatr. 2022;10:905421.
  112. Vanderbrink BA, Rink RC, Cain MP, Kaefer M, Meldrum KK, Misseri R, Karmazyn B. Does preoperative genitography in congenital adrenal hyperplasia cases affect surgical approach to feminizing genitoplasty? J Urol. 2010;184(4 Suppl):1793-8.
  113. Eggers S, Sadedin S, van den Bergen JA, Robevska G, Ohnesorg T, Hewitt J, et al. Disorders of sex development: insights from targeted gene sequencing of a large international patient cohort. Genome Biol. 2016;17(1):243.
  114. Baxter RM, Arboleda VA, Lee H, Barseghyan H, Adam MP, Fechner PY, et al. Exome sequencing for the diagnosis of 46,XY disorders of sex development. J Clin Endocrinol Metab. 2015;100(2):E333-44.
  115. Bacila IA, Lawrence NR, Badrinath SG, Balagamage C, Krone NP. Biomarkers in congenital adrenal hyperplasia. Clin Endocrinol (Oxf). 2023.
  116. Ye L, Zhao Z, Ren H, Wang W, Zhou W, Zheng S, et al. A Multiclassifier System to Identify and Subtype Congenital Adrenal Hyperplasia Based on Circulating Steroid Hormones. J Clin Endocrinol Metab. 2022;107(8):e3304-e12.
  117. Kamrath C, Friedrich C, Hartmann MF, Wudy SA. Metabotypes of congenital adrenal hyperplasia in infants determined by gas chromatography-mass spectrometry in spot urine. J Steroid Biochem Mol Biol. 2023;231:106304.
  118. Jha S, Turcu AF, Sinaii N, Brookner B, Auchus RJ, Merke DP. 11-Oxygenated Androgens Useful in the Setting of Discrepant Conventional Biomarkers in 21-Hydroxylase Deficiency. J Endocr Soc. 2021;5(2):bvaa192.
  119. Bileck A, Fluck CE, Dhayat N, Groessl M. How high-resolution techniques enable reliable steroid identification and quantification. J Steroid Biochem Mol Biol. 2019;186:74-8.
  120. Dhayat NA, Frey AC, Frey BM, d'Uscio CH, Vogt B, Rousson V, et al. Estimation of reference curves for the urinary steroid metabolome in the first year of life in healthy children: Tracing the complexity of human postnatal steroidogenesis. J Steroid Biochem Mol Biol. 2015;154:226-36.
  121. Enver EO, Vatansever P, Guran O, Bilgin L, Boran P, Turan S, et al. Adrenal steroids reference ranges in infancy determined by LC-MS/MS. Pediatr Res. 2022;92(1):265-74.
  122. Guibourdenche J, Lucidarme N, Chevenne D, Rigal O, Nicolas M, Luton D, et al. Anti-Müllerian hormone levels in serum from human foetuses and children: pattern and clinical interest. Mol Cell Endocrinol. 2003;211(1-2):55-63.
  123. Schmidt H, Schwarz HP. Serum concentrations of LH and FSH in the healthy newborn. Eur J Endocrinol. 2000;143(2):213-5.
  124. Johannsen TH, Main KM, Ljubicic ML, Jensen TK, Andersen HR, Andersen MS, et al. Sex Differences in Reproductive Hormones During Mini-Puberty in Infants With Normal and Disordered Sex Development. J Clin Endocrinol Metab. 2018;103(8):3028-37.
  125. Bertelloni S, Russo G, Baroncelli GI. Human Chorionic Gonadotropin Test: Old Uncertainties, New Perspectives, and Value in 46,XY Disorders of Sex Development. Sex Dev. 2018;12(1-3):41-9.
  126. Gomes NL, Batista RL, Nishi MY, Lerário AM, Silva TE, de Moraes Narcizo A, et al. Contribution of Clinical and Genetic Approaches for Diagnosing 209 Index Cases With 46,XY Differences of Sex Development. J Clin Endocrinol Metab. 2022;107(5):e1797-e806.
  127. Guerrero-Fernández J, González-Peramato P, Rodríguez Estévez A, Alcázar Villar MJ, Audí Parera L, Azcona San Julián MC, et al. Consensus guide on prophylactic gonadectomy in different sex development. Endocrinol Diabetes Nutr (Engl Ed). 2022;69(8):629-45.
  128. Patel V, Casey RK, Gomez-Lobo V. Timing of Gonadectomy in Patients with Complete Androgen Insensitivity Syndrome-Current Recommendations and Future Directions. J Pediatr Adolesc Gynecol. 2016;29(4):320-5.
  129. Sircili MH, e Silva FA, Costa EM, Brito VN, Arnhold IJ, Dénes FT, et al. Long-term surgical outcome of masculinizing genitoplasty in large cohort of patients with disorders of sex development. J Urol. 2010;184(3):1122-7.
  130. Yan M, Dilihuma J, Luo Y, Reyilanmu B, Shen Y, Mireguli M. Novel Compound Heterozygous Variants in the LHCGR Gene in a Genetically Male Patient with Female External Genitalia. J Clin Res Pediatr Endocrinol. 2019;11(2):211-7.
  131. Berg JS, French SL, McCullough LB, Kleppe S, Sutton VR, Gunn SK, Karaviti LP. Ethical and legal implications of genetic testing in androgen insensitivity syndrome. J Pediatr. 2007;150(4):434-8.
  132. Wisniewski AB. Complete Androgen Insensitivity Syndrome: Long-Term Medical, Surgical, and Psychosexual Outcome. Journal of Clinical Endocrinology &amp; Metabolism. 2000;85(8):2664-9.
  133. Bianco S, Agrifoglio V, Mannino F, Cefalù E, Cittadini E. Successful pregnancy in a pure gonadal dysgenesis with karyotype 46,XY patient (Swyer's syndrome) following oocyte donation and hormonal treatment. Acta Eur Fertil. 1992;23(1):37-8.
  134. Hines M, Ahmed SF, Hughes IA. Archives of Sexual Behavior. 2003;32(2):93-101.
  135. Park S, Ha SH, Kim KS. Long-term follow-up after feminizing genital reconstruction in patients with ambiguous genitalia and high vaginal confluence. Journal of Korean medical science. 2011;26(3):399-403.
  136. Sircili MHP, de Queiroz e Silva FA, Costa EMF, Brito VN, Arnhold IJP, Dénes FT, et al. Long-Term Surgical Outcome of Masculinizing Genitoplasty in Large Cohort of Patients With Disorders of Sex Development. Journal of Urology. 2010;184(3):1122-7.
  137. Wisniewski AB, Migeon CJ, Gearhart JP, Rock JA, Berkovitz GD, Plotnick LP, et al. Congenital Micropenis: Long-Term Medical, Surgical and Psychosexual Follow-Up of Individuals Raised Male or Female. Hormone Research in Paediatrics. 2001;56(1-2):3-11.
  138. Mendonca BB, Costa EM, Belgorosky A, Rivarola MA, Domenice S. 46,XY DSD due to impaired androgen production. Best Pract Res Clin Endocrinol Metab. 2010;24(2):243-62.
  139. Batista RL, Mendonca BB. Integrative and Analytical Review of the 5-Alpha-Reductase Type 2 Deficiency Worldwide. Appl Clin Genet. 2020;13:83-96.
  140. Bonnet E, Winter M, Mallet D, Plotton I, Bouvattier C, Cartigny M, et al. Changes in the clinical management of 5α-reductase type 2 and 17β-hydroxysteroid dehydrogenase type 3 deficiencies in France. Endocr Connect. 2023;12(3).
  141. Duranteau L, Rapp M, van de Grift TC, Hirschberg AL, Nordenskjöld A. Participant- and Clinician-Reported Long-Term Outcomes After Surgery in Individuals with Complete Androgen Insensitivity Syndrome. J Pediatr Adolesc Gynecol. 2021;34(2):168-75.
  142. Dessens AB, Slijper FME, Drop SLS. Gender Dysphoria and Gender Change in Chromosomal Females with Congenital Adrenal Hyperplasia. Archives of Sexual Behavior. 2005;34(4):389-97.
  143. Long DN, Wisniewski AB, Migeon CJ. Gender Role Across Development in Adult Women with Congenital Adrenal Hyperplasia Due to 21-Hydroxylase Deficiency. Journal of Pediatric Endocrinology and Metabolism. 2004;17(10).
  144. Wisniewski AB, Migeon CJ, Malouf MA, Gearhart JP. PSYCHOSEXUAL OUTCOME IN WOMEN AFFECTED BY CONGENITAL ADRENAL HYPERPLASIA DUE TO 21-HYDROXYLASE DEFICIENCY. Journal of Urology. 2004;171(6 Part 1):2497-501.
  145. Lee PA, Houk CP. Outcome Studies Among Men with Micropenis. Journal of Pediatric Endocrinology and Metabolism. 2004;17(8).
  146. Lee PA, Houk CP, Husmann DA. Should Male Gender Assignment be Considered in the Markedly Virilized Patient With 46,XX and Congenital Adrenal Hyperplasia? Journal of Urology. 2010;184(4S):1786-92.
  147. de Jesus LE, Costa EC, Dekermacher S. Gender dysphoria and XX congenital adrenal hyperplasia: how frequent is it? Is male-sex rearing a good idea? Journal of Pediatric Surgery. 2019;54(11):2421-7.
  148. Crissman HP, Warner L, Gardner M, Carr M, Schast A, Quittner AL, et al. Children with disorders of sex development: A qualitative study of early parental experience. International journal of pediatric endocrinology. 2011;2011(1):10-.
  149. Yau M, Khattab A, New MI. Prenatal Diagnosis of Congenital Adrenal Hyperplasia. Endocrinology and Metabolism Clinics of North America. 2016;45(2):267-81.
  150. Van't Westeinde A, Karlsson L, Messina V, Wallensteen L, Brösamle M, Dal Maso G, et al. An update on the long-term outcomes of prenatal dexamethasone treatment in congenital adrenal hyperplasia. Endocr Connect. 2023;12(4).
  151. Kazmi D, Bailey J, Yau M, Abu-Amer W, Kumar A, Low M, Yuen T. New developments in prenatal diagnosis of congenital adrenal hyperplasia. The Journal of Steroid Biochemistry and Molecular Biology. 2017;165:121-3.
  152. Maher JY, Gomez-Lobo V, Merke DP. The management of congenital adrenal hyperplasia during preconception, pregnancy, and postpartum. Rev Endocr Metab Disord. 2023;24(1):71-83.
  153. Fedele D, Kirk K, Wolfe-Christensen C, Phillips T, Mazur T, Mullins L, et al. Primary Caregivers of Children Affected by Disorders of Sex Development: Mental Health and Caregiver Characteristics in the Context of Genital Ambiguity and Genitoplasty. International Journal of Pediatric Endocrinology. 2010;2010(1):690674.
  154. Hullmann SE, Fedele DA, Wolfe-Christensen C, Mullins LL, Wisniewski AB. Differences in adjustment by child developmental stage among caregivers of children with disorders of sex development. International journal of pediatric endocrinology. 2011;2011(1):16-.
  155. Kirk KD, Fedele DA, Wolfe-Christensen C, Phillips TM, Mazur T, Mullins LL, et al. Parenting Characteristics of Female Caregivers of Children Affected by Chronic Endocrine Conditions: A Comparison Between Disorders of Sex Development and Type 1 Diabetes Mellitus. Journal of Pediatric Nursing. 2011;26(6):e29-e36.
  156. Wolfe-Christensen C, Fedele DA, Kirk K, Phillips TM, Mazur T, Mullins LL, et al. Degree of External Genital Malformation at Birth in Children with a Disorder of Sex Development and Subsequent Caregiver Distress. Journal of Urology. 2012;188(4S):1596-600.
  157. Wisniewski AB, Batista RL, Costa EMF, Finlayson C, Sircili MHP, Dénes FT, et al. Management of 46,XY Differences/Disorders of Sex Development (DSD) Throughout Life. Endocrine Reviews. 2019;40(6):1547-72.
  158. Magritte E. Working together in placing the long term interests of the child at the heart of the DSD evaluation. Journal of Pediatric Urology. 2012;8(6):571-5.

 

Fibromyalgia

ABSTRACT

 

Fibromyalgia is a clinical entity characterized by the combination of chronic widespread pain and other non-pain symptoms, including fatigue, poor sleep, and cognitive disturbances, which can exhibit symptom variation not only between different patients, but also in the same patient during the course of the disease. These symptoms are relatively common and non-specific. They can be encountered in other disorders that may overlap with fibromyalgia, often without having clear boundaries, while their nature makes them difficult to be objectively defined and quantified. These issues have led to significant controversy over the definition and the diagnostic criteria of fibromyalgia. It has been suggested that the markers of physical and psychological distress have a continuous distribution in the general population with fibromyalgia patients being at the extreme end of this continuum. Genetic predisposition in combination with environmental factors, are responsible for each individual’s position in this this distribution. In recent years more knowledge has been obtained to better understand the environmental factors that seem to be important in triggering fibromyalgia. Most of them act as stressors superimposed onto a deranged stress-response system leading to dys-regulation of the nociceptive system and the appearance of clinical symptoms. The aim of the therapy is to relieve pain and motivate the patients to become more physically active using a multimodal individualized therapeutic strategy that includes education, exercise, cognitive-behavioral approaches and medications. The response to current therapeutic modalities varies significantly, with some patients responding adequately, while others do not seem to experience any long-term benefit. 

 

INTRODUCTION

 

Fibromyalgia is a clinical entity characterized by the combination of ill-defined symptoms including chronic widespread pain, with concomitant fatigue, sleeping disorders, and cognitive disturbances (1). The severity of these symptoms can vary significantly during the course of the disease. Fibromyalgia has been described as an arbitrarily created syndrome that lies at the extreme end of the spectrum of poly-symptomatic distress (2). The term poly-symptomatic was used to emphasize the variety of multiple different symptoms that can be found in fibromyalgia patients, while the distress can have a physical and/or a psychological component. This exact nature of fibromyalgia makes it difficult to be clearly defined, often overlapping with disorders that are characterized by similar symptoms. It is important to note that fibromyalgia is not an exclusion diagnosis as it can co-exist with other clinical conditions (3). 

 

CLINICAL FEATURES

 

The main presenting complains of patients with fibromyalgia include chronic widespread pain (also called multisite pain), fatigue, and poor sleep. Usually the pain is initially localized, but eventually it involves many muscle groups. It is characterized as persistent with varying intensity, while it can often be described as a sensation of burning, gnawing soreness, stiffness, or aching. Excessive sensitivity to normally painful stimuli, such as pressure or heat (hyperalgesia) and painful sensation to normally non-painful stimuli, such as touch (allodynia) are significant features of fibromyalgia. Often patients complain of swollen joints and paresthesias without though the presence of any objective clinical findings during physical examination. Pain is often aggravated by cold and humid weather, poor sleep, physical and mental stress. Additionally the patients may have a variety of less well understood pain symptoms, including abdominal pain, chest wall pain, symptoms suggestive of irritable bowel syndrome, pelvic pain, and bladder symptoms of frequency and urgency suggestive of interstitial cystitis (4–9).

 

Fatigue is present in almost all patients with fibromyalgia, while many complain of non-refreshing sleep, frequent awakening during the night, and difficulty falling back to sleep. Sleep apnea and nocturnal myoclonus can also be present along with a sensation of light-headedness, dizziness, and faintness. In addition, cognitive difficulties such as short-term memory loss, groping for words and poor vocabulary, are common among patients with fibromyalgia. Mood disturbances, including depression, anxiety and heightened somatic concern, may often also occur. Headaches, either muscular or migraine type, are also commonly present (6,7). Other often co-existing conditions include multiple chemical sensitivity, “allergic” symptoms, ocular dryness, palpitations, dyspnea, vulvodynia, dysmenorrhea, premenstrual syndrome, sexual dysfunction, weight fluctuations, night sweats, dysphagia, restless leg syndrome, temporomandibular joint pain, chronic fatigue syndrome (systemic exertion intolerance disease), Raynaud`s phenomenon, autonomic dysfunction,  and dysgeusia (6,8,9). These conditions cannot be used to support the diagnosis of fibromyalgia.

 

Approximately 40% of fibromyalgia patients have accompanying depression at the time of diagnosis, while 60% of patients have a lifetime history of depression. In addition, an anxiety disorder is present in 30% of the cases at the time of diagnosis while the lifetime prevalence of an anxiety disorder in fibromyalgia patients is approximately 60% (10–14). The levels of depression and anxiety in patients with fibromyalgia seem to be associated with the degree of cognitive impairment, as shown in a meta-analysis of 23 case-control studies (15). Based on the coexistence of depression and anxiety, fibromyalgia patients can be divided into 2 major groups. The first group comprises of patients without coexisting mood disorders, while the second of patients with concomitant depressive mood, often in combination with anxiety. According to the results of a study that intended to subgroup fibromyalgia patients based on: 1) mood status (evaluated by the Center for Epidemiologic Studies Depression Scale for depression and the State-Trait Personality Inventory for symptoms of trait-related anxiety), 2) cognition (by the catastrophizing and control of pain subscales of the Coping Strategies Questionnaire), and 3) hyperalgesia/tenderness (by dolorimetry and random pressure-pain applied at suprathreshold values), it was noted that fibromyalgia patients with depressive mood and anxiety are also ‘catastrophizing’. This term is used to indicate that such patients have a very negative, pessimistic view of what their pain is and what is causing, while they have no sense that they can control their pain. On the contrary fibromyalgia patients who are neither depressed nor anxious and therefore do not catastrophize, have a moderate sense that they can control their pain. These patients can be further divided into 2 subgroups based on the degree of hyperalgesia/tenderness, the first subgroup comprises of patients with high hyperalgesia/tenderness, while the second one of patients with moderate hyperalgesia/tenderness (11). In addition, it has been proposed that depressed fibromyalgia patients can also be divided into 2 subgroups, in the first one depression is a co-morbid condition, while in the second depression is the cause of fibromyalgia (16). All these fibromyalgia subgroups are illustrated in Figure 1.

 

Figure 1. Subgroups of fibromyalgia patients.

 

In another study fibromyalgia patients were classified as dysfunctional, inter-personally distressed, or adaptive copers, based on their responses to the Multidimensional Pain Inventory. The dysfunctional patients experienced more pain behaviors and overt expressions of pain, distress, and suffering, such as slowed movement, bracing, limping, and grimacing compared to the inter-personally distressed or the adaptive copers (17).

 

It is of interest that up to  25% of patients correctly diagnosed with a systemic rheumatic disease (e.g. rheumatoid arthritis, systemic lupus erythematosus) will also fulfill the classification criteria for fibromyalgia (18). This is also the case for many patients who experience persistent various forms of pain (including widespread myalgias, arthralgias, and headache), fatigue, neurocognitive dysfunction, and sleep disturbances after an infection with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) that caused a mild to moderate coronavirus disease 2019 (long COVID) (19,20). The most commonly encountered comorbid conditions in fibromyalgia patients are shown in Table 1.

 

-    Table 1. The Most Commonly Encountered Co-Morbid Conditions in Fibromyalgia

Sleep disorders

-    Non restorative sleep (alpha-delta sleep anomaly)

-    Sleep apnea

-    Restless leg syndrome

-    Nocturnal myoclonus

Chronic fatigue syndrome (systemic exertion intolerance disease)

 

Psychiatric disorders

 

-    Anxiety disorders

-    Depression

-    Obsessive compulsive disorder

Headache

-    Tension type headache

-    Migraine

Irritable bowel syndrome

 

Musculofascial pain syndrome

-    Temporomandibular disorders

-    Interstitial cystitis

Dysmenorrhea

 

Premenstrual syndrome

 

Non-cardiac chest pain

 

Raynaud’s phenomenon

 

Systemic autoimmune diseases

-    Rheumatoid arthritis

-    Systematic lupus erythematosus

-    Sjögren’s syndrome

-    Ankylosing spondylitis and other seronegative spondylarthritis

-    Polymyalgia rheumatica

Long COVID

 

 

ASSESSMENT

 

Fibromyalgia is a chronic illness, with a variety of symptoms that can change during the course of the disease and after treatment. Therefore, its core symptoms should be evaluated both in clinical practice and in treatment trials. A working group within OMERACT (Outcome Measures in Rheumatology) reached a consensus regarding the domains that need to be assessed in clinical trials for fibromyalgia, using Delphi exercises within patients and expert clinicians (21). The fibromyalgia core symptom domains include pain intensity, tenderness, fatigue, sleep disturbance, multidimensional function (including health related quality of life and physical function), patient global impression of change, cognitive dysfunction, and depression. However at the present time, there is no consensus on how to evaluate these domains, in order to quantify fibromyalgia disease activity state and/or response (22).

 

Pain intensity can be assessed using visual analog scales. It has been proposed to use the wording “please rate your pain by circling on the number that best describes your pain on average” and has anchors that vary from “no pain” to “pain as bad as you can imagine” (23).

 

Tenderness can be measured by evaluating the alteration of the severity of pain at the tender points, using visual or analog scales, but not by the change in their number. The change in the number of tender points is poorly correlated with improvement in fibromyalgia treatment trials. It has been noted that the tender points measure the combination of tenderness and distress an individual has, rendering them inadequate for the evaluation of tenderness per se (24).

 

Fatigue can be evaluated by the Fatigue Severity Scale (FSS), which measures the functional outcomes related to fatigue (25). It has been shown that FSS has the most robust psychometric properties of 19 reviewed fatigue measures, while it had the best ability to act as an outcome measure sensitive to change with treatment, in chronically ill patients (26). Other instruments that have been used to assess fatigue in fibromyalgia patients include the Multidimensional Fatigue Index and the Multidimensional Assessment of Fatigue.

 

Sleep disturbance in fibromyalgia patients has been evaluated by the Medical Outcome Studies (MOS) sleep scale, the Functional Outcomes of Sleep Questionnaire (FOSQ), and the Jenkins Sleep Scale (JSS). Of these instruments MOS sleep scale lacks validity to assess changes in sleep symptoms in fibromyalgia treatment trials, FOSQ has not been adequately validated in fibromyalgia patients, while JSS has been criticized for possible high-recall bias, since it requires the patients to rate the frequency of their symptoms over a period of a month (23).

 

Multidimensional function, including health related quality of life and physical function, can be evaluated using the fibromyalgia impact questionnaire (FIQ). This represents a useful tool in assessing functional abilities in daily life and measures patient status, progress and outcomes. FIQ is self-administered and is highly sensitive to changes during the course of the disease. However, its functional items are orientated toward high levels of disability, resulting in a possible floor effect, while its physical function items are addressed to women living in affluent countries, generating gender and ethnic bias. To address these issues the Revised Fibromyalgia Impact Questionnaire (FIQR) was developed, having modified physical function questions and including questions on memory, tenderness, balance and environmental sensitivity, while keeping questions that evaluate overall impact and symptoms (27). Other tools for assessing overall function and quality of life include the Health Assessment Questionnaire, the Symptom Interpretation Questionnaire, the Western Ontario and McMaster Universities Osteoarthritis Index, the Patient Global Impression of Change scale (PGIC), and psychometric scales (28).

 

Patient global impression of change can be evaluated by the Patient Global Impression of Change scale (PGIC), as recommended by the Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (IMMPACT) (29).

 

Cognitive dysfunction can be evaluated by the Multiple Ability Self-report Questionnaire (MASQ), which is a self-report questionnaire measuring language, visuoperception, verbal memory and attention (30). However self-assessment is poorly correlated with objective measures of cognitive function and has poor discriminating ability for patients with mild cognitive impairment (31).

 

Depression can be evaluated using the Hospital Anxiety and Depression Scale (HADS) depression subscale (HADS-D). The use of the original Beck Depression Inventory (BDI) as well as the current BDI-II, should be avoided, since they tend to overestimate the presence of major depressive disorder in fibromyalgia patients by evaluating a number of non-depressive symptoms, which are often encountered in fibromyalgia (23). 

 

DIAGNOSIS

 

As already mentioned, fibromyalgia is characterized by symptoms that can vary in number and intensity not only between different patients, but also in the same patient during the course of the disease. These symptoms are also common in other disorders that can overlap with fibromyalgia, often without having clear boundaries. Additionally, the nature of the symptoms of fibromyalgia makes them difficult to be objectively defined and measured. All these issues have led to significant controversy over the definition and diagnosis of fibromyalgia. The clinical entity of fibromyalgia was first described in 1904, under the term “fibrositis”, after focusing on clinical evidence of muscle sensitivity (32). It was not until 1977 that specific criteria for the diagnosis of fibromyalgia were introduced (33). Since then a number of different criteria have been proposed, based on a combination of tender point examination and the presence of symptoms. In 1986 a committee of the American College of Rheumatology (ACR) started a multicenter study, trying to provide a definition of fibromyalgia and  establish classification criteria (34). In 2013, the Analgesic, Anesthetic, and Addiction Clinical Trial Translations Innovations Opportunities and Networks (ACTTION) public-private partnership with the US Food and Drug Administration (FDA) and American Pain Society (APS) initiated the ACTTION-APS Pain Taxonomy (AAPT) in an attempt to develop a diagnostic system that would be clinically useful and consistent across chronic pain disorders, including fibromyalgia (35).

 

In clinical practice either the 2016 revision of the 2010/2011 fibromyalgia diagnostic criteria or the AAPT criteria can be used to help physicians to diagnose fibromyalgia (36). However, these criteria should be viewed as an aid and not as a gold standard for diagnosing fibromyalgia in clinical practice. Good clinical judgment is necessary to interpret the findings of physical examination and to assess psychological factors and associated comorbidities so as to correctly identify the patients with fibromyalgia (37).

 

Relevant social, personal and family history can be helpful in establishing the diagnosis of fibromyalgia, since there is evidence that the symptoms of fibromyalgia can appear after a physical or emotional trauma, a medical illness, or asurgical operation, while a family history of fibromyalgia, makes the diagnosis of fibromyalgia more likely (38).

 

1990 ACR Classification Criteria

 

In 1990 the American College of Rheumatology (ACR) committee established criteria for the classification of fibromyalgia. According to these criteria fibromyalgia is defined as chronic widespread pain involving both sides of the body, above and below the waist, as well as the whole length of the spine, and excessive tenderness in the pressure of 11 of 18 specific muscle-tendon sites (9 pairs of tender points). The locations of the tender points are described in Figure2 and Table 2. Pressure equivalent of 4 kg/cm is applied to these points using the pulp of the thumb or the first two or three fingers. This can be accurately measured with a dolorimeter or it can be estimated, since 4 kg/cm is the pressure needed to be applied so as to whiten the examiner’s fingernail bed. These criteria specifically state that fibromyalgia is not an exclusionary diagnosis (34).

 

Figure 2. Fibromyalgia tender points.

 

 

Table 2. Description of the Location of Fibromyalgia Tender Points

Occiput:

at the insertions of one or more of the following muscles: trapezius, sternocleidomastoid, splenius capitus, semispinalis capitus

Trapezius:

at the midpoint of the upper border

Supraspinatus:

above the scapular spine near the medial border

Gluteal:

at the upper outer quadrant of the buttocks at the anterior edge of the gluteus maximus

Low cervical:

at the anterior aspect of the interspaces between the transverse processes of C5–C7

Second rib:

just lateral to the second costochondral junctions

Lateral epicondyle:

2 cm distal to the lateral epicondyle

Greater trochanter:

posterior to the greater trochanteric prominence

Knee:

at the medial fat pad proximal to the joint line

 

The tender points, which are examined in fibromyalgia, are not just areas that the patient feels pain. They are points that fibromyalgia patients are relatively more tender, compared to normal individuals, when pressure is applied on them. But fibromyalgia patients are more tender wherever you apply pressure, not only to some of these 18 specific tender points, including areas previously considered to be “control points” (39). There is evidence that these tender points are areas that everyone is generally more tender. In contrast, fibromyalgia patients are more tender not only to pressure but to other stimuli such as heat, cold and other sensory stimuli, most probably due to decreased pain threshold. The number of tender points an individual has is highly correlated with distress, as defined by the presence of anxiety, depression, sleep disturbance, fatigue and global severity. Tender points have been described as “a sedimentation rate of distress”. Consequently tender points measure the combination of tenderness and distress an individual has (24).

 

The use of the 1990 ACR classification criteria in clinical practice is surrounded by substantial controversy. Tender points were introduced as an objective physical finding. However, if the physician who performs the physical examination is not experienced enough, tender point counting is impossible to be performed accurately. Most of the physicians examining fibromyalgia patients did not know how to carry out the tender point examination. Consequently, tender point count was not routinely performed, and when performed it was performed inaccurately (40). Another shortcoming of the tender point counting is that it is not as objective as it was initially considered, since the physician can be biased by the patient’s interview that precedes the physical examination. These issues lead to a low inter-examiner reliability of the tender point count (41). 

 

Other more sophisticated measures of assessing tenderness, such as applying stimuli randomly, when the individual cannot anticipate what the next stimulus is going to be, are equally abnormal in fibromyalgia patients, but do not correlate with distress (39). These methods require special training and are more time consuming than the trigger point count. Other alternative assessment methods include functional magnetic resonance imaging (fMRI) and nociceptive flexion reflex (NFR) testing, which documents abnormal pain processing in fibromyalgia. Functional MRI demonstrates similar brain activation in regions involved in pain processing in fibromyalgia patients and normal individuals. However fibromyalgia patients have increased pain sensitivity and brain activation during comparable stimulus (42). Nociceptive flexion reflexes are sensory-motor responses elicited by electrical noxious stimuli, which involve activation of spinal and supraspinal neuronal circuits, providing an objective and quantitative assessment of the function of the pain-controlsystem. It has been demonstrated that the NFR threshold in patients with fibromyalgia is significantly decreased compared with that in controls (43). Of these methods fMRI is expensive and complex compared to NFR testing that appears to be more easily accessible and convenient, since standard electromyographic equipment can be used. This test also seems to eliminate subjective bias and dissimulation (44).

 

The 1990 ACR classification criteria, define fibromyalgia in terms of pain rather than its other features. However, patients with fibromyalgia apart from tenderness and pain, also have a number of other somatic symptoms. Although non-pain symptoms are important, there is no evidence to support the notion that they are more important than hyperalgesia and allodynia, which are key symptoms of fibromyalgia (44). Many clinicians with experience in fibromyalgia did not feel that the 1990 ACR classification criteria were sufficiently reliable for the diagnosis of fibromyalgia in clinical practice and were  considering other aspects of the disease in an attempt to reach a more accurate diagnosis (38).

 

2010 ACR Preliminary Diagnostic Criteria

 

To address the aforementioned issues the ACR in 2010 proposed the preliminary diagnostic criteria for fibromyalgia (Table 3), that were not meant to replace the 1990 ACR classification criteria, but to represent an alternative simple and easy method of diagnosis in clinical practice (45). These diagnostic criteria do not require a tender point count. Instead, they rely only on symptoms for the diagnosis of fibromyalgia. They introduced the widespread pain index (WPI), which counts the areas that the patient feels pain during one week preceding the examination, and the symptom severity (SS) scale, which describes the severity of fatigue, unrefreshing sleep, cognitive problems, and a number of associated somatic fibromyalgia symptoms. These symptoms need to be assessed and rated by a physician, therefore the 2010 ACR preliminary diagnostic criteria are in-adequate for patient self-diagnosis.

 

Two more conditions need to be fulfilled so as to diagnose fibromyalgia. The symptoms need to be present at a similar level for at least 3 months while alternate disorders that would otherwise explain the pain need to be excluded (Table 3). The authors of the 2010 ACR preliminary diagnostic criteria have clarified that the latter condition does not mean that fibromyalgia is an exclusion diagnosis according to these criteria. The diagnosis of fibromyalgia should not be made only when there is not another disease that could explain the pain that would otherwise be attributed to fibromyalgia. It should be noted that rheumatic diseases usually do not cause pain that can be confused with fibromyalgia (3).

 

Table 3. 2010 ACR Preliminary Diagnostic Criteria

Criteria:

A patient satisfies diagnostic criteria for fibromyalgia if the following 3 conditions are met:

1)    Widespread pain index (WPI) ≥7 and symptom severity (SS) scale score ≥5 or

Widespread pain index (WPI) 3-6 and symptom severity (SS) scale score ≥9.

2)    Symptoms have been present at a similar level for at least 3 months.

3)    The patient does not have a disorder that would otherwise explain the pain.

 

Ascertainment:

1)    WPI

Note the number of areas in which the patient has had pain over the last week. In how many areas has the patient had pain?

(Score will be between 0 and 19)

 

 

-Neck

-Upper arm, left

 

-Abdomen

 

-Upper leg, left

 

 

 

-Jaw, left

-Upper arm, right

 

-Upper back

 

-Upper leg, right

 

 

 

-Jaw, right

-Lower arm, left

 

-Lower back

 

-Lower leg, left

 

 

 

-Shoulder girdle, left

 

-Lower arm, right

 

-Hip (buttock, trochanter), left

 

-Lower leg, right

 

 

 

-Shoulder girdle, right

 

-Chest

 

-Hip (buttock, trochanter), right

 

 

 

 

2)    SS scale score

The SS scale score is the sum of the severity of the 3 symptoms (fatigue, waking unrefreshed, cognitive symptoms) plus the extent (severity) of somatic symptoms in general.

(The final score is between 0 and 12)

 

-For the each of the 3 symptoms below, indicate the level of severity over the past week using the following scale:

0 = no problem

1 = slight or mild problems, generally mild or intermittent

2 = moderate, considerable problems, often present and/or at a moderate level

3 = severe: pervasive, continuous, life-disturbing problems

Fatigue                     (0-3)

Waking unrefreshed (0-3)

Cognitive symptoms (0-3)

 

-Considering somatic symptoms in general, indicate whether the patient has: muscle pain, irritable bowel syndrome, fatigue/tiredness, thinking or remembering problem, muscle weakness, headache, pain/cramps in the abdomen, numbness/tingling, dizziness, insomnia, depression, constipation, pain in the upper abdomen, nausea, nervousness, chest pain, blurred vision, fever, diarrhea, dry mouth, itching, wheezing, Raynaud's phenomenon, hives/welts, ringing in ears, vomiting, heartburn, oral ulcers, loss of/change in taste, seizures, dry eyes, shortness of breath, loss of appetite, rash, sun sensitivity, hearing difficulties, easy bruising, hair loss, frequent urination, painful urination, and bladder spasms

0 = no symptoms

1 = few symptoms

2 = a moderate number of symptoms

3 = a great deal of symptoms

 

There is evidence that there is good agreement between the 1990 ACR classification criteria and the 2010 ACR preliminary diagnostic criteria (46–53). However, these criteria are expected not to agree completely, as the former are focused on the presence of tender points while the latter on the presence of symptoms. The 1990 criteria can diagnose fibromyalgia in patients who do not have sufficiently high symptom score according to the 2010 criteria, while the 2010 criteria can diagnose fibromyalgia in patients who do not have sufficient tender points according to the 1990 criteria.

 

The introduction of the 2010 ACR preliminary diagnostic criteria was surrounded by controversy too. In particular, they have been criticized for being completely symptom focused, ill-defined, and lucking some mechanistic features of fibromyalgia, such as hyperalgesia, central sensitization and dysfunctional pain modulation (54). Additionally, these diagnostic criteria are based on the subjective assessment of the patient’s somatic symptoms by the physician, adding ambiguity and influencing repeatability among different physicians (55). A self-reported version of the 2010 ACR preliminary diagnostic criteria was developed in 2011, so as to be used in survey research, and not in clinical practice (56). These criteria are known as the modified 2010 ACR preliminary diagnostic criteria or the 2011 ACR survey criteria. They introduced the fibromyalgia severity (FS) score (originally called fibromyalgianess scale) which is the sum of the self-reported WPI and SS score. This score can be used as an approximate measure of the severity of fibromyalgia. The FS score has also been called polysymptomatic distress (PSD) scale. It has been proposed that the markers of physical and psychological distress have a continuous distribution in the general population with fibromyalgia patients being at the extreme end of this distribution (57). The PSD scale could be useful to define the position of each individual in this continuum, without having to differentiate between patients with  fibromyalgia and those without, as this distinction can sometimes be unclear if not arbitrary (58).

 

2016 Revisions to the 2010/2011 Fibromyalgia Diagnostic Criteria

 

A limitation of the WPI is the fact that it counts the number of painful areas without considering their distribution in the body. Patients with regional pain disorders can fulfill the 2010 ACR preliminary diagnostic criteria since pain can be located in 3 or more areas in the same region (59). To overcome this issue the 2016 revision of the diagnostic criteria require the pain to be generalized (multisite pain). The areas WPI assesses are divided in 5 regions (Table 4) and the diagnosis of  fibromyalgia requires the distribution of pain in 4 out of 5 regions (60). The jaw, the chest and the abdomen area are problematic when they are used to define a region. In this way they are excluded from the definition of generalized pain (61). Since pain needs to be located in at least 4 areas according to the 2016 revision, the previous criterion for diagnosis, WPI of 3-6 and SS scale score ≥9 was changed to WPI of 4-6 and SS scale score ≥9.

 

The 2010 and 2011 ACR preliminary diagnostic criteria are extremely similar. Their difference is that the 2010 criteria are physician-based and can be used in clinical practice for the diagnosis of fibromyalgia, while the 2011 criteria are self-reported and can be used only in survey research. According to the 2010 criteria the SS scale assesses a wide range of somatic symptoms, which makes them impractical for use in questionnaires. With the 2016 revision the assessment of somatic symptoms that is included in the SS scale is limited to headaches, pain and cramps in the lower abdomen and depression. In this way, there is no longer need for different criteria for clinical practice and for survey research. The same criteria can be used in both settings having 2 different methods of administration.

 

One prerequisite for diagnosis of fibromyalgia according to the 2010 ACR preliminary diagnostic criteria is the patient not to have a condition that would otherwise explain the pain. The authors of these criteria clarified that this does not mean that the diagnosis of fibromyalgia is an exclusion diagnosis. However, this phrasing was not considered clear enough and caused significant misunderstanding. In this way this criterion was removed in the 2016 revision. The diagnosis of fibromyalgia can be valid even if there is another condition that can cause the pain that is attributed to fibromyalgia. According to this definition fibromyalgia can coexist with other clinically significant conditions that can cause pain.

 

Table 4. 2016 Revisions to the 2010/2011 Fibromyalgia Diagnostic Criteria

Criteria:

A patient satisfies diagnostic criteria for fibromyalgia if the following 3 conditions are met:

1)    Widespread pain index (WPI) ≥7 and symptom severity (SS) scale score ≥5 or

Widespread pain index (WPI) 4-6 and symptom severity (SS) scale score ≥9.

2)    Generalized pain: Pain must be present in at least 4 of 5 regions.

Jaw, chest, and abdominal pain are not included in generalized pain definition.

3)    Symptoms have been generally for at least 3 months.

4)    A diagnosis of fibromyalgia is valid irrespective of other diagnoses. A diagnosis of fibromyalgia does not exclude the presence of other clinically important illnesses.

 

Ascertainment:

1)    WPI

Note the number of areas in which the patient has had pain over the last week. In how many areas has the patient had pain?

       (Score will be between 0 and 19)

 

Region 1: Left Upper Region

-Jaw, left *

-Shoulder girdle, left

-Upper arm, left

-Lower arm, left

Region 2: Right Upper Region

-Jaw, right *

-Shoulder girdle, right

-Upper arm, right

-Lower arm, right

 

 

Region 5: Axial Region

-Neck

-Upper back

-Lower back

-Chest *

-Abdomen *

 

 

Region 3: Left Lower Region

-Hip (buttock, trochanter), left

-Upper leg, left

-Lower leg, left

Region 4: Right Lower Region

-Hip (buttock, trochanter), right

-Upper leg, right

-Lower leg, right

 

 

 

* Not included in generalized pain definition

 

2)    SS scale score

The SS scale score is the sum of the severity of the 3 symptoms (fatigue, waking unrefreshed, cognitive symptoms) plus the sum of the number of 3 symptoms (headaches, pain or cramps in lower abdomen, depression)

(The final score is between 0 and 12)

 

- For the each of the 3 symptoms below, indicate the level of severity over the past week using the following scale:

0 = no problem

1 = slight or mild problems, generally mild or intermittent

2 = moderate, considerable problems, often present and/or at a moderate level

3 = severe: pervasive, continuous, life-disturbing problems

Fatigue                          (0-3)

Waking unrefreshed     (0-3)

Cognitive symptoms     (0-3)

 

- During the previous 6 months indicate the number of the following symptoms the patient has been bothered by:

  - Headaches                                         (0-1)

  - Pain or cramps in lower abdomen     (0-1)

  - Depression                                         (0-1)

 

The fibromyalgia severity (FS) scale is the sum of the WPI and the SS scale

 

AAPT Diagnostic Criteria

 

In an attempt to improve the recognition of fibromyalgia in clinical practice, the AAPT fibromyalgia working group proposed new diagnostic criteria in 2018 (62). These criteria are similar to the ACR criteria as they require the pain to be generalized (multisite), require the presence of non-pain symptoms and require the symptoms to be present for at least 3 months (Table 5). These diagnostic criteria are more simple than the ACR criteria and they can be easily implemented in primary clinical practice, but some of their aspects have been criticized (63). According to the AAPT criteria the head, the abdomen and the chest are included in the areas that are assessed for the presence of generalized musculoskeletal pain. However, these regions are problematic since pain originating from the teeth, the heart and the bowel can be referred to these areas. Additionally, the AAPT criteria do not have the ability to quantify the severity of fibromyalgia as, apart from the generalized pain, they only assess the presence or absence of the 2 most common non-pain symptoms of fibromyalgia, abolishing all other somatic symptoms. Compared with the 2016 ACR diagnostic criteria, individuals with less symptom severity and fewer pain sites can be classified as fibromyalgia patients, when the AAPT diagnostic criteria are used (64).

 

Table 5. AAPT Diagnostic Criteria

Criteria:

1)    Multisite pain defined as 6 or more pain sites from a total of 9 possible sites:

- Head

- Left arm

- Right arm

- Chest

- Abdomen

- Upper back and spine

- Lower back and spine, including buttocks

- Left leg

- Right leg

2)    Moderate to severe sleep problems or fatigue

3)    Multisite pain plus fatigue or sleep problems must have been present for at least 3 months

 

NOTE. The presence of another pain disorder or related symptoms does not rule out a diagnosis of fibromyalgia. However, a clinical assessment is recommended to evaluate for any condition that could fully account for the patient’s symptoms or contribute to the severity of the symptoms.

 

EPIDEMIOLOGY

 

The prevalence of fibromyalgia depends on the criteria used to define it. Most studies use either the 1990 ACR classification criteria or the modified 2010 ACR preliminary diagnostic criteria (2011 ACR survey criteria) and the prevalence varies between 2-4% (65). Using the 2016 ACR diagnostic criteria the prevalence of fibromyalgia in the general population is 3-4% while with the AAPT diagnostic criteria the prevalence of fibromyalgia is 73% higher, ranging from 5% to 7% (64). In the general population the prevalence increases with age from 2% at the age of 20 to 8% at age of 70. Fibromyalgia appears more often in relatives of patients suffering from fibromyalgia (66), whereas there is a significant difference on the women to men ratio depending on the criteria used to define fibromyalgia. When the 1990 ACR classification criteria are used the women to men ratio is 7:1 while when the 2011 ACR survey criteria are used, that do not use the tender point count, the ratio ranges from 4:1 to 1:1 (58,67–69). Using the 2016 ACR or the AAPT diagnostic criteria there is no statistically significant difference in the prevalence of fibromyalgia between women and men (64).

 

DIFFERENTIAL DIAGNOSIS

 

Several conditions can mimic or overlap with fibromyalgia. In order to reach a differential diagnosis, careful history taking should be followed by a thorough physical examination. Careful neurologic and musculoskeletal examination needs to be performed in all fibromyalgia patients in order to exclude the presence of such conditions. Mood and functional impairment should also be evaluated. This can be easily performed using simple self-administered questionnaires. Patients with obvious mood disturbances should have a formal evaluation by a mental health professional. Baseline blood tests should be limited to a complete blood count, erythrocyte sedimentation rate, a comprehensive metabolic panel, and thyroid function tests. These tests are usually normal in fibromyalgia patients. Consequently, the identification of abnormalities in any of these examinations might suggest that a different condition is present. Additional tests are not recommended for a diagnosis, unless they are clinically indicated. The disorders that can mimic and/or overlap with fibromyalgia and the characteristic clinical features which differentiate them from fibromyalgia are described in Table 6. The clinical features of fibromyalgia, chronic fatigue syndrome (systemic exertion intolerance disease), depression, migraine, and irritable bowel syndrome often overlap being so interchangeable that some authors consider that these conditions should be approached as a “spectrum” of associated disorders (10). They are also considered as part of the spectrum of post-traumatic stress disorder (70,71).

 

 

Table 6. Disorders that can Mimic and/or Overlap with Fibromyalgia Along with Characteristic Clinical Features that Differentiate Them from Fibromyalgia

Disorders

Differentiating clinical features

Rheumatoid arthritis, Systematic Lupus Erythematosus, Sjögren’s syndrome

·      Characteristic synovitis and systemic features of connective tissue disease, apart from musculoskeletal pain, fatigue, Raynaud phenomenon, dry eyes and dry mouth, are usually not features of fibromyalgia.

·      Routine serologic tests are not recommended because of low positive predictive value.

Ankylosing spondylitis, other inflammatory back conditions

·      Generally, there is normal spinal motion in fibromyalgia.

·      Characteristic radiologic features of these disorders are not present in fibromyalgia.

Polymyalgia rheumatica

·      Tender points are not always present in polymyalgia rheumatica.

·      Stiffness is more prominent than pain in polymyalgia rheumatica.

·      Most patients with polymyalgia rheumatica have increased erythrocyte sedimentation rate, while it is normal in fibromyalgia.

·      Patients with polymyalgia rheumatica respond extremely well to modest doses of corticosteroids, in contrast to fibromyalgia patients.

Inflammatory myositis, metabolic myopathies

·      Myositis and myopathies can cause muscle weakness and muscle fatigue, but they are not usually associated with diffuse pain.

·      Patients with myositis or myopathies have abnormal muscle enzyme tests and specific histopathologic findings on muscle biopsy, in contrast to fibromyalgia patients (muscle biopsy should be limited to cases that there is clinical evidence suggestive of myositis or myopathy).

Statin myopathy

·      Statin myopathy symptoms are limited to muscle weakness and pain without other symptoms associated with fibromyalgia.

·      Statin myopathy pain is temporally associated with statin therapy.

·      Statin myopathy can be associated with abnormal muscle enzyme tests.

Infection:

chronic viral infection (e.g., infectious mononucleosis, HIV, HTLV, HBV, HCV, Lyme disease), long COVID

·      In fibromyalgia patients there is no objective evidence of inflammation or organ system dysfunction

Hypothyroidism

·      Although thyroid autoantibodies are common in fibromyalgia patients, thyroid function tests are usually normal.

Hyperparathyroidism

·      Hypercalcemia is not present in fibromyalgia.

Cushing’s syndrome

·      Cushing’s syndrome is associated with muscle weakness rather than pain.

·      The characteristic facial and skin signs of Cushing’s syndrome are not present in fibromyalgia.

Adrenal insufficiency

·      Adrenal insufficiency causes severe exhaustion, while it is not typically associated with chronic widespread pain.

Hypophosphatasia

·      Most hypophosphatasia patients have low alkaline phosphatase

Neurologic diseases:

peripheral neuropathies, cervical radiculopathy, entrapment syndromes (e.g., carpal tunnel syndrome), multiple sclerosis, myasthenia gravis

·      Multiple sclerosis and myasthenia gravis are associated with post-exercise muscle and generalized fatigue, but not with widespread pain.

·      Thorough neurologic examination can reveal neurologic signs characteristic of specific diseases.

Myofascial pain syndromes (they may include other common regional pain disorders such as tension headaches, occupational overuse syndrome, cumulative trauma disorder, work related musculoskeletal disorder,idiopathic low back and cervical strain disorders, chronic pelvic pain temporomandibular disorderand interstitial cystitis)

·      In myofascial pain syndromes the pain and the tenderness is confined in one anatomic region

Chronic fatigue syndrome

(systemic exertion intolerance disease)

·       Criteria for the diagnosis of chronic fatigue syndrome:

 

1.     According to the modified United States Centers for Disease Control and Prevention chronic fatigue syndrome is diagnosed when two criteria are fulfilled (72):

i)     Clinically evaluated, unexplained, persistent or relapsing fatigue that is of new or definite onset; is not the result of ongoing exertion; is not alleviated by rest; and results in substantial reduction in previous levels of occupational, educational, social, or personal activities

ii)    Four or more of the following symptoms that last six months or longer:

-    Impaired memory or concentration

-    Post-exertional malaise where physical or mental exertions bring on "extreme, prolonged exhaustion and sickness"

-    Unrefreshing sleep

-    Muscle pain

-    Arthralgia in multiple joints

-    Headaches of new kind or greater severity

-    Frequent or recurring sore throat

-    Tender cervical or axillary lymph nodes

 

2.     According to the proposed diagnostic criteria of the United States Institute of Medicine the chronic fatigue syndrome (systemic exertion intolerance disease) is diagnosed when two criteria are fulfilled (73):

i)    All of the following symptoms:

-    A substantial reduction or impairment in the ability to engage in pre-illness levels of occupational, educational, social, or personal activities that persists for more than 6 months and is accompanied by fatigue, which is often profound, is of new or definite onset (not lifelong), is not the result of ongoing excessive exertion, and is not substantially alleviated by rest

-    Post-exertional malaise*

-    Unrefreshing sleep*

ii)   Two or more of the following manifestations:

-    Cognitive impairment*

-    Orthostatic intolerance

-     

*   The diagnosis should be questioned if patients do not have these symptoms at least half of the time with moderate, substantial, or severe intensity.

 

·       Chronic widespread pain is not included in the criteria for diagnosis of chronic fatigue syndrome

Psychiatric disorders:

depression, anxiety disorders, posttraumatic stress disorder

·       In fibromyalgia patients with a concurrent psychiatric disorder, the attribution of symptoms to fibromyalgia or the psychiatric disorder is not always possible.

Sleep disorders:

obstructive sleep apnea, restless legs syndrome, periodic limb movement disorders

·       Detail history can identify the majority of the primary sleep disorders.

·       Chronic widespread pain is uncommon in primary sleep disorders.

Irritable bowel syndrome

·       According to the 2009 American College of Gastroenterology recommendations for the diagnosis of irritable bowel syndrome, it is defined by abdominal pain or discomfort that occurs in association with altered bowel habits over a period of at least three months (74).

Temporomandibular disorders

·       Temporomandibular disorders are characterized by recurrent facial/jaw pain and/or limitation in jaw opening occurring in the past six months.

Tension – Migraine headache

·       Tension – migraine headache is characterized by recurrent headaches (at least five for migraine, at least 10 for tension-type) lasting 30 minutes.

Interstitial cystitis

·       According to the American Urological Association guidelines interstitial cystitis is defined as an unpleasant sensation (pain, pressure, discomfort) perceived to be related to the urinary bladder, associated with lower urinary tract symptoms of more than six weeks duration, in the absence of infection or other identifiable causes (75).

HIV: human immunodeficiency virus, HTLV: human T-lymphotropic virus, HBV: hepatitis B virus, HCV: hepatitis C virus, COVID: coronavirus disease

 

PATHOPHYSIOLOGICAL MECHANISMS

 

Pain sensitivity in the population occurs over a wide continuum, forming a classic bell-shaped curve, just like any other physiologic variable. Genetic predisposition in combination with environmental factors, determine the place that each individual takes in this continuum. People who are placed in the right end of this curve are very sensitive to pain and they can probably develop pain even without having any inflammation or damage in the peripheral tissues. This pain can be either regional or widespread (39).

 

In the past fibromyalgia was thought to be a primary muscle disease. However, controlled studies found no evidence of significant pathologic or biochemical muscle abnormalities that can be the cause of chronic widespread pain and tenderness. Most investigators believe that any muscle pathology is secondary to chronic pain and inactivity, rather than primary in nature (76–79). Current research suggests that altered central nervous system (CNS) physiology might underlie the symptoms of fibromyalgia. Abnormal central sensory processing of pain signals seems to play a significant role in the pathogenesis of fibromyalgia. This dysregulation of the nociceptive system can arise from a combination of interactions between neurotransmitters, cytokines, hormones, the autonomic nervous system, behavioral constructs, and external stressors.

 

Abnormalities in Sensory Processing

 

Fibromyalgia overlaps with several other similar syndromes including chronic fatigue syndrome (systemic exertion intolerance disease) and myophasial pain syndrome (Table 6). It has been proposed that these disorders should be considered as members of the central sensitivity syndromes (Table 7). These similar and overlapping syndromes are bound by the common mechanism of central sensitization that involves hyper-excitement of the second-order neurons, especially the wide-dynamic-range neurons (WDR) in the dorsal horns of the spinal cord,  by various synaptic and neurotransmitter/neuromodulator activities (6). Central sensitization is clinically and physiologically characterized by hyperalgesia, allodynia, expansion of the receptive field (pain expanding beyond the area of the peripheral nerve supply, after the application of a nociceptive stimulus), prolonged electrophysiological discharge, and an after-stimulus unpleasant quality of the pain (e.g., burning, throbbing, tingling or numbness). Parallel to central sensitization, temporal summation takes place in the second-order neurons. It is characterized by a progressive increase in electrical discharges (and consequently increase in the perceived intensity of pain) in response to repetitive short noxious stimuli. Temporal summation involves the production of second pain, which is described as dull or burning, and leaves an after-stimulus unpleasant sensation (80).

 

Table 7. Central Sensitivity Syndromes

·       Fibromyalgia

·       Chronic fatigue syndrome (systemic exertion intolerance disease)

·       Irritable bowel syndrome

·       Tension type headaches

·       Migraine

·       Temporomandibular disorder

·       Myophasial pain syndrome

·       Restless leg syndrome

·       Periodic limb movements in sleep

·       Primary dysmenorrhea

·       Interstitial cystitis

·       Posttraumatic stress disorder

 

N-methyl-D-aspartate (NMDA) receptors are mostly responsible for escalation of hyperexcitability of the second-order nociceptive neurons. The role of the major neurotransmitters of the nociceptive system that participate in signal conduction at the level of the spinal cord is briefly illustrated in Figure 3 (6).

 

Figure 3. The role of the major neurotransmitters of the nociceptive system that participate in signal conduction at the level of the spinal cord. SP: Substance P, NGF: nerve growth factor, NMDA: N-methyl-D-aspartate, D: dopamine.

 

The second-order neurons have ascending projections to the thalamus, hypothalamus, the limbic system and the somatosensory cortex. These supraspinal structures are involved in the sensory, evaluative and affective dimensions of pain (e.g., unpleasantness, emotional reaction). Several descending pathways from the cortico-reticular system, locus ceruleus, hypothalamus, brain stem, and local spinal cord interneurons terminate to the dorsal horn cells. These pathways utilize neurotransmitters that include serotonin (5-HT), norepinephrine, γ-amino-butyric acid (GABA), enkephalins and adenosine (6). This descending system, once thought to be predominantly inhibitory, is now known to have a facilitatory potential (81). Evidence suggests that the 5-HT3 receptor has a facilitatory function, while the 5H-T1Areceptor is inhibitory. The ascending and descending pathways should not be considered as dichotomous in function. They are interactive and their functions are bidirectional. Both pathways can either facilitate or inhibit pain, depending on the site of action and the neurotransmitters that are used (6).

 

The dysregulation of the nociceptive system, either at the level of the dorsal horns of the spinal cord, or at the level of the ascending and descending pathways, can lead to its hyper-excitability. In other words, it can lead to central sensitization. Several factors may amplify and sustain central sensitization through interactive and synergistic actions. These factors are summarized in Table 8 (80). Central sensitization can become self-sustained, even when the event that triggered it no longer exists, due to long-term CNS plasticity.

 

Table 8. Factors that may Amplify and Sustain Central Sensitization

·       Genetics

·       Sympathetic over-activity

·       Endocrine dysfunctions

·       Viral infection

·       Peripheral nociception generators (e.g., arthritis)

·       Poor sleep

·       Environmental stimuli (e.g., weather, noise, chemicals)

·       Psychological distress (e.g., adverse childhood experience)

 

Neuroimaging studies provide moderate evidence for structural changes in the brain of patients with fibromyalgia. Gray matter volume appears to be reduced in areas related with pain processing, such as the cingulate, the insular, and the prefrontal cortices (82). Functional MRI studies reveal alterations in the functional connectivity of brain areas responsible for pain processing and provide support of functional dysregulation of the ascending and descending pain pathways in fibromyalgia patients (82–85). Additionally, alterations in neuronal activity between the ventral and the dorsal spinal cord have been demonstrated in fibromyalgia patients (86).

 

Although nearly all of the research on sensory processing in fibromyalgia has focused on the processing of pain, there are some data suggesting a more generalized disturbance in sensory processing. There is evidence that fibromyalgia patients have a hypersensitivity to unpleasant stimuli of other sensory systems. For example, many patients experience reduced tolerance to loud noises, bright lights, odors, drugs, and chemicals (87,88).

 

Neurotransmitters

 

The levels of Substance P (SP) in the cerebrospinal fluid (CSF) in patients with fibromyalgia are significantly increased compared to normal individuals, whereas CSF levels of serotonin metabolites are decreased, as are metabolites of dopamine and norepinephrine (89).

 

The first direct evidence that fibromyalgia patients may have abnormal dopamine response to pain originated from positron emission tomography (PET) competitive binding studies using the D2/D3 receptor antagonist [11C] raclopride. It was shown that dopamine is released in response to tonic toxic noxious muscle stimulation, but not after non-painful muscle stimulation in healthy human subjects. In contrast the dopamine response in fibromyalgia patients did not differ between painful and non-painful muscle stimulation (87). There are indications that disturbances of the opioidergic system occur in fibromyalgia patients, as there is an up-regulation of opioid receptors in the periphery, with a reduction of the brain opioid receptors (90,91). This implies an increased baseline endogenous opioidergic activity. Opioids can activate glial cells, via a non-stereoselective activation of toll-like receptor 4 (TLR4). Glial cells in turn can mediate pain by releasing neuroexcitatory, pro-inflammatory products (92).

 

In a study where fibromyalgia patients were evaluated for cortical excitability and intracortical modulation using transcranial magnetic stimulation of the motor cortex, it was shown that there were deficits in intracortical modulation of GABAergic and glutamatergic mechanisms (93). Diminished inhibitory neurotransmission resulting from lower concentrations of GABA within the right anterior insula of patients with fibromyalgia was documented using proton magnetic resonance spectroscopy (94). Evidence for enhanced glutaminergic neurotransmission in fibromyalgia patients is derived from studies that used magnetic resonance spectroscopy. It was shown that fibromyalgia patients have significantly higher levels of glutamine within the posterior insula and in the right amygdala (95,96). The levels of brain-derived neurotrophic factor, which is involved in neuronal survival and synaptic plasticity of the central and peripheral nervous system, have been found to be increased both in the brain and in the plasma of fibromyalgia patients (97).

 

Cytokines

 

Although fibromyalgia is not considered an inflammatory disorder, the interaction of immunological mechanisms with pain physiology, has led to the identification of alterations in the levels of various cytokines (98). However, it is not clear whether cytokine changes are the cause of pain in these patients, or just its consequence.

 

The serum levels of interleukin 1 receptor antibody (IL-1Ra), IL-6, and IL-8, and the plasma levels of IL-8 are higher in fibromyalgia patients, compared to controls (99). Inflammatory cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) have been detected in skin biopsies taken from fibromyalgia patients, possibly indicating an element of neurogenic inflammation (100). Lower levels of the anti-inflammatory cytokines IL-4 and IL-10 have been reported in fibromyalgia patients compared to healthy controls (101,102). However, the interpretation of cytokine levels is not always easy. Most cytokines are expressed in low levels and a sensitive bioassay is needed for their detection. Consequently, a negative result can merely be the ramification of a not sensitive enough method. Additionally, cytokine levels can be vigorously influenced by a number of factors, including circadian rhythmicity, physical activity, and co-morbid conditions, including depression.

 

Inflammatory cytokines like IL-1β, IL-6 and TNFα can elicit pain, induce hyperalgesia and they are associated with neuropathic pain (103), although they do not appear to be involved in “normal” pain. Although serum cytokines do pass the blood brain barrier the release of pro-inflammatory cytokines by immune cells in the body leads, in turn, to release of pro-inflammatory cytokines by glial cells within the brain and spinal cord (104).Inflammatory cytokines like IL-1β, IL-6 and TNFα can also cause activation of the hypothalamic-pituitary-adrenal (HPA) axis alone, or in synergy with each other. There is evidence to suggest that IL-6, which is the main endocrine cytokine, plays the most significant role in the immune stimulation of the axis, especially in chronic inflammatory stress (105). IL-6 can stimulate the hypothalamic secretion of corticotropin-releasing hormone (CRH) and arginine vasopressin (AVP), leading to the increase of serum adrenocorticotropic hormone (ACTH) and cortisol levels (106).

 

Hypothalamic-Pituitary-Adrenal Axis

 

There is substantial data supporting an abnormal function of the HPA axis in fibromyalgia. However, the results from studies on the HPA axis function in fibromyalgia patients are relatively heterogeneous and partially contradictory (107). The 24-hour urinary free cortisol (UFC) has been found to be reduced or normal (108). Findings regarding alterations in diurnal variation of cortisol secretion are also inconsistent. Although normal diurnal patterns of ACTH and cortisol have been reported, there is data demonstrating flattened cortisol diurnal rhythm with normal morning peak and higher evening cortisol, levels (109). It has been demonstrated that there is a significant decrease in the rate of decline from acrophase (peak) to nadir for diurnal cortisol levels in fibromyalgia patients, compared to controls, while there is no change in the ACTH to cortisol ratio (108). This implies a decreased ability of the HPA axis to return to baseline after a physiologic stimulation by meals, several other activities or even pain (108). Decreased morning cortisol release and reduced frequency of cortisol pulses over 24 hours have also been reported. There is evidence to suggest  that the reduced cortisol release in fibromyalgia patients is associated with depressive symptoms and experiences of childhood trauma (109).

 

In line with studies suggesting reduced adrenal output in fibromyalgia patients, reduced cortisol secretion has been reported in response to pharmacological challenge with synthetic ACTH1-24 and  insulin tolerance test (109). Patients with fibromyalgia exhibit increased ACTH, but normal cortisol response to CRH stimulation, compared to controls. This finding suggests a sensitization of the pituitary in combination with a degree of adrenal insufficiency (109). Arginine vasopressin (AVP), an ACTH secretagogue, has been found to be more increased in response to the postural challenge test in fibromyalgia patients, compared to controls (110). Alterations in the feedback regulation of HPA axis have also been reported in fibromyalgia patients, using the overnight dexamethasone suppression test (DST). Increased rates of non-suppression following the standard (1 mg) DST have been reported in fibromyalgia patients, compared to controls, but this finding was difficult to be interpreted as it was associated with depression. Interestingly, other studies have revealed lower rates of non-suppression in fibromyalgia patients (111).

 

There are indications that there is a dissociation between total and free cortisol levels in fibromyalgia patients, with normal salivary and plasma free cortisol despite diminished total cortisol levels. One possible explanation of this finding is a reduced concentration of the glucocorticoid binding globulin (CBG). Reduced levels of CBG have been reported in fibromyalgia patients compared to controls. It is of particular interest that chronic social stress might result in reduced CBG levels, whereas IL-6 and IL-1β that can also inhibit the production of CBG may contribute further (109). Apart from HPA axis abnormalities in fibromyalgia patients, abnormal levels of growth hormone have also been found in some, but not all reports (112). On the other hand the levels of sex hormones have not been clearly shown to differ between female fibromyalgia patients and controls (113).

 

Autonomic Nervous System

 

Sympathetic hyperactivity, often associated with sympathetic hypo-activity in response to stressors, or parasympathetic underactivity has been described in fibromyalgia. Attenuated sympathetic and parasympathetic activity was demonstrated in a study where fibromyalgia patients and healthy controls were assessed for 24 hours in a controlled hospital setting, including relaxation, a test with prolonged mental stress and sleep. The urinary catecholamine levels were found to be lower in fibromyalgia patients compared to controls. Patients with fibromyalgia had significantly lower adrenaline levels during the night and the second day of the study, and significantly lower dopamine levels during the first day, the night, and the second day. Furthermore, heart rate during relaxation and sleep was significantly higher in patients than in controls (114). In another study, plasma catecholamines, ACTH, and cortisol levels were reduced in 16 fibromyalgia patients compared to 16 healthy controls while performing static knee extension until exhaustion (115). Nocturnal heart rate variability indices have been shown to be significantly different in fibromyalgia women compared to healthy individuals, indicating a sympathetic predominance (116).  In addition, orthostatic hypotension and increased pain in response to tilt table test have been described along with increased resting supine heart rate and decreased heart rate variability  (117,118). IL-6 administration causes exaggerated norepinephrine responses and increases in heart rate, as well as delayed ACTH release, suggesting an incapacitated stress-regulating system (119). In-vitro testing of beta adrenergic receptor mediated cyclic AMP generation has revealed decreased responsiveness to beta-adrenergic stimulation (120).

 

Overall, it has been suggested that sympathetic dysfunction can not only cause diffuse pain, but also contribute to other symptoms like sleep disturbances, due to sustained nocturnal sympathetic activity, and fatigue, due to deranged sympathetic response to stress (6).

 

Psychological, Cognitive, and Behavioral Factors

 

Pain apart from a sensory-discriminative dimension, which includes the location and the intensity of pain, has a very significant psychological component. This includes the affective dimension of pain, the emotional valance of pain in other words, as well as attention and cognitive aspects, which are based on CNS mechanisms. Emotion and selective attention can enhance pain perception, with the involvement of the descending pathways that have a facilitatory effect on the spinal cord dorsal horn neurons (80). Catastrophizing has been shown to be related to decreased pain threshold and tolerance to heat stimuli in fibromyalgia patients. However there is a subgroup of fibromyalgia patients that is very tender, despite the fact that they do not catastrophize and they have a moderate control over their pain (11). A fMRI study has shown that although depression is associated with the magnitude of neuronal activation in brain regions that process the affective-motivational dimension of pain, neither the extent of depression nor the presence of comorbid major depression modulated the sensory-discriminative aspects of pain processing in fibromyalgia patients (121). Catastrophizing, has been associated with increased activity in brain areas related to anticipation, attention and the emotional aspects of pain, as shown by fMRI in response to pressure stimuli. This study also revealed an association between catastrophizing and increased activity in the secondary somatosensory cortex, indicating that the way patients think about their pain might actually influence its sensory processing (80).

 

Genetic Predisposition

 

It is currently well established that familial aggregation is a characteristic of fibromyalgia. First degree relatives of fibromyalgia patients are 8.5 times more likely to have fibromyalgia than relatives of patients with rheumatoid arthritis (12). As with other complex and multifactorial syndromes, the occurrence of familial aggregation in the case of fibromyalgia does not necessarily imply a genetic basis. Shared environmental factors and learned patterns of behavior that may evolve within families are equally valid targets of investigation.

 

Genome-wide association studies have shown significant differences in allele frequencies between fibromyalgia patients and controls. However many of the results are inconsistent, without being replicated. Additionally the small sample size of these studies limits the genetic variants that can be identified only to those with large effects (122–126). The coexistence of other comorbidities in fibromyalgia patients further obscures these results. In a large scale genome-wide association study of 26,749 individuals the overall estimated heritability of fibromyalgia was 14%. There was a significant difference between age groups, with the heritability in individuals less than 50 years of age to be 23.5%, while in those over 60 years of age it was only 7.5% (126).

 

Although no specific candidate gene has been identified, the following genes have been associated with fibromyalgia:

 

SEROTONIN TRANSPORTER (5-HTT) GENE

 

An increased frequency of the S/S genotype of the 5-HTT gene has been found in fibromyalgia patients compared to controls (127,128). However this putative association may be limited to patients with concomitant affective disorders, since it was not confirmed in fibromyalgia patients without depression or anxiety (129).

 

D4 RECEPTOR GENE

 

Polymorphisms affecting the number of repeats in the third cytoplasmic loop of the dopamine D4 receptor gene have been shown to be significantly decreased in frequency in fibromyalgia patients (130).

 

CATECHOL-O-METHYL TRANSFERASE (COMT) GENE

 

The homozygous low activity (met/met) and the heterozygous low activity (val/met) COMT genotypes occur more often in fibromyalgia patients than in controls, whereas the homozygous high activity (val/val) genotype is less frequent (131). However in a meta-analysis COMT gene val(158)met polymorphism was not associated with an increased risk for fibromyalgia (132). The met/met genotype has been associated with greater fibromyalgia illness severity across the domains of pain, fatigue, sleep disturbance, and psychological distress, while fibromyalgia patients with the met/met polymorphism experienced a greater decline in exhibiting a positive attitude on days when pain was elevated than did patients with the val/met or val/val genotype (133).

 

OPIOID RECEPTOR μ 1 GENE (OPRM1)

 

The 118G allele frequency has been described to be significantly lower in patients with fibromyalgia than in the control group (134).

 

ADRENERGIC RECEPTOR GENES

 

The presence fibromyalgia and its symptom severity is associated with various adrenergic receptor gene polymorphisms (135).

 

Other genes associated with the regulation of nociceptive and analgesic neuronal pathways

Specific variants of trace amine-associated receptor 1 (TAAR1) gene, regulator of G-protein signaling 4 (RGS4) gene, cannabinoid receptor 1 (CNR1) gene, and glutamate receptor, ionotrophic, AMPA 4 (GRIA4) gene, have been associated with fibromyalgia (123).  

 

External Stressors

 

Almost all diseases are caused by a combination of genetic predisposition and the effect of environmental factors. We are now beginning to better understand the environmental factors that seem to be important in triggering fibromyalgia. Most of them act as “stressors” that when superimposed onto a deranged stress-response system can lead to the dysregulation of the nociceptive system.

 

PERIPHERAL PAIN SYNDROME

 

Pain due to damage or inflammation of peripheral tissues may trigger fibromyalgia. Additionally, small fiber neuropathy can be associated with fibromyalgia (136–138). Chronic localized – regional pain can lead to central sensitization and pain dis-inhibition, causing pain hypersensitivity and widespread pain. Systematic autoimmune diseases can be associated with fibromyalgia too. Approximately 20-25% of patients with rheumatoid arthritis, systemic lupus erythematosus and ankylosing spondylitis, have co-morbid fibromyalgia (18). In such cases, it is important to realize that many symptoms may be attributed to fibromyalgia rather than the underlying disorder. This recognition has significant clinical implications.

 

INFECTIONS

 

Various infections have been linked to the development of fibromyalgia and chronic fatigue syndrome (systemic exertion intolerance disease). Epstein-Barr virus, parvovirus, Lyme disease, Q fever, HIV and hepatitis C virus (HCV), have been suggested as triggers of fibromyalgia or chronic fatigue syndrome (systemic exertion intolerance disease), but more robust evidence is needed. The role of vaccination in precipitating fibromyalgia and related syndromes is still not clear (139,140).

 

PHYSICAL TRAUMA

 

Various forms of physical trauma have been considered as culprits of triggering the pathogenesis of fibromyalgia. Many patients report the initiation or the exacerbation of their symptoms after a traumatic event such as whiplash injury, while increased rates of fibromyalgia have been demonstrated among patients undergoing cervical trauma during motor vehicle accidents (141,142).

 

PSYCHOLOGICAL DISTRESS

 

It has been considered that psychological factors that give rise to chronic stress may initiate the chain of events that leads to fibromyalgia. The chronic stress can be a result of the accumulation of daily stress events. Emotional stress, catastrophic events such as war, job loss, marital discord and excess family responsibilities such as caring for sick elders, have been implicated as triggers of fibromyalgia (143). However the data that supports the notion that psychological stress and distress directly causes fibromyalgia is rather weak (39).

 

MANAGEMENT

 

The treatment of fibromyalgia is challenging because of our limited understanding of its pathogenesis and the poor response of patients to conventional pain treatments. The aim of the therapy is to relieve pain and increase function using a multimodal individualized therapeutic strategy which, in most cases, includes pharmacologic and non-pharmacologic interventions. Current clinical-based evidence supports the use of a multimodal program that includes education, exercise, cognitive-behavioral approaches and medications. The treatment should be individualized based on the symptoms, the comorbidities and the preferences of the patient, who should be encouraged to participate in the decision-making process of selecting the optimal therapies (144,145). Coexisting disorders are common in fibromyalgia patients. Their identification and effective treatment can have beneficial effects on fibromyalgia symptoms. It is also important to assure that adequate adherence to both pharmacological and non-pharmacological treatment is maintained, so as to achieve the optimal benefit from these treatments.

 

Non-Pharmacological Management

 

PATIENT EDUCATION

 

The first step should be the education of the patient. The patients with fibromyalgia need to understand their illness before any treatment modality is used (146). Providing a diagnosis, “labeling” the patient with fibromyalgia, may have beneficial effects. It has been shown that fewer symptoms and an improvement in health status is noted after the patients are informed of their diagnosis (147,148). The physician should clarify that fibromyalgia is a real illness and the symptoms the patient experiences are not imaginary. The role of neurotransmitters and neuromodulators in pain perception, fatigue, abnormal sleep and mood disturbances should be discussed, so as the patient to understand the rationale of the pharmacologic therapy, especially when antidepressant drugs are used. The significance of good sleep hygiene should be reviewed and poor sleep habits should be addressed. Fibromyalgia patients who are overweight or obese should be informed for the adverse effect of increase body mass index to fibromyalgia symptoms and quality of life (149). For these patients weight reduction should be encouraged. The patient also needs to acknowledge that fibromyalgia is a chronic relapsing condition without though being life-threatening nor deforming.

 

EXERCISE

 

Another potent non-pharmacological treatment for fibromyalgia is exercise. It has been reported that an exercise program incorporating aerobic, strengthening, and flexibility elements can lead to greater benefits than a relaxation program. Exercise in fibromyalgia patients should have two major components: strengthening to increase soft-tissue length and joint mobility, and aerobic conditioning to increase fitness and function, reduce fibromyalgia symptoms and improve quality of life (144,150–153). Exercise should be of low impact and of sufficient intensity so as to be able to change aerobic capacity (28). Successful interventions include fast walking, biking, swimming, water aerobics, tai chi, and yoga. Land and aquatic training appears to be equally beneficial (154). An improvement in the severity of fibromyalgia symptoms has also been achieved with web-based exercise programs (155). A gradual incremental increase in physical activity should be encouraged as it is common for fibromyalgia patients to initially perceive an aggravation of their pain and fatigue at the beginning of a training program. It has been suggested that in the presence of exercise-induced pain, the intensity and duration of exercise should be reduced, while its frequency should be maintained, so as to avoid any further decrease in exercise tolerance (144). The type and intensity of the exercise program should be individualized and should be based upon patient preference and the presence of any other cardiovascular, pulmonary, or musculoskeletal comorbidities.

 

COGNITIVE-BEHAVIORAL APPROACHES

 

One of the goals of the management should be to help patients understand the effect of thoughts, beliefs and expectations on their symptoms. This can help them to abolish the perception of helplessness and the catastrophizing thoughts that can adversely influence their condition. Patients with greater self-efficacy are more likely to have a good response to treatment programs and experience better outcomes. The beneficial effect of cognitive-behavioral therapies in fibromyalgia patients with anxiety and depression disorders is limited to a reduction of negative mood, while the rest of the patients also demonstrate a reduction of pain and fatigue. Psychologically based interventions, have been proven to be useful when they are compared to no treatment or treatment other than aerobic exercise (156). In a 2021 systematic review and meta-analysis there was high quality evidence that cognitive-behavioral therapy can significantly reduce the pain intensity in fibromyalgia patients for 3 months (157). Preliminary data from functional MRI studies suggest that cognitive-behavioral therapies have the ability to restore the alterations in the functional connectivity of brain areas responsible for pain processing observed in fibromyalgia patients (158,159).

 

COMPLEMENTARY AND ALTERNATIVE APPROACHES

 

Acupuncture

 

Acupuncture is the insertion of needles in the human body. There are different styles of acupuncture depending on the location and the depth the needles are inserted. The inserted needles can be stimulated by heat, electrical current (electro-acupuncture), mechanical pressure (acupressure), or laser (laser acupuncture). The most common type of acupuncture involves skin penetration without stimulation (manual acupuncture).  Sham or fake acupuncture is a research tool to control the effects of real acupuncture. It can involve skin contact with the needles without actual penetration or needle insertion in areas other than the ones usually targeted.

 

In a high quality meta-analysis it was demonstrated that the effects of manual acupuncture on pain, sleep quality and global well-being did not differ significantly from the effects of sham acupuncture. On the contrary electro-acupuncture significantly reduced pain, fatigue, and stiffness, while it improved sleep quality and global well-being when compared to sham acupuncture. Additionally, electro-acupuncture significantly improved pain, stiffness, and global well-being when compared to non-acupuncture. The beneficial effects of acupuncture could be observed at 1 month after treatment, but they were not maintained at 6-7 months (160).

 

Other

 

The effectiveness of meditative movement therapies (qigong, yoga, tai chi) on sleep and fatigue improvement and of hydrotherapy on pain reduction has been supported by some studies (161,162). A number of other modalities has also been utilized for the treatment of fibromyalgia including biofeedback, chiropractic therapy, massage therapy, hypnotherapy, guided imagery, electrothermal therapy, phototherapeutic therapy, music therapy, journaling / storytelling, static magnet therapy, transcutaneous electrical nerve stimulation, and transcranial direct current stimulation. However there are no well-designed studies to advocate their general use (145).

 

Pharmacologic Treatment

 

A wide range of drugs has been used in the treatment of fibromyalgia including antidepressants, sedatives, muscle relaxants and antiepileptic drugs. The choice of medication is influenced by patient preference; prominence of particular symptoms, including fatigue, insomnia, and depression; potential adverse effects; patient tolerance of individual medications; cost and regulatory limitations on prescription choice (163,164). Nonsteroidal anti-inflammatory drugs and opioids, although often prescribed for fibromyalgia, are not an effective form of treatment (39,165). However analgesics and anti-inflammatory medications can be useful in case of coexisting conditions that cause regional pain, like arthritis, which can aggravate or trigger the fibromyalgia symptoms. Regarding opioids, with the exception of tramadol, apart from not being effective for the treatment of fibromyalgia symptoms, their long-term use also curries a dose-dependent risk for serious adverse effects, including overdose, abuse, fractures, myocardial infarction and sexual dysfunction (166). Additionally opioids in fibromyalgia patients can reduce the effectiveness of psychological therapy (167), while their long-term use can cause sleep disturbances (168).

 

Patients should be informed that for most pharmacologic therapies several weeks may be needed until they experience a benefit. Initially a single drug should be administered. However, in the case of non-responsiveness combination therapy should be considered. Since therapeutic responses are rarely durable, physicians should not be surprised when the initial efficacy of a medication is abolished. Successful treatment of fibromyalgia may require regular reassessment and possible rotation or combination of medications (169). Adequate dose prescription and patient adherence are significant for the effectiveness and tolerability of pharmacologic treatment (170). The doses of the most commonly used medications with strong and moderate evidence of effectiveness are shown in Table 9.

 

ANTIDEPRESSANTS

 

Tricyclic Antidepressants (TCAs)

 

TCAs are often used as initial treatment for fibromyalgia. Their analgesic effect is independent of their antidepressant action and is thought to be mediated by inhibition of norepinephrine (rather than serotonin) reuptake at spinal dorsal horn synapses, with secondary activity at the sodium channels. The most widely studied drugs of this group are amitriptylineand cyclobenzaprine. They should be administered at lower doses than those required for the treatment of depression, a few hours before bedtime, and their dose should be escalated very slowly. A clinically important improvement is observed in 25-45% of patients treated with TCAs compared to 20% in those taking placebo (171–175). However their use is limited by the fact that they are ineffective or intolerable in 60-70% of patients (144), while their efficacy may decrease over time (171,176).

 

Amitriptyline is more efficient compared to the serotonin-norepinephrine reuptake inhibitors duloxetine and milnacipran in reducing pain, sleep disturbance, and fatigue, without differences in acceptability, as it was shown in a systematic review and meta-analysis (177). In a 2022 network meta-analysis comparing amitriptyline, duloxetine and pregabalin it was shown that treatment with amitriptyline 25 mg was superior to duloxetine and pregabalin for the reduction of pain intensity for at least 50% (178). The combination of 20 mg of fluoxetine in the morning with 25 mg of amitriptyline at bedtime has been shown to be more effective than either medication alone (179). Side effects of amitriptyline include dry mouth, constipation, fluid retention, weight gain, difficulty in concentrating and possibly cardiotoxicity.

 

Cyclobenzaprine has a similar tricyclic structure and presumed mode of action with amitriptyline in fibromyalgia, but is thought to have minimal antidepressant effect (163). A meta-analysis of five placebo-controlled trials has revealed improvement of the global functioning, with a similar effect size as this reported for amitriptyline. The group that received cyclobenzaprine had a significant decrease in pain for 4 weeks, compared to those treated with placebo, but the decrease in pain was not significantly different after 8 and 12 weeks. Sleep was improved at all time points in both cyclobenzaprine and placebo groups, while no effect was noted on fatigue (171–173). It has been demonstrated that the use of very low-dose cyclobenzaprine (1 to 4 mg at bedtime) can improve the symptoms of fibromyalgia, including pain, fatigue, and depression, compared to symptoms at baseline and to placebo. Significantly more patients who received the very low-dose of cyclobenzaprine experienced improved restorative sleep, based upon analysis of cyclic alternating pattern of sleep by electroencephalography. The increase in nights with improved sleep by this measure correlated with improvements in fatigue and depression (180).

 

Desipramine has fewer anticholinergic and sedative effects than other TCAs, which can make it a possible alternative, although its efficacy is not well studied in fibromyalgia.

 

Serotonin-Norepinephrine Reuptake Inhibitors (SNRIs)

 

SNRIs are similar to TCAs in their ability to inhibit the reuptake of both serotonin and norepinephrine, but they differ from TCAs in being devoid of significant activity at other receptor systems, resulting in diminished side effects and increased tolerance. Venlafaxine, duloxetine and milnacipran have been shown to be effective in diminishing fibromyalgia symptoms (147,181,182). These drugs can be used in fibromyalgia patients who do not respond to a trial of low-dose TCAs or who have intolerable side effects. They can also be administered as an alternative to amitriptyline for initial therapy especially for patients with significant fatigue or depression. Of these medications duloxetine and milnacipranare better studied and they are preferred to be administered to patients with fibromyalgia. There are more limited data regarding the efficacy of venlafaxine for fibromyalgia, while withdrawal symptoms if a dose is missed occur more often, because of the short half-life of this medication (183). A meta-analysis has shown that fibromyalgia patients treated with duloxetine at 60mg daily are more likely to have more than 50% reduction in pain, compared to patients taking placebo (184). However, duloxetine at 30mg daily does not significantly reduce pain (185). The efficacy of duloxetine can be maintained at 3 and 6 months of treatment (186). In a 2018 systematic review and meta-analysis it was shown that duloxetine and milnacipran were not superior to placebo in the frequency of pain relief of at least 50%, but there was a benefit in reducing the pain at least by 30% and in the patient's global impression to be much or very much improved. Additionally, there was not a significant difference in the reduction of fatigue, in the reduction of sleep problems, nor in the improvement of health-related quality of life (187). Another meta-analysis has shown that duloxetine, pregabalin and milnacipran were superior to placebo for pain relief, while duloxetine and pregabalin were superior to milnacipran. These drugs also differed in their effects on sleep disturbances, depression and fatigue (188). A fMRI study in fibromyalgia patients treated with milnacipran and placebo demonstrated that pain reduction with milnacipran treatment was associated with decreased functional connectivity between the insular cortex and the rostral part of the anterior cingulate cortex as well as the periaqueductal gray, while these changes were not demonstrated with the placebo (189).Regarding their side effects, headaches and nausea are more common with duloxetine and milnacipran treatment, while diarrhea is more common with duloxetine treatment. Other side effects related to SNRIs include dry mouth, constipation, somnolence, dizziness and insomnia (188).

 

Monoamine Oxidase Inhibitors

 

Monoamine oxidase inhibitors block the catabolism of serotonin, increasing its levels in the brain. It has been indicated that pirlindole and moclobemide have a significant beneficial effect on pain, without a significant effect on sleep nor fatigue. Their use for the treatment of fibromyalgia patients is limited (190).

 

ANTICONVULSANTS

 

Antiepileptic medications useful for the treatment of fibromyalgia patients include pregabalin and gabapentin. Both of these medications are structurally related to GABA and they bind with high affinity to the alpha2-delta subunit site of cellular voltage-dependent calcium channels. Although their exact mechanism of action is unknown their therapeutic effects can be mediated by blocking the release of various neurotransmitters. They can be used in cases where other medications initially administered to the fibromyalgia patients become intolerable or ineffective, or as the initial treatment for patients with significant sleep disturbance in addition to pain.  

 

Pregabalin has been reported to be efficient against pain, sleep disturbances and fatigue in fibromyalgia. In a recent meta-analysis a reduction of at least 50% in pain intensity was found in 22-24% of patients taking pregabalin 300-600 mg per day, approximately 9% higher compared to the placebo group. A reduction in pain intensity of at least 30% was found in 39-43% of patients on pregabalin 300-600 mg per day, compared to 28% of patients taking placebo (191). In addition to pain, pregabalin at doses 300-600 mg per day can improve sleep and patient function, as it is demonstrated in a 2018 review of clinical trials, meta-analyses, combination studies and post-hoc analyses (192). The improvement in pain and sleep can be apparent as early as 1-2 days after the onset of treatment (193). In a 2022 network meta-analysis it was shown that treatment with pregabalin 450 mg per day was superior to duloxetine 30 mg for the reduction of pain intensity of at least 30% (178). A randomized placebo-controlled neuroimaging study demonstrated that the reduction in pain intensity from pregabalin was associated with a reduction in connectivity between the posterior insula and the default mode network (DMN) and that pregabalin but not placebo can reduce the response of the DMN to experimental pain (194). It is of interest that baseline patterns of brain connectivity have been used in a machine-learning model to successfully distinguish fibromyalgia patients who have a favourable response to pain intensity after the treatment with milnacipran from those who achieve a reduction of pain intensity after the treatment with pregabalin (195). Common side effects of pregabalin include somnolence, dizziness, weight gain and peripheral oedema. Discontinuation due to side effects is approximately 10% higher in patients treated with pregabalin compared to placebo, while discontinuation due to inefficiency of treatment is 6% lower (191). The intensity of adverse effects and the frequency of discontinuation of the treatment due to adverse effects is dose dependent. It is important for pregabalin to be titrated to the maximally tolerated therapeutic dose for each patient (192).  

 

Gabapentin has been shown to be efficient in treating fibromyalgia associated pain, while it was well tolerated (196). Side effects include dizziness, sedation, lightheadedness, and weight gain. Its efficacy and tolerability is not well studied in fibromyalgia patients, however it can be considered as an acceptable alternative in case pregabalin cannot be administered due to its cost or due to regulatory limitations (197).

 

SEDATIVE HYPNOTIC AGENTS

 

Zopiclone and zolpidem have been used in fibromyalgia. It has been suggested that they can improve the sleep and perhaps fatigue, without any significant effects on pain (144).

 

Sodium oxibate, a precursor of GABA with powerful sedative properties has been shown to improve pain, fatigue and sleep architecture in fibromyalgia (198). However, in view of safety concerns the European Medicines Agency and the US Food and Drug Administration have not approved it for use in fibromyalgia patients.

 

TRAMADOL

 

Tramadol has multiple analgesic effects, since it inhibits norepinephrine and serotonin reuptake, and its major metabolite binds weakly to opioid μ receptors (144). The use of tramadol (with or without acetaminophen) is both effective and well tolerated for the management of pain in fibromyalgia (199,200). There are some concerns regarding the long-term potential of abuse of tramadol, although the risk is less than that of more potent narcotic analgesics that have also been used in fibromyalgia.

 

Table 9. The Doses of the Most Commonly used Medications with Strong and Moderate Evidence of Effectiveness in Fibromyalgia

Drugs

Doses

Tricyclic antidepressants

Amitriptyline

Start 5-10 mg at bedtime, increase up to 25-50 mg

Cyclobenzaprine

Start 10 mg at bedtime, increase up to 30-40mg,

decrease to 5mg if 10mg too sedating

Serotonin-norepinephrine reuptake inhibitors

Duloxetine

Start 10-15mg twice daily,

gradually increased to 30 mg twice daily

Milnacipran

Start 12.5mg in the morning,

gradually increase to 50mg twice daily

Venlafaxine

167 mg per day

Anticonvulsants

Gabapentin

Start 100mg at bedtime,

increase to 1200-2400 mg per day

Pregabalin

Start 25-50mg at bedtime,

increase to 300-450 mg/day

Other

Tramadol

37.5 mg four times daily

 

CONCLUSION

 

Fibromyalgia is a common disease that is often underdiagnosed. Genetic predisposition, in combination with exposure to external stressors may lead to dysregulation of the nociceptive system and to the appearance of clinical symptoms. Fibromyalgia patients do not form a homogenous group with some patients responding adequately to current therapeutic modalities, and some others not experiencing any long-term benefit. Patients treated by primary care physicians in the community have a much better prognosis, compared to patients treated in tertiary referral centers. Certain psychological factors, such as an increased sense of control over pain, a belief that one is not disabled, that pain is not a sign of damage, and behaviors like seeking help from others, decreased guarding during examination, exercising more and having pacing activities are associated with better prognosis. Conversely, catastrophizing is associated with increased awareness of pain and with worsening symptoms.

 

REFERENCES

 

  1. Bennett RM. Clinical manifestations and diagnosis of fibromyalgia. Rheum. Dis. Clin. North Am. 2009;35(2):215–232.
  2. Wessely S, Hotopf M. Is fibromyalgia a distinct clinical entity? Historical and epidemiological evidence. Baillieres Best Pract Res Clin Rheumatol 1999;13(3):427–436.
  3. Wolfe F. Editorial: the status of fibromyalgia criteria. Arthritis & Rheumatology (Hoboken, N.J.) 2015;67(2):330–333.
  4. Weir PT, Harlan GA, Nkoy FL, Jones SS, Hegmann KT, Gren LH, Lyon JL. The incidence of fibromyalgia and its associated comorbidities: a population-based retrospective cohort study based on International Classification of Diseases, 9th Revision codes. J Clin Rheumatol 2006;12(3):124–128.
  5. Mease P, Arnold LM, Bennett R, Boonen A, Buskila D, Carville S, Chappell A, Choy E, Clauw D, Dadabhoy D, Gendreau M, Goldenberg D, Littlejohn G, Martin S, Perera P, Russell IJ, Simon L, Spaeth M, Williams D, Crofford L. Fibromyalgia syndrome. J Rheumatol 2007;34(6):1415–1425.
  6. Yunus MB. Fibromyalgia and overlapping disorders: the unifying concept of central sensitivity syndromes. Semin. Arthritis Rheum. 2007;36(6):339–356.
  7. Marcus DA, Bernstein C, Rudy TE. Fibromyalgia and headache: an epidemiological study supporting migraine as part of the fibromyalgia syndrome. Clin. Rheumatol. 2005;24(6):595–601.
  8. Rhodus NL, Fricton J, Carlson P, Messner R. Oral symptoms associated with fibromyalgia syndrome. J Rheumatol2003;30(8):1841–1845.
  9. Aydin G, Başar MM, Keleş I, Ergün G, Orkun S, Batislam E. Relationship between sexual dysfunction and psychiatric status in premenopausal women with fibromyalgia. Urology 2006;67(1):156–161.
  10. Aaron LA, Burke MM, Buchwald D. Overlapping conditions among patients with chronic fatigue syndrome, fibromyalgia, and temporomandibular disorder. Arch. Intern. Med. 2000;160(2):221–227.
  11. Giesecke T, Williams DA, Harris RE, Cupps TR, Tian X, Tian TX, Gracely RH, Clauw DJ. Subgrouping of fibromyalgia patients on the basis of pressure-pain thresholds and psychological factors. Arthritis & Rheumatism2003;48(10):2916–2922.
  12. Arnold LM, Hudson JI, Hess EV, Ware AE, Fritz DA, Auchenbach MB, Starck LO, Keck PE Jr. Family study of fibromyalgia. Arthritis Rheum. 2004;50(3):944–952.
  13. Løge-Hagen JS, Sæle A, Juhl C, Bech P, Stenager E, Mellentin AI. Prevalence of depressive disorder among patients with fibromyalgia: Systematic review and meta-analysis. J Affect Disord 2019;245:1098–1105.
  14. Kleykamp BA, Ferguson MC, McNicol E, Bixho I, Arnold LM, Edwards RR, Fillingim R, Grol-Prokopczyk H, Turk DC, Dworkin RH. The Prevalence of Psychiatric and Chronic Pain Comorbidities in Fibromyalgia: an ACTTION systematic review. Semin Arthritis Rheum 2021;51(1):166–174.
  15. Wu Y-L, Huang C-J, Fang S-C, Ko L-H, Tsai P-S. Cognitive Impairment in Fibromyalgia: A Meta-Analysis of Case-Control Studies. Psychosom Med 2018;80(5):432–438.
  16. Müller W, Schneider EM, Stratz T. The classification of fibromyalgia syndrome. Rheumatol. Int. 2007;27(11):1005–1010.
  17. Thieme K, Spies C, Sinha P, Turk DC, Flor H. Predictors of pain behaviors in fibromyalgia syndrome. Arthritis Rheum. 2005;53(3):343–350.
  18. Goldenberg DL. Fibromyalgia syndrome a decade later: What have we learned? Arch Intern Med1999;159(8):777–785.
  19. Ursini F, Ciaffi J, Mancarella L, Lisi L, Brusi V, Cavallari C, D’Onghia M, Mari A, Borlandelli E, Faranda Cordella J, La Regina M, Viola P, Ruscitti P, Miceli M, De Giorgio R, Baldini N, Borghi C, Gasbarrini A, Iagnocco A, Giacomelli R, Faldini C, Landini MP, Meliconi R. Fibromyalgia: a new facet of the post-COVID-19 syndrome spectrum? Results from a web-based survey. RMD Open 2021;7(3):e001735.
  20. Clauw DJ, Calabrese L. Rheumatology and Long COVID: lessons from the study of fibromyalgia. Ann Rheum Dis2023:ard-2023-224250.
  21. Mease P, Arnold LM, Choy EH, Clauw DJ, Crofford LJ, Glass JM, Martin SA, Morea J, Simon L, Strand CV, Williams DA. Fibromyalgia syndrome module at OMERACT 9: domain construct. J. Rheumatol. 2009;36(10):2318–2329.
  22. Mease P, Clauw D, Christensen R, Crofford L, Gendreau M, Martin S, Simon L, Strand V, Williams D, Arnold L.Toward Development of a Fibromyalgia Responder Index and Disease Activity Score: OMERACT Module Update. J Rheumatol 2011;38(7):1487–1495.
  23. Boomershine CS. A Comprehensive Evaluation of Standardized Assessment Tools in the Diagnosis of Fibromyalgia and in the Assessment of Fibromyalgia Severity. Pain Research and Treatment 2012;2012:1–11.
  24. Endresen GKM. Fibromyalgia: a rheumatologic diagnosis? Rheumatol Int 2007;27(11):999–1004.
  25. Taylor RR, Jason LA, Torres A. Fatigue rating scales: an empirical comparison. Psychol Med 2000;30(4):849–856.
  26. Whitehead L. The measurement of fatigue in chronic illness: a systematic review of unidimensional and multidimensional fatigue measures. J Pain Symptom Manage 2009;37(1):107–128.
  27. Bennett RM, Friend R, Jones KD, Ward R, Han BK, Ross RL. The Revised Fibromyalgia Impact Questionnaire (FIQR): validation and psychometric properties. Arthritis Res. Ther. 2009;11(4):R120.
  28. Chakrabarty S, Zoorob R. Fibromyalgia. Am Fam Physician 2007;76(2):247–254.
  29. Dworkin RH, Turk DC, Wyrwich KW, Beaton D, Cleeland CS, Farrar JT, Haythornthwaite JA, Jensen MP, Kerns RD, Ader DN, Brandenburg N, Burke LB, Cella D, Chandler J, Cowan P, Dimitrova R, Dionne R, Hertz S, Jadad AR, Katz NP, Kehlet H, Kramer LD, Manning DC, McCormick C, McDermott MP, McQuay HJ, Patel S, Porter L, Quessy S, Rappaport BA, Rauschkolb C, Revicki DA, Rothman M, Schmader KE, Stacey BR, Stauffer JW, von Stein T, White RE, Witter J, Zavisic S. Interpreting the clinical importance of treatment outcomes in chronic pain clinical trials: IMMPACT recommendations. J Pain 2008;9(2):105–121.
  30. Seidenberg M, Haltiner A, Taylor MA, Hermann BB, Wyler A. Development and validation of a Multiple Ability Self-Report Questionnaire. J Clin Exp Neuropsychol 1994;16(1):93–104.
  31. Farias ST, Mungas D, Jagust W. Degree of discrepancy between self and other-reported everyday functioning by cognitive status: dementia, mild cognitive impairment, and healthy elders. Int J Geriatr Psychiatry 2005;20(9):827–834.
  32. Gowers WR. A Lecture on Lumbago: Its Lessons and Analogues: Delivered at the National Hospital for the Paralysed and Epileptic. Br Med J 1904;1(2246):117–121.
  33. Smythe HA, Moldofsky H. Two contributions to understanding of the “fibrositis” syndrome. Bull Rheum Dis1977;28(1):928–931.
  34. Wolfe F, Smythe HA, Yunus MB, Bennett RM, Bombardier C, Goldenberg DL, Tugwell P, Campbell SM, Abeles M, Clark P. The American College of Rheumatology 1990 Criteria for the Classification of Fibromyalgia. Report of the Multicenter Criteria Committee. Arthritis Rheum. 1990;33(2):160–172.
  35. Fillingim RB, Bruehl S, Dworkin RH, Dworkin SF, Loeser JD, Turk DC, Widerstrom-Noga E, Arnold L, Bennett R, Edwards RR, Freeman R, Gewandter J, Hertz S, Hochberg M, Krane E, Mantyh PW, Markman J, Neogi T, Ohrbach R, Paice JA, Porreca F, Rappaport BA, Smith SM, Smith TJ, Sullivan MD, Verne GN, Wasan AD, Wesselmann U. The ACTTION-American Pain Society Pain Taxonomy (AAPT): an evidence-based and multidimensional approach to classifying chronic pain conditions. J Pain 2014;15(3):241–249.
  36. Clauw D. Time to Stop the Fibromyalgia Criteria Wars and Refocus on Identifying and Treating Individuals With This Type of Pain Earlier in Their Illness. Arthritis Care Res (Hoboken) 2021;73(5):613–616.
  37. Goldenberg DL. Diagnosing Fibromyalgia as a Disease, an Illness, a State, or a Trait? Arthritis Care Res (Hoboken) 2019;71(3):334–336.
  38. Perrot S, Dickenson AH, Bennett RM. Fibromyalgia: harmonizing science with clinical practice considerations. Pain Pract 2008;8(3):177–189.
  39. Clauw DJ. Fibromyalgia: update on mechanisms and management. J Clin Rheumatol 2007;13(2):102–109.
  40. Fitzcharles M-A, Boulos P. Inaccuracy in the diagnosis of fibromyalgia syndrome: analysis of referrals. Rheumatology (Oxford) 2003;42(2):263–267.
  41. Wolfe F, Walitt BT, Häuser W. What is fibromyalgia, how is it diagnosed, and what does it really mean? Arthritis Care Res (Hoboken) 2014;66(7):969–971.
  42. Gracely RH, Petzke F, Wolf JM, Clauw DJ. Functional magnetic resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis Rheum. 2002;46(5):1333–1343.
  43. Desmeules JA, Cedraschi C, Rapiti E, Baumgartner E, Finckh A, Cohen P, Dayer P, Vischer TL. Neurophysiologic evidence for a central sensitization in patients with fibromyalgia. Arthritis Rheum. 2003;48(5):1420–1429.
  44. Harth M, Nielson WR. The fibromyalgia tender points: use them or lose them? A brief review of the controversy. J. Rheumatol. 2007;34(5):914–922.
  45. Wolfe F, Clauw DJ, Fitzcharles M-A, Goldenberg DL, Katz RS, Mease P, Russell AS, Russell IJ, Winfield JB, Yunus MB. The American College of Rheumatology Preliminary Diagnostic Criteria for Fibromyalgia and Measurement of Symptom Severity. Arthritis Care & Research 2010;62(5):600–610.
  46. Kim SM, Lee SH, Kim HR. Applying the ACR Preliminary Diagnostic Criteria in the Diagnosis and Assessment of Fibromyalgia. Korean J Pain 2012;25(3):173–182.
  47. Ferrari R, Russell AS. A questionnaire using the modified 2010 American College of Rheumatology criteria for fibromyalgia: specificity and sensitivity in clinical practice. J. Rheumatol. 2013;40(9):1590–1595.
  48. Marcus DA, Bernstein C, Albrecht KL. Brief, self-report fibromyalgia screener evaluated in a sample of chronic pain patients. Pain Med 2013;14(5):730–735.
  49. Usui C, Hatta K, Aratani S, Yagishita N, Nishioka K, Kanazawa T, Itoh K, Yamano Y, Nakamura H, Nakajima T, Nishioka K. The Japanese version of the modified ACR preliminary diagnostic criteria for fibromyalgia and the fibromyalgia symptom scale: reliability and validity. Mod Rheumatol 2013;23(5):846–850.
  50. Bennett RM, Friend R, Marcus D, Bernstein C, Han BK, Yachoui R, Deodhar A, Kaell A, Bonafede P, Chino A, Jones KD. Criteria for the diagnosis of fibromyalgia: validation of the modified 2010 preliminary American College of Rheumatology criteria and the development of alternative criteria. Arthritis Care Res (Hoboken)2014;66(9):1364–1373.
  51. Segura-Jiménez V, Aparicio VA, Álvarez-Gallardo IC, Soriano-Maldonado A, Estévez-López F, Delgado-Fernández M, Carbonell-Baeza A. Validation of the modified 2010 American College of Rheumatology diagnostic criteria for fibromyalgia in a Spanish population. Rheumatology (Oxford) 2014;53(10):1803–1811.
  52. Carrillo-de-la-Peña MT, Triñanes Y, González-Villar A, Romero-Yuste S, Gómez-Perretta C, Arias M, Wolfe F.Convergence between the 1990 and 2010 ACR diagnostic criteria and validation of the Spanish version of the Fibromyalgia Survey Questionnaire (FSQ). Rheumatol. Int. 2015;35(1):141–151.
  53. Jones GT, Atzeni F, Beasley M, Flüß E, Sarzi-Puttini P, Macfarlane GJ. The prevalence of fibromyalgia in the general population: a comparison of the American College of Rheumatology 1990, 2010, and modified 2010 classification criteria. Arthritis & Rheumatology (Hoboken, N.J.) 2015;67(2):568–575.
  54. Staud R, Price DD, Robinson ME. The provisional diagnostic criteria for fibromyalgia: One step forward, two steps back: Comment on the article by Wolfe et al. Arthritis Care & Research 2010;62(11):1675–1676.
  55. Toda K. Preliminary diagnostic criteria for fibromyalgia should be partially revised: Comment on the article by Wolfe et al. Arthritis Care & Research 2011;63(2):308–309.
  56. Wolfe F, Clauw DJ, Fitzcharles M-A, Goldenberg DL, Häuser W, Katz RS, Mease P, Russell AS, Russell IJ, Winfield JB. Fibromyalgia criteria and severity scales for clinical and epidemiological studies: a modification of the ACR Preliminary Diagnostic Criteria for Fibromyalgia. J. Rheumatol. 2011;38(6):1113–1122.
  57. Häuser W, Schmutzer G, Brähler E, Glaesmer H. A cluster within the continuum of biopsychosocial distress can be labeled “fibromyalgia syndrome”--evidence from a representative German population survey. J. Rheumatol.2009;36(12):2806–2812.
  58. Wolfe F, Brähler E, Hinz A, Häuser W. Fibromyalgia prevalence, somatic symptom reporting, and the dimensionality of polysymptomatic distress: results from a survey of the general population. Arthritis Care Res (Hoboken) 2013;65(5):777–785.
  59. Egloff N, von Känel R, Müller V, Egle UT, Kokinogenis G, Lederbogen S, Durrer B, Stauber S. Implications of proposed fibromyalgia criteria across other functional pain syndromes. Scand. J. Rheumatol. 2015;44(5):416–424.
  60. Wolfe F, Egloff N, Häuser W. Widespread Pain and Low Widespread Pain Index Scores among Fibromyalgia-positive Cases Assessed with the 2010/2011 Fibromyalgia Criteria. J. Rheumatol. 2016;43(9):1743–1748.
  61. Wolfe F, Clauw DJ, Fitzcharles M-A, Goldenberg DL, Häuser W, Katz RL, Mease PJ, Russell AS, Russell IJ, Walitt B. 2016 Revisions to the 2010/2011 fibromyalgia diagnostic criteria. Semin. Arthritis Rheum. 2016;46(3):319–329.
  62. Arnold LM, Bennett RM, Crofford LJ, Dean LE, Clauw DJ, Goldenberg DL, Fitzcharles M-A, Paiva ES, Staud R, Sarzi-Puttini P, Buskila D, Macfarlane GJ. AAPT Diagnostic Criteria for Fibromyalgia. J Pain 2019;20(6):611–628.
  63. Wolfe F. Letter to the editor, “Fibromyalgia Criteria.” J Pain 2019;20(6):739–740.
  64. Häuser W, Brähler E, Ablin J, Wolfe F. Modified 2016 American College of Rheumatology Fibromyalgia Criteria, the Analgesic, Anesthetic, and Addiction Clinical Trial Translations Innovations Opportunities and Networks-American Pain Society Pain Taxonomy, and the Prevalence of Fibromyalgia. Arthritis Care Res (Hoboken)2021;73(5):617–625.
  65. Queiroz LP. Worldwide epidemiology of fibromyalgia. Curr Pain Headache Rep 2013;17(8):356.
  66. Neumann L, Buskila D. Epidemiology of fibromyalgia. Curr Pain Headache Rep 2003;7(5):362–368.
  67. Wolfe F, Ross K, Anderson J, Russell IJ, Hebert L. The prevalence and characteristics of fibromyalgia in the general population. Arthritis Rheum. 1995;38(1):19–28.
  68. Vincent A, Lahr BD, Wolfe F, Clauw DJ, Whipple MO, Oh TH, Barton DL, St Sauver J. Prevalence of fibromyalgia: a population-based study in Olmsted County, Minnesota, utilizing the Rochester Epidemiology Project. Arthritis Care Res (Hoboken) 2013;65(5):786–792.
  69. Nakamura I, Nishioka K, Usui C, Osada K, Ichibayashi H, Ishida M, Turk DC, Matsumoto Y, Nishioka K. An epidemiologic internet survey of fibromyalgia and chronic pain in Japan. Arthritis Care Res (Hoboken)2014;66(7):1093–1101.
  70. Amital D, Fostick L, Polliack ML, Segev S, Zohar J, Rubinow A, Amital H. Posttraumatic stress disorder, tenderness, and fibromyalgia syndrome: are they different entities? J Psychosom Res 2006;61(5):663–669.
  71. Cohen H, Jotkowitz A, Buskila D, Pelles-Avraham S, Kaplan Z, Neumann L, Sperber AD. Post-traumatic stress disorder and other co-morbidities in a sample population of patients with irritable bowel syndrome. Eur. J. Intern. Med. 2006;17(8):567–571.
  72. Fukuda K, Straus SE, Hickie I, Sharpe MC, Dobbins JG, Komaroff A. The chronic fatigue syndrome: a comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann. Intern. Med. 1994;121(12):953–959.
  73. Committee on the Diagnostic Criteria for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome, Board on the Health of Select Populations, Institute of Medicine.Beyond Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Redefining an Illness. Washington (DC): National Academies Press (US); 2015. Available at: http://www.ncbi.nlm.nih.gov/books/NBK274235/. Accessed December 16, 2019.
  74. Brandt LJ, Chey WD, Foxx-Orenstein AE, Schiller LR, Schoenfeld PS, Spiegel BM, Talley NJ, Quigley EMM. An evidence-based position statement on the management of irritable bowel syndrome. Am. J. Gastroenterol.2009;104 Suppl 1:S1-35.
  75. Hanno PM, Burks DA, Clemens JQ, Dmochowski RR, Erickson D, Fitzgerald MP, Forrest JB, Gordon B, Gray M, Mayer RD, Newman D, Nyberg L Jr, Payne CK, Wesselmann U, Faraday MM. AUA guideline for the diagnosis and treatment of interstitial cystitis/bladder pain syndrome. J. Urol. 2011;185(6):2162–2170.
  76. Abeles AM, Pillinger MH, Solitar BM, Abeles M. Narrative review: the pathophysiology of fibromyalgia. Ann. Intern. Med. 2007;146(10):726–734.
  77. Simms RW, Roy SH, Hrovat M, Anderson JJ, Skrinar G, LePoole SR, Zerbini CA, de Luca C, Jolesz F. Lack of association between fibromyalgia syndrome and abnormalities in muscle energy metabolism. Arthritis Rheum.1994;37(6):794–800.
  78. Häkkinen A, Häkkinen K, Hannonen P, Alen M. Force production capacity and acute neuromuscular responses to fatiguing loading in women with fibromyalgia are not different from those of healthy women. J. Rheumatol.2000;27(5):1277–1282.
  79. Lund E, Kendall SA, Janerot-Sjøberg B, Bengtsson A. Muscle metabolism in fibromyalgia studied by P-31 magnetic resonance spectroscopy during aerobic and anaerobic exercise. Scand. J. Rheumatol. 2003;32(3):138–145.
  80. Yunus MB. Central sensitivity syndromes: a new paradigm and group nosology for fibromyalgia and overlapping conditions, and the related issue of disease versus illness. Semin. Arthritis Rheum. 2008;37(6):339–352.
  81. Suzuki R, Rygh LJ, Dickenson AH. Bad news from the brain: descending 5-HT pathways that control spinal pain processing. Trends Pharmacol. Sci. 2004;25(12):613–617.
  82. Cagnie B, Coppieters I, Denecker S, Six J, Danneels L, Meeus M. Central sensitization in fibromyalgia? A systematic review on structural and functional brain MRI. Semin. Arthritis Rheum. 2014;44(1):68–75.
  83. Truini A, Tinelli E, Gerardi MC, Calistri V, Iannuccelli C, La Cesa S, Tarsitani L, Mainero C, Sarzi-Puttini P, Cruccu G, Caramia F, Di Franco M. Abnormal resting state functional connectivity of the periaqueductal grey in patients with fibromyalgia. Clin. Exp. Rheumatol. 2016;34(2 Suppl 96):S129-133.
  84. Fallon N, Chiu Y, Nurmikko T, Stancak A. Functional Connectivity with the Default Mode Network Is Altered in Fibromyalgia Patients. PLoS ONE 2016;11(7):e0159198.
  85. Kaplan CM, Schrepf A, Vatansever D, Larkin TE, Mawla I, Ichesco E, Kochlefl L, Harte SE, Clauw DJ, Mashour GA, Harris RE. Functional and neurochemical disruptions of brain hub topology in chronic pain. Pain2019;160(4):973–983.
  86. Martucci KT, Weber KA, Mackey SC. Altered Cervical Spinal Cord Resting-State Activity in Fibromyalgia. Arthritis & Rheumatology (Hoboken, N.J.) 2019;71(3):441–450.
  87. Schweinhardt P, Sauro KM, Bushnell MC. Fibromyalgia: A Disorder of the Brain? Neuroscientist 2008. doi:10.1177/1073858407312521.
  88. Staud R, Godfrey MM, Robinson ME. Fibromyalgia Patients Are Not Only Hypersensitive to Painful Stimuli But Also to Acoustic Stimuli. J Pain 2021;22(8):914–925.
  89. Clauw DJ, Crofford LJ. Chronic widespread pain and fibromyalgia: what we know, and what we need to know. Best Pract Res Clin Rheumatol 2003;17(4):685–701.
  90. Salemi S, Aeschlimann A, Wollina U, Gay RE, Michel BA, Gay S, Sprott H. Up-regulation of delta-opioid receptors and kappa-opioid receptors in the skin of fibromyalgia patients. Arthritis Rheum. 2007;56(7):2464–2466.
  91. Harris RE, Clauw DJ, Scott DJ, McLean SA, Gracely RH, Zubieta J-K. Decreased central mu-opioid receptor availability in fibromyalgia. J. Neurosci. 2007;27(37):10000–10006.
  92. Watkins LR, Hutchinson MR, Rice KC, Maier SF. The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol. Sci. 2009;30(11):581–591.
  93. Mhalla A, de Andrade DC, Baudic S, Perrot S, Bouhassira D. Alteration of cortical excitability in patients with fibromyalgia. Pain 2010;149(3):495–500.
  94. Foerster BR, Petrou M, Edden RAE, Sundgren PC, Schmidt-Wilcke T, Lowe SE, Harte SE, Clauw DJ, Harris RE.Reduced insular γ-aminobutyric acid in fibromyalgia. Arthritis Rheum. 2012;64(2):579–583.
  95. Harris RE, Sundgren PC, Craig AD, Kirshenbaum E, Sen A, Napadow V, Clauw DJ. Elevated insular glutamate in fibromyalgia is associated with experimental pain. Arthritis Rheum. 2009;60(10):3146–3152.
  96. Valdés M, Collado A, Bargalló N, Vázquez M, Rami L, Gómez E, Salamero M. Increased glutamate/glutamine compounds in the brains of patients with fibromyalgia: a magnetic resonance spectroscopy study. Arthritis Rheum.2010;62(6):1829–1836.
  97. Haas L, Portela LVC, Böhmer AE, Oses JP, Lara DR. Increased plasma levels of brain derived neurotrophic factor (BDNF) in patients with fibromyalgia. Neurochem. Res. 2010;35(5):830–834.
  98. O’Mahony LF, Srivastava A, Mehta P, Ciurtin C. Is fibromyalgia associated with a unique cytokine profile? A systematic review and meta-analysis. Rheumatology (Oxford) 2021;60(6):2602–2614.
  99. Uçeyler N, Häuser W, Sommer C. Systematic review with meta-analysis: cytokines in fibromyalgia syndrome. BMC Musculoskelet Disord 2011;12:245.
  100. Salemi S, Rethage J, Wollina U, Michel BA, Gay RE, Gay S, Sprott H. Detection of interleukin 1beta (IL-1beta), IL-6, and tumor necrosis factor-alpha in skin of patients with fibromyalgia. J. Rheumatol. 2003;30(1):146–150.
  101. Üçeyler N, Valenza R, Stock M, Schedel R, Sprotte G, Sommer C. Reduced levels of antiinflammatory cytokines in patients with chronic widespread pain. Arthritis & Rheumatism 2006;54(8):2656–2664.
  102. Sturgill J, McGee E, Menzies V. Unique cytokine signature in the plasma of patients with fibromyalgia. J Immunol Res 2014;2014:938576.
  103. Sommer C, Kress M. Recent findings on how proinflammatory cytokines cause pain: peripheral mechanisms in inflammatory and neuropathic hyperalgesia. Neurosci. Lett. 2004;361(1–3):184–187.
  104. Staud R. Fibromyalgia pain: do we know the source? Curr Opin Rheumatol 2004;16(2):157–163.
  105. Tsigos C, Chrousos GP. Hypothalamic-pituitary-adrenal axis, neuroendocrine factors and stress. J Psychosom Res2002;53(4):865–871.
  106. Mastorakos G, Pavlatou M, Diamanti-Kandarakis E, Chrousos GP. Exercise and the stress system. Hormones (Athens) 2005;4(2):73–89.
  107. Beiner E, Lucas V, Reichert J, Buhai D-V, Jesinghaus M, Vock S, Drusko A, Baumeister D, Eich W, Friederich H-C, Tesarz J. Stress biomarkers in individuals with fibromyalgia syndrome: a systematic review with meta-analysis. Pain 2023;164(7):1416–1427.
  108. Crofford LJ, Young EA, Engleberg NC, Korszun A, Brucksch CB, McClure LA, Brown MB, Demitrack MA. Basal circadian and pulsatile ACTH and cortisol secretion in patients with fibromyalgia and/or chronic fatigue syndrome. Brain Behav. Immun. 2004;18(4):314–325.
  109. Wingenfeld K, Heim C, Schmidt I, Wagner D, Meinlschmidt G, Hellhammer DH.HPA axis reactivity and lymphocyte glucocorticoid sensitivity in fibromyalgia syndrome and chronic pelvic pain. Psychosom Med2008;70(1):65–72.
  110. Crofford LJ, Pillemer SR, Kalogeras KT, Cash JM, Michelson D, Kling MA, Sternberg EM, Gold PW, Chrousos GP, Wilder RL. Hypothalamic-pituitary-adrenal axis perturbations in patients with fibromyalgia. Arthritis Rheum.1994;37(11):1583–1592.
  111. Wingenfeld K, Wagner D, Schmidt I, Meinlschmidt G, Hellhammer DH, Heim C. The low-dose dexamethasone suppression test in fibromyalgia. J Psychosom Res 2007;62(1):85–91.
  112. Jones KD, Deodhar P, Lorentzen A, Bennett RM, Deodhar AA. Growth hormone perturbations in fibromyalgia: a review. Semin. Arthritis Rheum. 2007;36(6):357–379.
  113. Ablin J, Neumann L, Buskila D. Pathogenesis of fibromyalgia - a review. Joint Bone Spine 2008;75(3):273–279.
  114. Riva R, Mork PJ, Westgaard RH, Okkenhaug Johansen T, Lundberg U. Catecholamines and heart rate in female fibromyalgia patients. J Psychosom Res 2012;72(1):51–57.
  115. Kadetoff D, Kosek E. Evidence of reduced sympatho-adrenal and hypothalamic-pituitary activity during static muscular work in patients with fibromyalgia. J Rehabil Med 2010;42(8):765–772.
  116. Lerma C, Martinez A, Ruiz N, Vargas A, Infante O, Martinez-Lavin M. Nocturnal heart rate variability parameters as potential fibromyalgia biomarker: correlation with symptoms severity. Arthritis Res. Ther. 2011;13(6):R185.
  117. Cohen H, Neumann L, Shore M, Amir M, Cassuto Y, Buskila D. Autonomic dysfunction in patients with fibromyalgia: application of power spectral analysis of heart rate variability. Semin. Arthritis Rheum.2000;29(4):217–227.
  118. Cohen H, Neumann L, Alhosshle A, Kotler M, Abu-Shakra M, Buskila D. Abnormal sympathovagal balance in men with fibromyalgia. J. Rheumatol. 2001;28(3):581–589.
  119. Torpy DJ, Papanicolaou DA, Lotsikas AJ, Wilder RL, Chrousos GP, Pillemer SR. Responses of the sympathetic nervous system and the hypothalamic-pituitary-adrenal axis to interleukin-6: a pilot study in fibromyalgia. Arthritis Rheum. 2000;43(4):872–880.
  120. Maekawa K, Twe C, Lotaif A, Chiappelli F, Clark GT. Function of beta-adrenergic receptors on mononuclear cells in female patients with fibromyalgia. J. Rheumatol. 2003;30(2):364–368.
  121. Giesecke T, Gracely RH, Williams DA, Geisser ME, Petzke FW, Clauw DJ. The relationship between depression, clinical pain, and experimental pain in a chronic pain cohort. Arthritis Rheum. 2005;52(5):1577–1584.
  122. Lee YH, Choi SJ, Ji JD, Song GG. Candidate gene studies of fibromyalgia: a systematic review and meta-analysis. Rheumatol Int 2012;32(2):417–426.
  123. Smith SB, Maixner DW, Fillingim RB, Slade G, Gracely RH, Ambrose K, Zaykin DV, Hyde C, John S, Tan K, Maixner W, Diatchenko L. Large candidate gene association study reveals genetic risk factors and therapeutic targets for fibromyalgia. Arthritis Rheum 2012;64(2):584–593.
  124. Arnold LM, Fan J, Russell IJ, Yunus MB, Khan MA, Kushner I, Olson JM, Iyengar SK. The fibromyalgia family study: a genome-wide linkage scan study. Arthritis Rheum 2013;65(4):1122–1128.
  125. Docampo E, Escaramís G, Gratacòs M, Villatoro S, Puig A, Kogevinas M, Collado A, Carbonell J, Rivera J, Vidal J, Alegre J, Estivill X, Rabionet R. Genome-wide analysis of single nucleotide polymorphisms and copy number variants in fibromyalgia suggest a role for the central nervous system. Pain 2014;155(6):1102–1109.
  126. Dutta D, Brummett CM, Moser SE, Fritsche LG, Tsodikov A, Lee S, Clauw DJ, Scott LJ. Heritability of the Fibromyalgia Phenotype Varies by Age. Arthritis Rheumatol 2020;72(5):815–823.
  127. Offenbaecher M, Bondy B, de Jonge S, Glatzeder K, Krüger M, Schoeps P, Ackenheil M. Possible association of fibromyalgia with a polymorphism in the serotonin transporter gene regulatory region. Arthritis Rheum.1999;42(11):2482–2488.
  128. Cohen H, Buskila D, Neumann L, Ebstein RP.Confirmation of an association between fibromyalgia and serotonin transporter promoter region (5- HTTLPR) polymorphism, and relationship to anxiety-related personality traits. Arthritis Rheum. 2002;46(3):845–847.
  129. Gursoy S. Absence of association of the serotonin transporter gene polymorphism with the mentally healthy subset of fibromyalgia patients. Clin. Rheumatol. 2002;21(3):194–197.
  130. Buskila D, Dan B, Cohen H, Hagit C, Neumann L, Lily N, Ebstein RP. An association between fibromyalgia and the dopamine D4 receptor exon III repeat polymorphism and relationship to novelty seeking personality traits. Mol. Psychiatry 2004;9(8):730–731.
  131. Gürsoy S, Erdal E, Herken H, Madenci E, Alaşehirli B, Erdal N. Significance of catechol-O-methyltransferase gene polymorphism in fibromyalgia syndrome. Rheumatol. Int. 2003;23(3):104–107.
  132. Zhang L, Zhu J, Chen Y, Zhao J. Meta-analysis reveals a lack of association between a common catechol-O-methyltransferase (COMT) polymorphism val158met and fibromyalgia. Int J Clin Exp Pathol 2014;7(12):8489–8497.
  133. Finan PH, Zautra AJ, Davis MC, Lemery-Chalfant K, Covault J, Tennen H. Genetic influences on the dynamics of pain and affect in fibromyalgia. Health Psychol 2010;29(2):134–142.
  134. Solak Ö, Erdoğan MÖ, Yıldız H, Ulaşlı AM, Yaman F, Terzi ESA, Ulu S, Dündar Ü, Solak M. Assessment of opioid receptor μ1 gene A118G polymorphism and its association with pain intensity in patients with fibromyalgia. Rheumatol. Int. 2014;34(9):1257–1261.
  135. Vargas-Alarcón G, Fragoso J-M, Cruz-Robles D, Vargas A, Martinez A, Lao-Villadóniga J-I, García-Fructuoso F, Vallejo M, Martínez-Lavín M. Association of adrenergic receptor gene polymorphisms with different fibromyalgia syndrome domains. Arthritis Rheum. 2009;60(7):2169–2173.
  136. Üçeyler N, Zeller D, Kahn A-K, Kewenig S, Kittel-Schneider S, Schmid A, Casanova-Molla J, Reiners K, Sommer C. Small fibre pathology in patients with fibromyalgia syndrome. Brain 2013;136(Pt 6):1857–1867.
  137. Giannoccaro MP, Donadio V, Incensi A, Avoni P, Liguori R. Small nerve fiber involvement in patients referred for fibromyalgia. Muscle Nerve 2014;49(5):757–759.
  138. Grayston R, Czanner G, Elhadd K, Goebel A, Frank B, Üçeyler N, Malik RA, Alam U. A systematic review and meta-analysis of the prevalence of small fiber pathology in fibromyalgia: Implications for a new paradigm in fibromyalgia etiopathogenesis. Semin Arthritis Rheum 2019;48(5):933–940.
  139. Clauw DJ, Chrousos GP. Chronic pain and fatigue syndromes: overlapping clinical and neuroendocrine features and potential pathogenic mechanisms. Neuroimmunomodulation 1997;4(3):134–153.
  140. Buskila D, Atzeni F, Sarzi-Puttini P. Etiology of fibromyalgia: the possible role of infection and vaccination. Autoimmun Rev 2008;8(1):41–43.
  141. Buskila D, Neumann L, Vaisberg G, Alkalay D, Wolfe F. Increased rates of fibromyalgia following cervical spine injury. A controlled study of 161 cases of traumatic injury. Arthritis Rheum. 1997;40(3):446–452.
  142. Wynne-Jones G, Jones GT, Wiles NJ, Silman AJ, Macfarlane GJ. Predicting new onset of widespread pain following a motor vehicle collision. J. Rheumatol. 2006;33(5):968–974.
  143. Peterson EL. Fibromyalgia--management of a misunderstood disorder. J Am Acad Nurse Pract 2007;19(7):341–348.
  144. Sarzi-Puttini P, Buskila D, Carrabba M, Doria A, Atzeni F. Treatment strategy in fibromyalgia syndrome: where are we now? Semin. Arthritis Rheum. 2008;37(6):353–365.
  145. Macfarlane GJ, Kronisch C, Dean LE, Atzeni F, Häuser W, Fluß E, Choy E, Kosek E, Amris K, Branco J, Dincer F, Leino-Arjas P, Longley K, McCarthy GM, Makri S, Perrot S, Sarzi-Puttini P, Taylor A, Jones GT. EULAR revised recommendations for the management of fibromyalgia. Ann. Rheum. Dis. 2017;76(2):318–328.
  146. Burckhardt CS, Bjelle A. Education programmes for fibromyalgia patients: description and evaluation. Baillieres Clin Rheumatol 1994;8(4):935–955.
  147. White KP, Nielson WR, Harth M, Ostbye T, Speechley M. Does the label “fibromyalgia” alter health status, function, and health service utilization? A prospective, within-group comparison in a community cohort of adults with chronic widespread pain. Arthritis Rheum. 2002;47(3):260–265.
  148. Ehrlich GE. Pain is real; fibromyalgia isn’t. J. Rheumatol. 2003;30(8):1666–1667.
  149. Atzeni F, Alciati A, Salaffi F, Di Carlo M, Bazzichi L, Govoni M, Biasi G, Di Franco M, Mozzani F, Gremese E, Dagna L, Batticciotto A, Fischetti F, Giacomelli R, Guiducci S, Guggino G, Bentivegna M, Gerli R, Salvarani C, Bajocchi G, Ghini M, Iannone F, Giorgi V, Farah S, Bonazza S, Barbagli S, Gioia C, Marino NG, Capacci A, Cavalli G, Cappelli A, Carubbi F, Nacci F, Riccucci I, Cutolo M, Sinigaglia L, Sarzi-Puttini P. The association between body mass index and fibromyalgia severity: data from a cross-sectional survey of 2339 patients. Rheumatol Adv Pract2021;5(1):rkab015.
  150. Busch AJ, Webber SC, Brachaniec M, Bidonde J, Bello-Haas VD, Danyliw AD, Overend TJ, Richards RS, Sawant A, Schachter CL. Exercise therapy for fibromyalgia. Curr Pain Headache Rep 2011;15(5):358–367.
  151. McDowell CP, Cook DB, Herring MP. The Effects of Exercise Training on Anxiety in Fibromyalgia Patients: A Meta-analysis. Med Sci Sports Exerc 2017;49(9):1868–1876.
  152. Andrade A, Dominski FH, Sieczkowska SM. What we already know about the effects of exercise in patients with fibromyalgia: An umbrella review. Semin Arthritis Rheum 2020;50(6):1465–1480.
  153. Estévez-López F, Maestre-Cascales C, Russell D, Álvarez-Gallardo IC, Rodriguez-Ayllon M, Hughes CM, Davison GW, Sañudo B, McVeigh JG. Effectiveness of Exercise on Fatigue and Sleep Quality in Fibromyalgia: A Systematic Review and Meta-analysis of Randomized Trials. Arch Phys Med Rehabil 2021;102(4):752–761.
  154. Bidonde J, Busch AJ, Webber SC, Schachter CL, Danyliw A, Overend TJ, Richards RS, Rader T. Aquatic exercise training for fibromyalgia. Cochrane Database Syst Rev 2014;(10):CD011336.
  155. Salaffi F, Di Carlo M, Farah S, Marotto D, Giorgi V, Sarzi-Puttini P. Exercise therapy in fibromyalgia patients: comparison of a web-based intervention with usual care. Clin Exp Rheumatol 2020;38 Suppl 123(1):86–93.
  156. Bernardy K, Klose P, Busch AJ, Choy EHS, Häuser W. Cognitive behavioural therapies for fibromyalgia. Cochrane Database Syst Rev 2013;(9):CD009796.
  157. Mascarenhas RO, Souza MB, Oliveira MX, Lacerda AC, Mendonça VA, Henschke N, Oliveira VC. Association of Therapies With Reduced Pain and Improved Quality of Life in Patients With Fibromyalgia: A Systematic Review and Meta-analysis. JAMA Intern Med 2021;181(1):104–112.
  158. Jensen KB, Kosek E, Wicksell R, Kemani M, Olsson G, Merle JV, Kadetoff D, Ingvar M. Cognitive Behavioral Therapy increases pain-evoked activation of the prefrontal cortex in patients with fibromyalgia. Pain2012;153(7):1495–1503.
  159. Lazaridou A, Kim J, Cahalan CM, Loggia ML, Franceschelli O, Berna C, Schur P, Napadow V, Edwards RR.Effects of Cognitive-Behavioral Therapy (CBT) on Brain Connectivity Supporting Catastrophizing in Fibromyalgia. Clin J Pain 2017;33(3):215–221.
  160. Deare JC, Zheng Z, Xue CCL, Liu JP, Shang J, Scott SW, Littlejohn G. Acupuncture for treating fibromyalgia. Cochrane Database Syst Rev 2013;(5):CD007070.
  161. Langhorst J, Klose P, Dobos GJ, Bernardy K, Häuser W. Efficacy and safety of meditative movement therapies in fibromyalgia syndrome: a systematic review and meta-analysis of randomized controlled trials. Rheumatol. Int.2013;33(1):193–207.
  162. Langhorst J, Musial F, Klose P, Häuser W. Efficacy of hydrotherapy in fibromyalgia syndrome--a meta-analysis of randomized controlled clinical trials. Rheumatology (Oxford) 2009;48(9):1155–1159.
  163. Schmidt-Wilcke T, Clauw DJ. Fibromyalgia: from pathophysiology to therapy. Nat Rev Rheumatol 2011;7(9):518–527.
  164. Boomershine CS, Crofford LJ. A symptom-based approach to pharmacologic management of fibromyalgia. Nat Rev Rheumatol 2009;5(4):191–199.
  165. Goldenberg DL BC. MAnagement of fibromyalgia syndrome. JAMA 2004;292(19):2388–2395.
  166. Chou R, Turner JA, Devine EB, Hansen RN, Sullivan SD, Blazina I, Dana T, Bougatsos C, Deyo RA. The effectiveness and risks of long-term opioid therapy for chronic pain: a systematic review for a National Institutes of Health Pathways to Prevention Workshop. Ann Intern Med 2015;162(4):276–286.
  167. Hwang J-M, Lee B-J, Oh TH, Park D, Kim C-H. Association between initial opioid use and response to a brief interdisciplinary treatment program in fibromyalgia. Medicine (Baltimore) 2019;98(1):e13913.
  168. Curtis AF, Miller MB, Rathinakumar H, Robinson M, Staud R, Berry RB, McCrae CS. Opioid use, pain intensity, age, and sleep architecture in patients with fibromyalgia and insomnia. Pain 2019;160(9):2086–2092.
  169. Abeles M, Solitar BM, Pillinger MH, Abeles AM. Update on fibromyalgia therapy. Am. J. Med. 2008;121(7):555–561.
  170. Liu Y, Qian C, Yang M. Treatment Patterns Associated with ACR-Recommended Medications in the Management of Fibromyalgia in the United States. J Manag Care Spec Pharm 2016;22(3):263–271.
  171. Tofferi JK, Jackson JL, O’Malley PG. Treatment of fibromyalgia with cyclobenzaprine: A meta-analysis. Arthritis Rheum. 2004;51(1):9–13.
  172. O’Malley PG, Balden E, Tomkins G, Santoro J, Kroenke K, Jackson JL. Treatment of fibromyalgia with antidepressants: a meta-analysis. J Gen Intern Med 2000;15(9):659–666.
  173. Arnold LM, Keck PE Jr, Welge JA.Antidepressant treatment of fibromyalgia. A meta-analysis and review. Psychosomatics 2000;41(2):104–113.
  174. Carette S, Bell MJ, Reynolds WJ, Haraoui B, McCain GA, Bykerk VP, Edworthy SM, Baron M, Koehler BE, Fam AG. Comparison of amitriptyline, cyclobenzaprine, and placebo in the treatment of fibromyalgia. A randomized, double-blind clinical trial. Arthritis Rheum. 1994;37(1):32–40.
  175. Carette S, Oakson G, Guimont C, Steriade M. Sleep electroencephalography and the clinical response to amitriptyline in patients with fibromyalgia. Arthritis Rheum. 1995;38(9):1211–1217.
  176. Uçeyler N, Häuser W, Sommer C. A systematic review on the effectiveness of treatment with antidepressants in fibromyalgia syndrome. Arthritis Rheum. 2008;59(9):1279–1298.
  177. Häuser W, Petzke F, Üçeyler N, Sommer C. Comparative efficacy and acceptability of amitriptyline, duloxetine and milnacipran in fibromyalgia syndrome: a systematic review with meta-analysis. Rheumatology (Oxford)2011;50(3):532–543.
  178. Alberti FF, Becker MW, Blatt CR, Ziegelmann PK, da Silva Dal Pizzol T, Pilger D. Comparative efficacy of amitriptyline, duloxetine and pregabalin for treating fibromyalgia in adults: an overview with network meta-analysis. Clin Rheumatol 2022;41(7):1965–1978.
  179. Goldenberg D, Mayskiy M, Mossey C, Ruthazer R, Schmid C. A randomized, double-blind crossover trial of fluoxetine and amitriptyline in the treatment of fibromyalgia. Arthritis Rheum. 1996;39(11):1852–1859.
  180. Moldofsky H, Harris HW, Archambault WT, Kwong T, Lederman S. Effects of bedtime very low dose cyclobenzaprine on symptoms and sleep physiology in patients with fibromyalgia syndrome: a double-blind randomized placebo-controlled study. J. Rheumatol. 2011;38(12):2653–2663.
  181. Arnold LM, Gendreau RM, Palmer RH, Gendreau JF, Wang Y. Efficacy and safety of milnacipran 100 mg/day in patients with fibromyalgia: results of a randomized, double-blind, placebo-controlled trial. Arthritis Rheum.2010;62(9):2745–2756.
  182. Branco JC, Zachrisson O, Perrot S, Mainguy Y. A European multicenter randomized double-blind placebo-controlled monotherapy clinical trial of milnacipran in treatment of fibromyalgia. J. Rheumatol. 2010;37(4):851–859.
  183. Sayar K, Aksu G, Ak I, Tosun M. Venlafaxine treatment of fibromyalgia. Ann Pharmacother 2003;37(11):1561–1565.
  184. Lunn MPT, Hughes RAC, Wiffen PJ. Duloxetine for treating painful neuropathy, chronic pain or fibromyalgia. Cochrane Database Syst Rev 2014;1:CD007115.
  185. Arnold LM, Zhang S, Pangallo BA. Efficacy and safety of duloxetine 30 mg/d in patients with fibromyalgia: a randomized, double-blind, placebo-controlled study. Clin J Pain 2012;28(9):775–781.
  186. Russell IJ, Mease PJ, Smith TR, Kajdasz DK, Wohlreich MM, Detke MJ, Walker DJ, Chappell AS, Arnold LM.Efficacy and safety of duloxetine for treatment of fibromyalgia in patients with or without major depressive disorder: Results from a 6-month, randomized, double-blind, placebo-controlled, fixed-dose trial. Pain 2008;136(3):432–444.
  187. Welsch P, Üçeyler N, Klose P, Walitt B, Häuser W. Serotonin and noradrenaline reuptake inhibitors (SNRIs) for fibromyalgia. Cochrane Database Syst Rev 2018;2:CD010292.
  188. Häuser W, Petzke F, Sommer C. Comparative efficacy and harms of duloxetine, milnacipran, and pregabalin in fibromyalgia syndrome. J Pain 2010;11(6):505–521.
  189. Schmidt-Wilcke T, Ichesco E, Hampson JP, Kairys A, Peltier S, Harte S, Clauw DJ, Harris RE. Resting state connectivity correlates with drug and placebo response in fibromyalgia patients. Neuroimage Clin 2014;6:252–261.
  190. Häuser W, Bernardy K, Uçeyler N, Sommer C. Treatment of fibromyalgia syndrome with antidepressants: a meta-analysis. JAMA 2009;301(2):198–209.
  191. Derry S, Cording M, Wiffen PJ, Law S, Phillips T, Moore RA. Pregabalin for pain in fibromyalgia in adults. Cochrane Database Syst Rev 2016;9(9):CD011790.
  192. Arnold LM, Choy E, Clauw DJ, Oka H, Whalen E, Semel D, Pauer L, Knapp L. An evidence-based review of pregabalin for the treatment of fibromyalgia. Curr Med Res Opin 2018;34(8):1397–1409.
  193. Arnold LM, Emir B, Pauer L, Resnick M, Clair A. Time to improvement of pain and sleep quality in clinical trials of pregabalin for the treatment of fibromyalgia. Pain Med 2015;16(1):176–185.
  194. Harris RE, Napadow V, Huggins JP, Pauer L, Kim J, Hampson J, Sundgren PC, Foerster B, Petrou M, Schmidt-Wilcke T, Clauw DJ. Pregabalin rectifies aberrant brain chemistry, connectivity, and functional response in chronic pain patients. Anesthesiology 2013;119(6):1453–1464.
  195. Ichesco E, Peltier SJ, Mawla I, Harper DE, Pauer L, Harte SE, Clauw DJ, Harris RE. Prediction of Differential Pharmacologic Response in Chronic Pain Using Functional Neuroimaging Biomarkers and a Support Vector Machine Algorithm: An Exploratory Study. Arthritis Rheumatol 2021;73(11):2127–2137.
  196. Arnold LM, Goldenberg DL, Stanford SB, Lalonde JK, Sandhu HS, Keck PE Jr, Welge JA, Bishop F, Stanford KE, Hess EV, Hudson JI. Gabapentin in the treatment of fibromyalgia: a randomized, double-blind, placebo-controlled, multicenter trial. Arthritis Rheum. 2007;56(4):1336–1344.
  197. Cooper TE, Derry S, Wiffen PJ, Moore RA. Gabapentin for fibromyalgia pain in adults. Cochrane Database Syst Rev 2017;1(1):CD012188.
  198. Perrot S, Russell IJ. More ubiquitous effects from non-pharmacologic than from pharmacologic treatments for fibromyalgia syndrome: a meta-analysis examining six core symptoms. Eur J Pain 2014;18(8):1067–1080.
  199. Roskell NS, Beard SM, Zhao Y, Le TK. A meta-analysis of pain response in the treatment of fibromyalgia. Pain Pract 2011;11(6):516–527.
  200. MacLean AJB, Schwartz TL. Tramadol for the treatment of fibromyalgia. Expert Rev Neurother 2015;15(5):469–475.

 

Immune System Effects on the Endocrine System

ABSTRACT

 

Among the most important and complex systems in the human body are the endocrine and immune systems. Emerging research over the last decade has shed light on their remarkable interplay, revealing a multitude of bidirectional communication pathways and reciprocal regulation mechanisms. Endocrine diseases, such as autoimmune thyroiditis, diabetes mellitus type 1 and type 2, osteoporosis, and disorders of the hypothalamic-pituitary-adrenal (HPA) axis, as well as endocrine malignancies, such as thyroid cancer, are highly interconnected with dysregulations of the immune system.Thus, multiple cytokines, chemokines, and evolving inflammatory processes are involved in the pathogenesis of immune-related endocrine disorders, providing potential targets for immune-based therapeutic approaches. In this chapter, we provide a comprehensive overview of the molecular mechanisms underlying these complex endocrine-immune interactions, and discuss the implications of immune system function or dysfunction in endocrine disorders.

 

INTRODUCTION

 

The immune system is a host defense system that comprises numerous biological structures and processes to defend the human body against potentially harmful substances and invading pathogens. It functions through a series of coordinated mechanisms, including innate and adaptive immunity. The innate immune response consists of i) phagocytosis by macrophages, neutrophils, monocytes, and dendritic cells, and ii) cytotoxicity by natural killer cells, providing an immediate, nonspecific defense against a wide range of invaders by recognizing common patterns shared by many pathogens (1).

 

Adaptive immunity, on the other hand, is an acquired, specially designed defense system; it develops over time and utilizes highly specialized immune cells involving antibody-dependent complement or cell-mediated cytotoxicity produced by T cells that recognize injurious agents, such as heat shock proteins or microbial antigens. During adaptive immunity, antigens taken up by antigen-presenting cells (APCs) are presented to T cells through binding with major histocompatibility complex (MHC) molecules on the surface of these cells. Activated CD4 helper T cells stimulate the release of cytokines, such as interleukin (IL)-2, which i) induce T cell proliferation and activation, ii) stimulate killer cell activity by CD8 suppressor T cells, and iii) activate B cells to differentiate into plasma cells and produce antibodies. Naïve T cells differentiate mainly into two main subsets that produce a different set of cytokines and regulate distinct immune functions. T-helper 1 (Th1) cells produce mainly interferon-γ (IFN)-γ, tumor necrosis factor-α (TNF)-, and IL-12 to regulate cell-mediated responses, while T-helper 2 (Th2) cells secrete IL-4, IL-5, and IL-13, to stimulate antibody production. In addition, three other subsets of T helper cells have been identified: Th22, which secrete IL-22, Th17, which secrete IL-17, and Treg, which secrete transforming growth factor (TGF)- b, all of which also play a role in the pathogenesis of autoimmune diseases (2).

 

Multiple regulatory mechanisms are involved in maintaining central and peripheral T and B cell tolerance (1). Defects in the processes that ensure immune cell tolerance may induce a maladaptive immune response to a self-antigen and lead to the development of autoimmune diseases.

 

Emerging research of the last few decades has shed light on the remarkable interplay between endocrine and immune systems, revealing a multitude of bidirectional communication pathways and reciprocal regulation.

 

Autoimmune endocrine diseases, such as Hashimoto thyroiditis, diabetes mellitus type 1 (DM1), and Addison disease, as well as endocrine malignancies, such as differentiated, anaplastic, and medullary thyroid cancer (3), and other endocrine disorders, including diabetes mellitus type 2 (DM2), osteoporosis, as well as a dysfunctional hypothalamic-pituitary-adrenal (HPA) axis response to stress and inflammation, are highly interconnected with dysregulations of the immune system.

 

This chapter seeks to elucidate the interplay between the endocrine and immune systems, exploring their interconnections and highlighting the impact of their crosstalk on health and disease. We present a comprehensive overview of the molecular mechanisms underlying this interaction and discuss the potential therapeutic implications of targeting the immune system for the management of endocrine diseases.

 

IMMUNE SYSTEM AND THYROID DISEASE

 

Autoimmune Thyroid Disease

 

Autoimmune thyroid disease (AITD) results from a dysregulation of the immune system that leads to loss of tolerance to thyroid antigens and to an autoimmune attack on the thyroid gland. The most common clinical manifestations of AITD are Hashimoto's thyroiditis and Graves’ disease, while less prevalent manifestations are drug-induced thyroiditis, postpartum thyroiditis, or thyroiditis associated with polyglandular syndromes (i.e., autoimmune polyglandular syndromes type 1 and type 2). The underlying molecular mechanisms of AITD involve both circulating autoantibodies and T cell immune mechanisms, while genetic background, as well as cross-reactivity to external antigens (4-6), are also implicated.

 

There are three major thyroid autoantigens that are targeted during autoimmune thyroid attack and are critical for thyroid homeostasis, namely, thyroglobulin (Tg), thyroid peroxidase (TPO), and the thyrotropin receptor (TSHR) (Figure 1).

 

Figure 1. Thyroid proteins that serve as autoantigens. Thyroglobulin (Tg) function as storage protein in thyroid cells, playing a critical role in the synthesis and release of thyroid hormones. Thyroid peroxidase (TPO) catalyzes iodination of tyrosines in thyroglobulin, which attaches one or two iodine molecules to form monoiodotyrosine (MIT) or diiodotyrosine (DIT), respectively. In addition, thyroid peroxidase catalyzes the coupling of iodotyrosine residues to form triiodothyronine (T3) and thyroxine (T4) attached to thyroglobulin. Thyrotropin receptor (TSHR) is a transmembrane G-protein coupled receptor that upon stimulation by circulating TSH activates the expression of downstream effector genes to regulate thyroid growth, thyrocyte differentiation, and thyroid hormone synthesis. Sodium/iodide symporter (NIS) is a membrane glycoprotein, which actively cotransports two sodium cations per each iodide anion, using the electrochemical sodium gradient generated by the Na+/K+-ATPase. Pendrin is involved in the apical iodide efflux in thyroid cells. It can also exchange chloride and bicarbonate. [Modified by Boguslawska et al (7)].

 

Thyroglobulin is a soluble glycoprotein homodimer composed of two subunits of ~330 kDa in size and is the most abundant glycoprotein in the thyroid gland. It is the scaffold for the synthesis of thyroid hormones and the storage-form of thyroid hormones inside the gland. Recently, researchers described the first atomic structure of full-length Tg and identified its hormone-forming tyrosine residues (8). Anti-Tg antibodies (TgAb) act mainly through antibody-dependent cytotoxicity cells rather than through complement fixation (7). In AITD, the prevalent TgAb species recognize native rather than denatured antigens and bind to a number of overlapping epitopic domains located mainly in the central region and C-terminal end of Tg. Of note, TgAb in the serum of healthy subjects have a different epitopic pattern (9).

 

Another major thyroid antigen is TPO, a glycosylated heme-containing homodimer of two 107-kDa transmembrane subunits located in the apical membrane of thyrocytes (Figure 1). It catalyzes the iodination of tyrosyl residues in Tg and the coupling of iodotyrosine residues to form iodothyronines attached to Tg (10). Both humoral and cellular immune responses are directed against TPO. TPO autoantibodies (TPOAb) occur in almost all patients with Hashimoto thyroiditis and approximately 75% of individuals with Graves’ disease (11). In addition, TPOAb may be involved in autoimmune thyroid cell death via antibody-dependent cytotoxic cells and C3 complement-mediated cytotoxicity (7). Specific patterns of TPOAb recognition remain stable in an individual over time and are genetically transmitted through family lineages (12).

 

Thyrotropin receptor is primarily expressed on the basolateral membrane of thyrocytes (Figure 1) and belongs to the transmembrane G protein-coupled receptor family. The intracellular signaling pathway that is activated by the interaction of TSH with TSHR is indispensable for the synthesis of thyroid hormones and the proliferation of follicular epithelial cells. TSHR-stimulating antibodies (TSAb) act as TSHR agonists and stimulate thyroid growth and production of thyroid hormone in an autonomous and unregulated manner. On the other hand, TSHR-blocking antibodies (TBAb) act as antagonists that block the intracellular signaling of TSH, leading to decrease of thyroid hormone synthesis and subsequently hypothyroidism. Neutral antibodies to TSHR, which may also be detected in the serum of patients with Graves’ disease, bind to the receptor but do not alter its activity (13). The exact antigenic sites of TSHR-specific TBAb and TSAb overlap and are mostly directed to the extracellular A subunit of the receptor (Figure 1) (14,15).

 

Other thyroid antigens include the iodide transporters, sodium iodide symporter (NIS), and pendrin (Figure 1). The presence of NIS antibodies is increased in AITDs, especially in patients with Graves’ disease, whereas their expression in euthyroid individuals is rare (16). Pendrin is an apical membrane-bound iodide transporter, but the diagnostic value of antibodies against pendrin is rather low (16).

 

Although the above-described circulating autoantibodies are useful markers of thyroid autoimmunity, it is the T cell immune mechanism, i.e., the loss of immune self-tolerance, that is the core of AITD pathophysiology. Loss of immune self-tolerance may result from either: i) the loss of central tolerance (i.e., disturbed deletion of autoreactive T cells in the thymus), ii) dysfunction of peripheral tolerance (i.e., impaired apoptosis of self-reactive T cells and inhibition of the activity of T-regulatory cells), or iii) disturbed energy (i.e., disturbance of the functional inactivation that prevents the lymphocytes from activating an immune reaction against the antigen).

 

Patients with AITD express IFN-γ-induced MHC class II molecules, which promotes the presentation of thyroid autoantigens and activates T cells (4). The subsequent infiltration of the thyroid gland by APCs (dendritic cells and macrophages) may be triggered by inflammation resulting from either viral or bacterial infection or exposure to toxins. The common mechanism involved in the initiation and perturbation of the inflammatory processes in AITDs and in other autoimmune endocrine diseases (e.g., type 1 diabetes and Addison disease) is the Th1-cytokine/chemokine axis (17). Upon activation, Th1 lymphocytes produce IFN-γ -and TNF-α, which stimulate thyrocytes and retroorbital cells (in Graves ophthalmopathy) to secrete chemokines (CXCL10, CXCL9, and CXCL11). Chemokines, in turn, bind and activate the CXCR3 receptor on Th1 cells and further enhance IFN-γ and TNF-α secretion in a positive feedback loop which aggravates recruitment and activation of inflammatory cells in the affected organs (Figure 2). In advanced thyroiditis, the thyroid gland is infiltrated by B cells (representing up to 50% of the infiltrating immune cells), as well as cytotoxic T lymphocytes and CD4+ cells (7).

 

Figure 2. Depiction of the molecular mechanism involved in the inflammatory processes in autoimmune thyroid diseases. Activation of Th1 lymphocytes by antigen presenting cells produce INF-γ -and IL-2 and 12, which stimulate thyrocytes and retroorbital cells to secrete chemokines (CXCL10, CXCL9, and CXCL11). Chemokines bind and activate the CXCR3 receptor on Th1 cells, further enhancing IFN-γ and ILs secretion. APC, antigen presenting cells; IFΝ-γ, interferon gamma; IL-2, interleukin 2; IL-12, interleukin-12; CXCR3, C-X-C Motif Chemokine Receptor 3.

 

In Hashimoto thyroiditis, the humoral immune response is characterized by the presence of autoantibodies to TPO or Tg and is related to thyroid lymphocytic infiltration. These autoantibodies are themselves cytotoxic or may affect antigen processing or presentation to T cells. Th1 cells are the predominant T cell clones found in patients with Hashimoto thyroiditis: they promote apoptosis of thyrocytes through secretion of IL-12, TNF-α, and INF-γ, which activate cytotoxic T lymphocytes and macrophages (18). In addition, the Toll-like receptor-3 protein is overexpressed in human thyrocytes surrounded by immune cells in all patients with Hashimoto thyroiditis, but not in Graves’ disease or in euthyroid individuals (17).

 

In patients with Graves’ disease, the predominant antibodies are directed against the TSHR. T cells are activated through the presentation of TSHR peptides and, in turn, trigger B-cells and plasma cells that infiltrate the thyroid to produce autoantibodies directed against the TSHR. Activity of B-cells and plasma cells is also regulated by liver-produced insulin growth factor 1 (IGF1). In contrast to TPO and Tg, TSHR is widely expressed in extrathyroidal tissues and cells, including lymphocytes, adipose tissue, and fibroblasts. In consequence, the presence of TSHR antibodies contributes to the extrathyroidal manifestations of Graves’ disease, such as Graves ophthalmopathy, Graves dermopathy, and Graves-associated thymus hyperplasia (19).

 

Particularly in Graves ophthalmopathy, the TSHR autoantigen is presented by macrophages and B cells recruited to the orbit (20). Activated T cells, in turn, initiate an immunological attack on the orbital fibroblasts expressing TSHR. In response to the cytokines released by Th cells, orbital fibroblasts and adipocytes —both expressing TSHR— produce and deposit large amounts of glycosaminoglycans (e.g., hyaluronan), leading to increased osmotic pressure and water uptake, swelling of the extraorbital muscles, and increased accumulation of orbital adipose tissue. The process is enhanced by the IGF1/IGF1R signaling pathway in fibroblasts and adipocytes, as well as the crosstalk between TSHR and IGF1R intracellular signaling (21). A similar molecular pathophysiology may underlie Graves dermopathy (19).

 

NON-CODING RNAs

 

In recent years, non-coding RNAs, which are known to modulate gene transcription at the post-transcriptional level, have attracted a great deal of attention as potential biomarkers in various endocrine diseases. Although the study of microRNAs and circular RNAs is still in its infancy, these newly discovered non-coding, single-stranded RNA molecules have been implicated in the development and progression of AITDs.

 

A cluster of biomarkers consisting of miR-205/miR-20a-3p/miR-375/miR-296/miR-451/miR-500a/miR-326 has been reported to be differentially expressed in patients with Hashimoto thyroiditis (22,23). Similarly, multi-miRNA-based biomarkers, such as miR-762/miR-144-3p or miR-210/miR-155/miR-146, were differentially expressed in the serum of patients with Graves’ disease compared to healthy individuals. These dysregulated miRNAs can target key genes involved in the immune response and thyroid function. Regarding their prognostic role, higher miR-21-5p expression is associated with a worse prognosis for patients with Graves’ disease, whereas impaired expression of miR-155 correlates with the size of the goiter (22,24). Further understanding of the role of miRNAs in AITDs could provide valuable insights into disease mechanisms and potentially identify novel therapeutic targets. Data on other non-coding RNAs (such as long-noncoding RNAs and circular RNAs) are scarcer. Impaired expression of n335641, TCONS-00022357-xloc-010919, and n337845 was found in B cells of patients with Graves’ disease (25), while altered expression of 627 circRNAs in PBMCs of patients with Hashimoto thyroiditis has been tested for their potential prognostic value (26).

 

GUT MICROBIOME

 

Emerging evidence suggests that the composition and function of the gut microbiome may be involved in AITDs. The gut microbiome refers to the complex community of microorganisms residing in the gastrointestinal tract, which plays a vital role in various aspects of human health, such as prevention of intestinal colonization by pathogenic bacteria, fermentation/degradation of food debris, and production of nutrients. Studies have indicated that alterations in the gut microbiome can influence immune responses and contribute to the development of autoimmune diseases (7). In this context, dysbiosis, or an imbalance in the gut microbiota, has been observed in patients with Hashimoto thyroiditis and Graves’ disease, thus distinguishing them from healthy controls, while it was associated with the stage of disease, the level of thyroid autoantibodies, and the response to therapy.

 

Targeting the microbiome through dietary interventions or probiotics may represent a promising potential therapeutic avenue. It is therefore of interest to note that in hypothyroid patients treated with LT4, symbiotic supplementation for 8 weeks resulted in decrease of TSH concentration and LT4 dose (27). Moreover, the results of a large clinical study, INDIGO, reported a significant effect of LAB4 probiotics on the gut microbiota composition of patients with Graves’ disease and a temporary reduction in the serum level of IgG and IgA antibodies (28). Further research involving patients from different populations is required to fully understand the relationship between the gut microbiome and AITDs and to explore potential strategies for intervention.

 

Postpartum Thyroiditis

 

Postpartum thyroiditis may occur up to 12 months after delivery. Usually it presents as transient hyperthyroidism (median time of onset, 13 weeks post-delivery), followed by transient hypothyroidism (median time of onset, 19 weeks post-delivery). In the majority of patients, restoration of normal thyroid function occurs. Pregnancy triggers hormonal changes and immune system alterations, such as a shift from Th1 to Th2 cytokine production followed by a "rebound" shift back to Th1 after delivery, and fluctuations in transforming growth factor-beta1 TGF-β1) serum levels (29,30).

 

Anti-TPO and anti-Tg antibodies are found in almost all patients with postpartum thyroiditis. Notably, up to 50% of women who had anti-TPO antibodies at the end of the first trimester of gestation (i.e., before thyroid antibody titers start to decline during pregnancy) developed postpartum thyroiditis. Furthermore, there is evidence that the anti-TPO antibody titer at 16 weeks of gestation is related to the severity of postpartum thyroiditis (31), while activation of the complement is also involved in the pathogenesis (23).

 

Euthyroid Sick Syndrome

 

Euthyroid sick syndrome (ESS) is a condition characterized by alterations in thyroid hormone levels despite normal thyroid gland function. The characteristic laboratory abnormalities of the ESS include low T3 and/or fT3, elevated reverse T3 (rT3), normal or low TSH, and normal or low serum T4 or fT4 concentrations. Clinical conditions that trigger the development of ESS include systemic inflammation, myocardial infarction, starvation, sepsis, surgery, trauma, chronic degenerative diseases, malignancy, and every other condition associated with severe illness.

 

During illness or periods of severe physiological stress, the immune system releases various proinflammatory cytokines, such as in IL-6, TNF-α, IL-1β, IFN-γ, and TGF-β2. These cytokines disrupt the hypothalamic-pituitary-thyroid axis and interfere with the normal synthesis, secretion, and metabolism of thyroid hormones (Figure 3), while the more severe the illness, the more extensive the hormonal alterations.

 

Figure 3. Secretion of proinflammatory cytokines during severe illness or stress inhibits the activity of hepatic deiodinase type 1- suppressing the peripheral conversion of T4 to T3. They also suppress intrathyroidal hormone synthesis, TRH release and TSH secretion from the pituitary. IL-6, interleukin-6; IL-1, interleukin-1; TNF-α, tumor necrosis factor alpha; INF-γ, interferon gamma; TGF-β, tumor growth factor beta; TG, thyroglobulin; NIS, sodium/iodide symporter; TSH, thyroid stimulating hormone; TRH, Thyrotropin-releasing hormone.

 

Proinflammatory cytokines suppress the peripheral conversion of T4 to T3, resulting in low T3 levels and increase rT3 levels, by inhibiting the activity of hepatic deiodinase type 1, which promotes conversion of T4 to T3 and of rT3 to diiodothyronine (32). They also suppress TRH release and inhibit TSH response to TRH stimulation, leading to a decrease in TSH levels. Thus, the cause of the decreased T3 concentration in ESS is decreased T3 production, whereas the cause of the increased rT3 concentration is the result of impaired degradation. Prolonged and severe illness is marked by a decrease in circulating total T4 along with low T3 and high rT3, with very low T4 levels displaying a poor prognosis and having been associated with an increased mortality rate (32). In addition, cytokines reduce iodine uptake by inhibiting sodium-iodine symporter protein expression (25-27), and decrease thyrocyte growth (33), iodide organification (34,35), and thyroglobulin synthesis (36,37).

 

The central role of cytokines in the pathophysiology of ESS has been further elucidated in studies involving cytokine administration to humans. TNF-α administration in healthy volunteers caused a decrease in serum T3 and an increase in serum rT3 concentration (38). Unlike IL-6, serum TNF-α levels did not correlate with any of the typical thyroid parameters, such as low T3, increased rT3, or decreased TSH levels seen in ESS (39,40), suggesting that the changes of thyroid hormone profile following TNF-α administration might be indirect (i.e., through TNF-α increase in circulating IL-6 levels). Furthermore, both IL-6 and TNF-α can upregulate type 2 deiodinase in the anterior pituitary, affecting TSH release and contributing to the development of the non-thyroidal illness syndrome (41,42).

 

Leptin, a hormone primarily known for its role in regulating appetite and energy balance, has also been implicated in the development of EES. During acute illness or chronic inflammation, leptin levels are usually elevated. A link between leptin and the proinflammatory cytokines TNF-α and IL-6 in chronic inflammatory diseases, such as chronic obstructive pulmonary disease (43) and ankylosing spondylitis (44), respectively, has also been proposed previously.

 

The primary effect exerted by leptin on the hypothalamic-pituitary-thyroid axis is alteration of the setpoint for feedback sensitivity of hypophysiotropic TRH-producing neurons in the paraventricular nucleus of the hypothalamus to thyroid hormones (mainly T3) by lowering of the setpoint when leptin levels are suppressed during fasting (45). Two anatomically distinct and functionally antagonistic populations of neurons in the arcuate nucleus of the hypothalamus, α-melanocortin-stimulating hormone (α-MSH)-producing neurons that co-express cocaine- and amphetamine-regulated transcript and neuropeptide Y (NPY)-producing neurons that co-express agouti-related peptide (AGRP), are responsible for the effects of leptin on hypophysiotropic TRH. It has also been proposed that leptin directly affects hypophysiotropic TRH neurons (46). Leptin has been found to inhibit the conversion of T4 to T3 in peripheral tissues and increase the activity of the enzyme type 3 deiodinase, which converts T4 to rT3. These data suggest that leptin can disturb thyroid function in seriously ill patients via two different independent mechanisms (cytokine-dependent and directly).

 

Amiodarone-Induced Thyroid Disease

 

Amiodarone, a benzofuran derivative with a similar structure to that of thyroid hormones, is a highly effective antiarrhythmic agent widely used in the treatment of various types of tachyarrhythmias (supraventricular and ventricular arrhythmias). Amiodarone contains two iodine atoms per molecule, which is approximately 37.5% iodine by molecular weight (47).

 

Treatment with amiodarone may be related to an increase in lymphocyte subsets leading to an exacerbation of pre-existing autoimmunity (48,49). The relative proportion of patients developing either thyrotoxicosis or hypothyroidism depends on the iodine content of the local diet and pre-existing thyroid autoimmunity. In relatively iodine-replete areas, approximately 25% of patients with amiodarone-induced thyroid dysfunction become thyrotoxic, accounting for approximately 3% of amiodarone-treated individuals (50).

 

Amiodarone-induced hypothyroidism is attributed to an increased susceptibility to the inhibitory effect of iodide on thyroid hormone synthesis and/or to a failure to escape the Wolff-Chaikoff effect (49). Hashimoto thyroiditis is the most common risk factor for amiodarone-induced hypothyroidism and it is considered the most likely reason for the female preponderance of this clinical entity (51). Female patients with positive anti-TPO and anti-Tg autoantibodies have a relative risk of 13.5% for developing amiodarone-induced hypothyroidism compared to men without thyroid autoantibodies (20).

 

The pathogenesis of amiodarone-induced thyrotoxicosis is complex, although two distinct forms, type 1 and type 2, are recognized. Type 1 develops in patients with latent thyroid disease, predominantly nodular goiter, in whom the amiodarone iodine load triggers increased synthesis of thyroid hormones. Type 2 is the result of a destructive thyroiditis in a previously normal gland, with leakage of preformed thyroid hormones despite a reduction in hormone synthesis (47,50,52,53).

 

Differentiating between the two types of amiodarone-induced thyrotoxicosis is an essential step in their management, as treatment of each type is different (50). Type 1 usually responds to thionamide therapy, which blocks hormone synthesis, and perchlorate, which blocks active transport of iodine into the thyroid, whereas type 2 responds to high-dose glucocorticoids (50,53-55). Nevertheless, several studies now suggest that these two types should be treated concomitantly; thus, currently, patients with amiodarone-induced thyrotoxicosis receive both antithyroid drugs and prednisolone. In cases resistant to medical treatment and/or in patients with severe cardiac diseases who cannot interrupt amiodarone or require quick amiodarone reintroduction, total thyroidectomy may be offered after rapid correction of thyrotoxicosis following combination treatment with thionamides, KClO4, glucocorticoids, and a short course of iopanoic acid (56).

 

Thyroid Cancer

 

Thyroid cancer is the most common endocrine cancer, the incidence of which has steadily increased over the past few decades (57).

 

The association of chronic inflammation induced by Hashimoto thyroiditis and thyroid cancer has been long recognized (58). However, the immune response triggered against thyroid cancer and AITDs differs significantly. In thyroid cancer, the immune response is more tolerant and allows tumor growth, whereas in AITDs, the response is more aggressive, triggering cell destruction and reduction of the function of the gland (59). Hashimoto thyroiditis is considered both a risk factor for the development of thyroid cancer (60) and a favorable prognostic factor due to chronic lymphocytic infiltration, which can downregulate tumor aggressiveness (60,61). In Graves’ disease, the presence of a strong humoral immune response appears to be protective against thyroid cancer. Patients with increased anti-TPO and anti-Tg levels show lower distant metastasis rates than patients without thyroid autoantibodies (62,63), suggesting their potentially protective role (59).

 

Immune infiltrates in the tumor microenvironment differ between the different thyroid neoplasm subtypes (Figure 4). In general, differentiated thyroid cancer (DTC) has a higher number of tumor-associated lymphocytes and regulatory T cells (Tregs), while anaplastic thyroid cancer (ATC) and medullary thyroid cancer (MTC) display a high density of tumor-associated macrophages (64). The number of tumor-associated macrophages has been associated with dedifferentiation, lymph node metastases, and reduced survival (65). It is important to note, however, that most of the studies analyzing the immune milieu of DTC have used papillary thyroid cancer (PTC) tumor samples (66-69). Myeloid-derived suppressor cells are associated with aggressive characteristics of differentiated thyroid cancer and are related to poor prognosis (65).

 

Figure 4. Immune infiltrates differ in different types of thyroid cancer [Modified by Garcia-Alvarez et al, (70)]. TAM, tumor-associated macrophages; MDSC, myeloid-derived suppressor cell.

 

The dendritic cells, which play a critical role in antigen presentation, are increased in PTC, while neutrophils are found in more aggressive thyroid cancers (such as poorly differentiated or anaplastic). In addition, natural killer cells that play an important role in immunosurveillance are also increased in PTC and are negatively correlated with tumor stage, while lymphocytic infiltration is associated with better overall survival and low recurrence rate (71,72).

 

Cytokines, which may be produced by thyroid follicular cells and by immune cells infiltrating thyroid tumors, are also related to tumor development. IL-1 and IL-6 stimulate thyroid cell proliferation and tumor growth, while TGF-β, which is a suppressive cytokine, is overexpressed in aggressive cancers (73). In addition, multiple chemokines may be secreted by thyroid cancer or immune cells and affect chemiotaxis, angiogenesis, and lymphangiogenesis (73).

 

Immunomodulatory proteins, such as programmed death-ligand 1 (PDL1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), T cell immunoglobulin and mucin-domain containing-3 (TIM-3), lymphocyte activation gene-3 (LAG-3), and T cell immunoglobulin and ITIM domain (TIGIT), which are considered major immune coinhibitory receptors and promising immunotherapeutic targets in cancer treatment, are also expressed in thyroid cancer, being associated with more aggressive tumor characteristics and a poor prognosis (64). Programmed death-ligand 1 staining by immunohistochemistry has shown higher expression in ATC than in other subtypes (70). Six cohort studies have been published to-date reporting positive PD-L1 expression, varying between 22% and 65%, this being higher compared to that detected in DTC and poorly differentiated thyroid cancer (74-79). TIM-3 expression was observed in 48% of patients with MTC, and in the majority of cases (84.4%) its expression was restricted to tumor cells (80). Other coinhibitory receptors, such as LAG-3 and T cell TIGIT, were observed in a lesser percentage of cases (approximately 3%) (80). Nevertheless, results from clinical trials with immunotherapy as monotherapy or combinations have shown limited efficacy (70). In one phase Ib KEYNOTE-028 trial assessing the efficacy of pembrolizumab in patients with PD-L1+ (membranous staining on ≥1%) locally advanced or metastatic follicular or papillary thyroid cancer, pembrolizumab achieved an objective response rate of 9% and a median progression-free survival of 7 months (81). Several clinical trials further investigating the efficacy of combination therapy of immune checkpoint inhibitors are currently ongoing.

 

IMMUNE SYSTEM AND DIABETES MELLITUS

 

The immune system plays a crucial role in the pathogenesis of both type 1 and type 2 diabetes.

Diabetes type 1, which is immune-mediated in more than 95% of cases, is an organ-specific autoimmune disease characterized by lymphocytic infiltration and inflammation that leads to pancreatic β-cell destruction and absolute insulin deficiency (82). The immune system’s attack on pancreatic β-cells is usually triggered by a number of factors, including genetic predisposition and environmental triggers such as viral infections (e.g., Coxsackie B4, mumps, and rubella) or dietary compounds (e.g., cow’s milk) (82). The process involves the activation of immune cells, particularly T cells, which recognize self-autoantigens in pancreatic β-cells and initiate an immune response.

 

On the other hand, in type 2 diabetes, the immune system also plays a different role from that in DM1. Chronic low-grade inflammation, often associated with obesity, leads to immune cell activation and the release of proinflammatory cytokines. These cytokines interfere with the normal functioning of insulin and promote insulin resistance. Macrophages infiltrate adipose tissue and release inflammatory molecules, further exacerbating insulin resistance with increasing adiposity. The immune system, thus, contributes to the development and progression of insulin resistance and eventually promotes the onset of DM2.

 

Understanding the intricate relations between the immune system and diabetes pathogenesis is essential for the development of effective treatments and interventions for the management of both types of diabetes.

 

Diabetes Type 1

 

The susceptibility to develop DM1 is associated with multiple alleles of the major histocompatibility complex MHC I and II locus. More than 90% of patients with DM1 express either HLA DR3, DQ2 or DR4, or DQ8 (83), whereas HLA haplotype DR2, DQ6 is protective against DM1 development.

 

The primary pathological presentation of DM1 is inflammation of the pancreatic islets, also known as insulitis, caused by infiltration of immune cells, including CD4 and CD8 T cells along with B cells (84-86). Although the initial events triggering autoreactive responses remain unclear, presentation of pancreatic islet autoantigens by the associated MHC class II molecules contribute to priming and expansion of pathogenic T cells.

 

CD4 T helper cells are required for the development of the autoimmune process in the pancreatic islets, while CD8 cytotoxic T cells are the cells responsible for β-cell destruction (Figure 5). T cell receptors recognize peptides bound to MHC molecules on the surface of antigen-presenting cells (B-cells, dendritic cells, and macrophages). Each T cell then generates a unique receptor for the recognition of an autoantigen presented in the MHC molecule. The interaction between T cell receptor/autoantigen/MHC leads to activation of the T cells.

 

Figure 5. T cell mediated destruction of β- cells in diabetes type 1. CD4+ effector T cells recognize an insulin or islet peptide presented by diabetes-risk conferring HLA-DQ8 or DR4 on APCs, migrate to the pancreas and promote pancreatic β-cell destruction. Inside the pancreatic islets CD8+ effector T cells recognize islet antigens presented by HLA-A2 leading to the destruction of the β-cells. [Modified by Mitchel et al (87)]. TCR, T cell receptor; APC, antigen presenting cell; HLA-A2, human leukocyte antigen-A2.

 

Three different mechanisms have been proposed to explain T cell activation in DM1. One mechanism is thought to involve molecular mimicry-activated T cell proliferation. The hypothesis for this mechanism is based on the assumption that epitopes of proteins expressed by infectious agents can be shared by unrelated molecules encoded by host genes (88). A second mechanism that may trigger molecular mimicry-activated T cell proliferation is “bystander” T cell proliferation. This mechanism involves the stimulation of non-antigen-specific T cells by various cytokines during infection, simply because they are in the area. The cytokines thought to be involved in this nonspecific stimulation are IFN-α and IFN-β (89). The 3rd mechanism might involve a superantigen-mediated T cell proliferation mechanism: this theory proposes that autoreactive T cells can be inappropriately primed to react against self-structures through an encounter with a superantigen (90).

 

Multiple autoantigens in the pancreatic islets have been identified, including non-specific islet cell autoantigens (ICA), insulin, glutamic acid decarboxylase 65 (GAD65), insulinoma antigen-2 (IA-2), heat shock protein (HSP), islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), imogen-38, zinc transporter-8 (ZnT8), and the most recently identified pancreatic duodenal homeobox factor 1 (PDX1), chromogranin A (CHGA), and islet amyloid polypeptide (IAPP) (91). Autoantibodies against these autoantigens may be detected long before the onset of hyperglycemia and usually decline during the course of the disease (92).

 

Most patients diagnosed with DM1 have circulating islet cell autoantibodies directed against pancreatic islet autoantigens. However, although the detection of autoantibodies may be useful for DM1 diagnosis and prediction, it is the cellular immune system that eventually infiltrates the pancreatic islets and causes β-cell destruction. In turn, further β-cell destruction leads to more self-antigen presentation and ensuing amplification of the immune response (82,93,94).

 

Among several proinflammatory cytokines, IL-21, produced by CD8 T cells, is required for the development of DM1, while TNF-α may also be involved (95,96). IL-6 plays an important role in the pathogenesis of vitiligo-associated DM1 (97). In contrast, in vivo experiments with non-obese diabetic mice have shown that IL-4, produced by Th2 cells, may be protective against developing diabetes (95,96,98,99).

 

Understanding both the pathophysiology and the regulatory mechanisms involved in DM1 is a critical step towards the development of antigen-specific, β-cell-directed, immunomodulatory or cellular treatment modalities (100).

 

Investigations into the efficacy and safety of various immunotherapeutic strategies against the development of DM1 have been carried out in recent clinical trials and are still ongoing in current trials (101). Among them, T cell-directed therapies that aim at a favorable balance between effector T cell depletion and regulatory T cell preservation have shown the most promising results. Teplizumab, an anti-CD3-directed monoclonal antibody, was the first immunomodulatory agent to demonstrate a significant delay in disease progression in high-risk individuals before clinical onset (102) and has recently (November 17, 2022) been approved by the FDA as the first disease-modifying therapy for DM1 in adults and in children aged 8 years and older.

 

The Enigma of Pancreatic α-Cell Resistance in Diabetes Mellitus Type 1

 

Although both insulin-producing β-cells and glucagon-producing α-cells of the pancreatic islets share a similar embryonic origin and are directly exposed to the deleterious immune signals in DM1, it appears that the immune system selectively destroys β-cells, while α-cells survive even in long-term DM1 (103). α-Cells are located in close proximity to β-cells in human pancreatic islets (104), creating a closed communication loop that regulates their function and secretory capacity (105).  Dysfunction of α-cells plays a significant role in the pathogenesis of both types of diabetes (106). It has long been proposed that the reduced functional β-cell mass in DM1 and the consequent hypoglycemia were the key mechanisms indirectly inducing dysfunction of α-cells in diabetes (107). Advanced molecular technology of the last decade has, however, challenged this notion, showing that dysfunction of α-cells in diabetes is not secondary to β-cell pathology but is instead directly immune-induced (108-110). Despite this dysfunction, α-cells exhibit a remarkable autoimmune resistance that enables them to survive over β-cells in long-standing diabetes (103,111). Recent studies using single-cell RNA sequencing (109,112) have demonstrated important differences between these two cell types in terms of expression of: i) anti-apoptotic genes, ii) endoplasmic reticulum (ER) stress-related genes, III) innate immune response genes, and iv) antiviral response genes, all of which render α-cells less immunogenic and more resistant to viral infections and ER stress (113). In addition, CD8+ T cells invading the islets in DM1 are reactive to preproinsulin but not to glucagon. Furthermore, although β-cells are essential to life (neither humans nor animal models can survive without them), mice with 98% α-cell ablation retain near-normal glucose homeostasis (114): this points to the knowledge gap we have, from an evolutionary point of view, regarding the question of why β-cells are more fragile than α-cells. Certainly, greater understanding of the underlying mechanisms responsible for the autoimmune resistance of α-cells is critical since it is likely to reveal intracellular pathways amenable to therapeutic interventions that would increase the resistance of the β-cells themselves to the immune attack of the host’s immune system.

 

Diabetes Type 2

 

DM2 accounts for 90% of cases of diabetes worldwide (115). The increasing prevalence of DM2 around the world has been largely attributed to an unhealthy lifestyle and resultant development of obesity and overweight (116). Obesity is strongly related to DM2 mainly through inducing insulin resistance in the insulin sensitive tissues in the periphery.

 

The concept that a smoldering inflammatory process plays an important part in the pathogenesis of DM2 (117) has attracted much attention and is supported by evidence of inflammation in islets, adipose tissue, liver, and muscle that can provoke insulin resistance and β-cell dysfunction (118-120). Adipose tissue is characterized by infiltration by macrophages and other immune cells that produce cytokines and chemokines and contribute to the development of local and systemic chronic low-grade inflammation, this inflammatory milieu being the link between obesity, insulin resistance, and diabetes mellitus (121,122).

 

Earlier in vivo studies demonstrated that levels of TNF-α (123-125), IL-6, C-reactive protein, plasminogen activator inhibitor, and other inflammation mediators were elevated in adipose tissue and plasma of obese mice (126,127). It was also observed that these inflammatory mediators, together with saturated free fatty acids and reactive oxygen species (ROS), inhibited serine phosphorylation of the insulin receptor substrates (IRS-1 and 2) (128-130) in insulin sensitive tissues, such as adipose tissue and the liver (131), promoting insulin resistance (Figure 6). TNF-α is associated with increased release of free fatty acids by adipose tissue and leads to impaired insulin secretion and signaling (123,132).

 

Figure 6. Insulin resistance and inflammation in diabetes type 2. Insulin binds to insulin receptor (IR) in insulin sensitive tissues, and autophosphorylates tyrosine molecules of IRS-1 and -2 substrates. In the presence of obesity, and hyperlipidemia, the influx of free fatty acids, inflammatory cytokines and glucose activates IKKβ and JNK, which are the mediators for stress and inflammatory. In turn IKKβ and JNK inhibit tyrosine phosphorylation of IRS1 and 2 and promote transcriptional activation of genes related to inflammatory and stress responses resulting in insulin resistance. [Modified by Berbudi et al (133)]. IRS 1 -2, insulin receptor substrates; ER, Endoplasmic reticulum; IKKβ, inhibitory kappa B kinase β; JNK1 and c-Jun N-terminal kinase I.

 

In line with these early in vivo studies, studies in humans have shown that elevated levels of i) nonspecific indicators of inflammation, such as white cell count, fibrinogen, and CRP levels (134-136), ii) markers of reduced fibrinolysis, such as plasminogen activator inhibitor-1 (PAI-1), and tissue plasminogen activator (tPA), iii) von Willebrand factor (vWf), which is a marker of endothelial injury, and iv) early markers of inflammation, such as monocyte chemotactic protein-1 (MCP-1), IL-8, and interferon-γ-inducible protein-10 (137), were predictive of DM2 development.

 

Furthermore, CRP and/or IL-6 were associated with the incidence of DM2 independently of adiposity or insulin resistance (136,138,139). Visceral adipose tissue appears to be a major source of circulating IL-6 in humans, and obese people with insulin resistance display high levels of plasma IL-6 concentration, also predictive of DM2 development (113). TNF-α is also increased in obese individuals with insulin resistance (124,140) and it plays a major role in the pathogenesis of obesity-linked DM2 (141).

 

At the cellular level, chronic exposure of adipocytes to low doses of TNF-α led to a dramatic decrease in insulin-stimulated auto-phosphorylation of the IRS 1-2 (142). Treatment of cultured murine adipocytes with TNF-α induced serine phosphorylation of IRS-1 and convert IRS-1 into an inhibitor of IR tyrosine kinase activity in vitro. TNF-α has also been shown to downregulate glucose transporter GLUT4 mRNA levels in adipocyte and myocyte cultures as well (125,143,144).

 

Oxidative stress, as a result of increased cytokine levels in DM2, is also thought to play an important role in activating inflammatory genes (145,146) (Figure 6). It is possible that oxidative stress markers do not adequately reflect the impact of increased ROS on β-cells or insulin signaling, while inflammatory, procoagulant or endothelial dysfunction markers are more specific to the pathophysiology of hyperglycemia (145,146). Hasnain et al. showed that islet-endogenous and exogenous IL-22 suppressed oxidative and ER stress caused by cytokines or glucolipotoxicity in mouse and human β-cells by regulating oxidative stress pathways. In obese mice, antibody neutralization of IL-23 or IL-24 partially reduced β-cell ER stress and improved glucose tolerance, whereas IL-22 administration modulated oxidative stress regulatory genes in islets, suppressed ER stress and inflammation, promoted secretion of high-quality efficacious insulin, and fully restored glucose homeostasis, followed by reinstitution of insulin sensitivity (147).

 

The chemokine system is also associated with obesity and insulin resistance. MCP-1, which acts on monocytes, macrophages, T cells and NK cells, is increased in obese compared to lean subjects and is related to non-alcoholic fatty liver disease and other lipid overload states (148-153). A short-term program of 4-month lifestyle modification significantly decreases MCP-1 levels, with favorable effects on the glycemic and lipid profile (151).

 

Collectively, these findings supported the investigation of new therapeutic approaches that target inflammation to ameliorate diabetes and its complications. Regarding the latter, it is important to note that multiple sources of evidence support a pathogenic connection between rheumatoid arthritis (RA) and the mechanisms of DM2, via formation of a vicious circle that is perpetuated by impaired glucose metabolism and inflammation. In this context, ongoing clinical studies have shown that the inhibition of interleukin IL-1 and IL-6 may allow the treatment of RA and concomitant T2D at the same time (154).

 

Treatment with anakinra, a recombinant form of human IL-1 receptor antagonist that works as a competitive inhibitor of IL-1β, achieved significant improvements of glycemia and secretory function of β-cells (155,156), which was maintained after anakinra withdrawal during a 39-week follow-up (157). Canakinumab, a monoclonal antibody against IL-1β, significantly reduced inflammatory proteins, such as CRP, IL-6, and fibrinogen, in patients with DM2 and high cardiovascular risk (158,159), while regarding glycemic control, it reduced values of HbA1c during the first 6–9 months of treatment, without, however, consistent long-term benefits (159). Antagonists of IL-6 receptor tocilizumab and sarilumab have also been investigated in patients with RA with or without concomitant DM2. Tocilizumab showed a positive effect on insulin resistance in some studies (160-162), while other studies failed to report any beneficial effect on glycemic control (163,164). The efficacy of sarilumab was assessed in a post hoc analysis (165) of three randomized clinical trials in patients with RA with or without DM2 (166-168). Sarilumab, as monotherapy or in combination, significantly reduced HbA1c compared to adalimumab monotherapy or placebo plus methotrexate/convectional DMARDs in patients with RA and DM2 (165).

 

Several studies have analyzed the effects of TNF inhibitors (i.e., adalimumab, etanercept, and infliximab) on glucose metabolism, demonstrating a potential favorable effect on insulin resistance and insulin sensitivity (169,170). In a meta-analysis combining data from 22 randomized controlled trials and three cohort studies (171), new-onset DM2 was delayed in RA patients treated with TNF inhibitors.

 

The Effect of Diabetes on the Immune System

 

Like a mirror image, chronic hyperglycemia in diabetic patients impairs the host’s immune response, which in turn fails to control the spread of invading pathogens, rendering diabetic patients more susceptible to infections (133,172). Both innate immune response defects and defective adaptive immune response are implicated in this incapacity of the immune system to defend against invading pathogens in patients with diabetes. Several mechanisms have been proposed by experimental and human studies including: i) leukocyte recruitment inhibition (173,174), ii) defective pathogen recognition (175), iii) dysfunction of neutrophils (176-178), macrophages resulting in impairment of phagocytosis (179), iv) functional defects in natural killer cells (180), and v) dysfunction of complement activation (181).

 

OSTEOPOROSIS AND THE IMMUNE SYSTEM

 

Osteoporosis is a clinical condition characterized by low bone mass and impaired bone microarchitecture associated with an increased risk of fragility fractures. Growing evidence of the last few decades has demonstrated the effect of the immune system on bone metabolism, leading to the emergence of the new field of osteoimmunology (182-185) and immunology of osteoporosis (named as immunoporosis) (184-188).

 

States of immune dysfunction such as immunodeficiency, inflammatory diseases, or immune response to infections are associated with increased osteoclastic bone resorption and, therefore, increased bone loss and increased fracture risk.

 

Bone Remodeling and Bone Cells

 

Bone remodeling is a dynamic and continuous process that is responsible for the maintenance of skeletal health throughout life. It involves three consecutive phases: i) osteoclast-mediated bone resorption; ii) the reversal phase, during which mesenchymal derived osteoblasts are recruited to the bone site of bone resorption; and iii) osteoblast-mediated bone formation. The two processes of bone resorption and bone formation are tightly coupled and under control of the matrix-embedded osteocytes, capable of sensing and integrating mechanical and chemical signals from their environment to regulate bone formation and resorption at the bone surfaces.

 

Osteoclasts originate from the same myeloid precursor that derives macrophage and dendritic cells and are specialized in bone degradation (186). Osteoblasts are the main bone-forming cells and are derived from mesenchymal stem cells. Osteoclast formation and differentiation is regulated by macrophage colony-stimulating factor (M-CSF) and the receptor activator of nuclear factor-kB (RANK) ligand (RANKL) produced by osteoblasts and osteocytes (189,190). Osteocytes are a significant source of RANKL and its decoy receptor, osteoprotegerin (OPG), which binds to RANKL, preventing its interaction with RANK, while they also secrete the Wnt signaling inhibitor sclerostin, which regulates bone formation (189). RANKL is additionally expressed by fibroblasts and immune cells, including antigen-stimulated T cells and dendritic cells (191-193), while OPG is also produced by B lymphocytes and dendritic cells (194).

 

Inflammatory Diseases

 

Activated T cells increase the production of TNF-α and RANKL and stimulate osteoclastogenesis during inflammation (182-185,192-195). Multiple cytokines may promote osteoclastogenesis mainly by regulating the RANK/RANKL/OPG axis. TNF-α, IL-1, IL-6, IL-7, IL-11, Il-17, and IL-23 promote osteoclast differentiation, while IFN-α, IFN-γ, IL-3, IL-4, IL-10, IL-27, and IL-33 are considered anti-osteoclastogenic cytokines that protect bone integrity (195). Th17 cells are considered an osteoclastogenic subset of T cells as they enhance osteoclastogenesis by secreting IL-1, IL-6, Il-17, RANKL, TNF-α, and IFN-γ. Activation of Th2 leads to enhanced production of PTH and promotes the anabolic activity of osteoblasts in several inflammatory states. Furthermore, Th2 lymphocytes are associated with a low RANKL/OPG ratio and inhibition of bone loss (196). In addition, β-cells produce RANKL and OPG and may influence bone formation and absorption, while it has been observed that in HIV infected patients, there is an altered β-cell RANKL/OPG ratio that is inversely correlated with BMD (197).

 

Interleukin 6, produced by both stromal and osteoblastic cells (198) in response to stimulation by systemic hormones such as PTH, PTH-related peptide (PTH-rP), thyroid hormones, and 1,25-dihydroxyvitamin D3 and other cytokines (i.e., TGF-β, IL-1, and TNF-α), plays a major role in osteoclast development and function. Increased IL-6 levels contribute significantly to the abnormal bone resorption observed in patients with multiple myeloma (199), Paget’s disease of bone (200), rheumatoid arthritis (201), and Langerhans cell histiocytosis (202). Effects of increased osteoclast-induced bone resorption are not solely attributable to IL-6, but to all IL-6 family cytokines (203).

 

TNF-α has also been shown to induce bone resorption and plays an important part in inflammatory bone diseases (192). TNF-α promotes RANKL expression in osteoclast precursors and the formation of multinucleated osteoclasts in the presence of M-CSF. Furthermore, TNF-α increases RANKL and M-CSF expression in osteoblasts, stromal cells, and T lymphocytes, while RANKL can also enhance TNF-α mediated osteoclastogenesis (195). IL-1β increases RANKL expression and stimulates osteoclast formation and bone resorption while also promoting TNF-α induced osteoclastogenesis (204,205).

 

Postmenopausal Osteoporosis

 

Estrogen deficiency is a state of increased bone remodeling associated with an increased rate of bone resorption relative to bone formation, resulting in net bone loss. It has been shown that estrogen deficiency-induced bone loss has a complex mechanism mainly involving the immune system rather than a direct effect of estrogen on bone cells (206). Estrogen loss during menopause is associated with an expansion of T and B lymphocytes (207,208) leading to increased production of RANKL (209). In addition, an increased level of proinflammatory cytokines, including TNF-a and IL-1b, is observed in postmenopausal women (210,211). In addition, B lymphocytes may partially contribute to trabecular bone loss in postmenopausal osteoporosis (212). In the absence of estrogens, dendritic cells live longer, increasing their expression of IL-7 and IL-15. In turn, IL-7 and IL-15 induce IL-17 and TNF-a production in a subset of memory T cells, independent of antigen activation (213). These proinflammatory cytokines contribute to inflammation-induced bone loss in postmenopausal osteoporosis by activating low-grade inflammation. In contrast, Treg cells have a bone-protective role in postmenopausal osteoporosis (210). However, it has been shown that Th17/Treg balance is disturbed in estrogen deficiency. Treg cells lose their immunosuppressive function and convert to Th17 cells, which explains the imbalance of Th17/Treg in postmenopausal osteoporosis (214).

 

Senile Osteoporosis

 

Senile osteoporosis, on the other hand, is a low-bone turnover disease with decreased bone resorption and significantly reduced bone formation (215), which commonly occurs in older people, above 65, and affects both males and females. In recent years, it was demonstrated that aging is usually accompanied by systemic low-grade chronic inflammation and enhanced inflammatory mediators, such as IL-6 and TNF-a (216). A recent study found that senescent immune cells, such as macrophages and neutrophils, accumulate in bone marrow during aging in rats and mice (217). The senescent macrophages and neutrophils repress osteogenesis by promoting bone marrow mesenchymal stromal cell adipogenesis. In addition to directly inhibiting osteogenesis, the senescent immune cells contribute to chronic inflammation, thus leading to inflammatory bone resorption (217). Aging can tilt the balance of Th1/Th2 toward Th2 cells, resulting in an increased inflammatory response (218) and low-level chronic inflammation, ultimately leading to continuous bone loss.

 

Thyrotoxicosis-Induced Osteoporosis

 

IL-6 and IL-8 play a major role in thyrotoxicosis-induced osteoporosis and are increased in patients with thyrotoxicosis due to Graves’ disease or toxic multinodular goiter (219). In addition, patients with thyroid carcinoma on TSH suppressive therapy have significantly increased circulating levels of IL-6 and IL-8 compared to controls (219), which are tightly associated with serum T3 and fT4 concentrations. Both IL-6 and IL-8 have also been shown to be released by human bone marrow stromal cell cultures containing osteoblast progenitor cells in response to T3 (196). TNF-α elevations due to low TSH signaling in human hyperthyroidism also contribute to the bone loss that has traditionally been attributed solely to high thyroid hormone levels (220). Hyperthyroid mice lacking TSHR had greater bone resorption than hyperthyroid wild-type mice, demonstrating that the absence of TSH signaling contributes to low bone mass (221) in the hyperthyroid state.

 

Primary Hyperparathyroidism-Induced Osteoporosis

 

Bone resorption in primary hyperparathyroidism (PHP) also appears to be related to immune system effects. Circulating levels of IL-6 and TNF-α, which are significantly increased in patients with PHP, are strongly correlated with biochemical markers of resorption, returning to normal after successful parathyroidectomy (222). The hypothesis that IL-6 mediates the catabolic effects of parathyroid hormone (PTH) on the skeleton has been further strengthened by the finding that neutralizing IL-6 in vivo attenuates PTH-induced bone resorption in mice, while the resorptive response to PTH was also reduced in IL-6 knockout mice (223). Furthermore, it has been observed that transplantation of parathyroid from humans with hyperparathyroidism to mice lacking T cells was not associated with bone loss, suggesting a possible role of T lymphocytes in PTH-related osteoporosis (224). A direct action of PTH on T lymphocytes may also contribute, as deletion of the PTH receptor from T cells failed to induce bone loss (225). It has been proposed that PTH action on T cells results in secretion of TNF-α and, in combination with RANKL increase and OPG suppression, guides their differentiation to Th17 subsets, with subsequent IL-17 secretion and further RANKL amplification (226).

 

Drug-Induced Osteoporosis

 

The immune cells are also involved in drug-induced osteoporosis, such as glucocorticoid and chemotherapy-induced osteoporosis. A recent study demonstrated that glucocorticoid-induced osteoporosis could not be induced in T cell-deficient mice, while re-establishment was found to be possible by transferring splenic T cells from wild-type mice (227).

 

Cyclophosphamide, is a chemotherapy drug that causes immunosuppression and is associated with increased risk of osteoporosis (228-230). By improving the functional status of immune cells in an immunosuppressive mouse model induced by cyclophosphamide, bone loss was dramatically reduced (231), pointing to the possible contribution of immune cells in cyclophosphamide-induced osteoporosis.

 

EFFECTS OF THE IMMUNE SYSTEM ON THE STRESS SYSTEM

 

The Hypothalamic-Pituitary-Adrenal (HPA) Axis

 

The relations between the immune and the stress systems are complex and bidirectional, denoting that while stress can affect immune function, immune responses can also influence stress levels through various ways and mechanisms.

 

INFLAMMATION

 

During acute inflammation, the immune system is activated in response to infection or injury and releases proinflammatory cytokines and other inflammation-related factors into the central nervous system (CNS), plasma, and endocrine glands. Inflammatory cytokines, such as TNF-α, IL-1, and IL-6, produced by a variety of cells, including monocytes, macrophages, astrocytes, endothelial cells, and fibroblasts, activate the HPA axis leading to an increase of corticotropin-releasing hormone (CRH), adrenocorticotrophic hormone (ACTH) and, finally, glucocorticoids (38, 184). In turn, increased circulating levels of glucocorticoids exert suppressive effects on the inflammatory reaction, controlling the immune response and helping the organism to reach its prior healthy homeostasis (232). Similarly, in acute stress, the amplitude and synchronization of CRH secretory pulses is increased, and this is reflected in the levels of ACTH and cortisol in the systemic circulation (233).

 

The proinflammatory cytokine IL-1, especially its β form, is probably the most important molecule capable of modulating cerebral functions during systemic and localized inflammation. Systemic IL-1β injection activates the neurons involved in the control of autonomic functions, and neutralizing antibodies or IL-1 receptor antagonists are capable of preventing numerous responses during inflammatory stimuli (234). Similarly to IL-1β, intravenous IL-6 stimulates the hypothalamic-pituitary unit, leading to the secretion of cortisol by the adrenal glands and subsequent termination of the inflammatory cascade (235). All three inflammatory cytokines (IL-1, IL-6, and TNF-α) have the capacity to activate the HPA-axis, but it appears that IL-6 is the most critical component of this cascade. Studies in rats have demonstrated that immunoneutralization of IL-6 abolishes the effects of the other two cytokines on the HPA-axis (236). TNF-α and IL-1, on the other hand, stimulate the production of IL-6, which in turn stimulates the HPA-axis. The final end-product of HPA activation, glucocorticoids, inhibit IL-6 secretion at the transcriptional level through interaction of the ligand-activated glucocorticoid receptor with nuclear factor-kappa B, creating a negative feedback loop. In this way, IL-6 stimulates glucocorticoid release to control inflammation, and glucocorticoids subsequently inhibit IL-6 release through a negative feedback loop preventing uncontrolled and potentially harmful sequalae of inflammatory mechanisms, including tissue damage (237,238).

 

In an older study involving patients with Cushing disease studied before and after transsphenoidal adenectomy, plasma IL-6 concentration was increased when patients were hypocortisolemic, experiencing symptoms of glucocorticoid deficiency, as part of the “steroid withdrawal syndrome” (i.e., pyrexia, headache, anorexia, nausea, fatigue, malaise, arthralgias, myalgias, and somnolence of variable degree). Notably, IL-6 levels did not increase in patients who did not become hypocortisolemic after surgery, while following glucocorticoid replacement, a dramatic decrease of IL-6 levels concomitantly with relief of the observed symptoms was reported (239).

 

While acute stimulation with IL-6 activates the HPA axis mainly through the hypothalamic CRH neurons, chronic exposure to IL-6 may also directly stimulate the pituitary corticotropic cells and the adrenal cells via CRH receptor-independent mechanisms (240).

 

In vivo experiments have shown a stimulatory effect of IL-6 on cortisol production by the adrenal cortex in the absence of CRH and subsequent activation of the HPA axis (240) in cytomegalovirus-infected mice (241), as well as in murine colitis (242). In addition, experiments in mice deficient in CRH (CRH KO) and deficient in both CRH and IL-6 (CRH KO)/IL-6 KO) have demonstrated that IL-6 during prolonged immunological challenge may surpass CRH in augmentation of adrenal function (240). Protein expression of IL-6 and IL-6 receptor has been detected in primary cultures of human adrenocortical cells depleted of macrophages (CD68-positive cells), predominantly in the zona reticularis but also in the zona fasciculata and in single cells within the zona glomerulosa and the medulla (243). Moreover, IL-6 was able to induce adrenal steroidogenesis in vitro in a time- and dose-dependent manner in the absence of macrophages, suggesting that IL-6 may be a long-term stimulator of steroidogenesis but with no acute effects (243).

 

In humans, IL-6 may stimulate cortisol release directly at the level of the adrenal gland in long-term stress situations (244), and the same may also apply in chronic inflammatory diseases, although direct evidence is still lacking (245). In a recent clinical study, increased daily, and especially evening, saliva cortisol secretion, in the context of the acute viral infection COVID-19, appeared to be mostly driven by hypersecretion of IL-6, independently of ACTH (246). However, the hypothesis that IL-6 may partially replace ACTH when there is an acute requirement for increased cortisol secretion has yet to be tested.

 

PROLONGED OR CHRONIC STRESS AND INFLAMMATION

 

Acute versus chronic stress and inflammation are distinct conditions that exert extremely different effects on the immune system, each altering its function in a distinct manner. Chronic stress is associated with a blunted circadian cortisol rhythm, a suppressed inflammatory response, and a shift from Th1   to Th 2 and a Th reg to Th 17 immunity. It has clearly been shown that in chronic stress, endogenous glucocorticoids fail to terminate the stress response and, in fact, cause body composition changes reminiscent of those in hypercortisolism, such as visceral adiposity, sarcopenia, and osteoporosis (247-249).

 

On the other hand, following a period of intense stress, there may be glucocorticoid-induced suppression of the HPA axis (238). In this case, long-term “inadequate” cortisol secretion may unleash its inhibitory effect on immune system activation, resulting in immune dysregulation and expression of sickness-syndrome manifestations. In this case, the post-stress sustained HPA axis suppression and hypocortisolism is reminiscent of the clinical picture of the intrinsic hypocortisolemia of primary adrenal insufficiency (Addison’s disease), associated with immune perturbations due to failure of cortisol to appropriately suppress the increased secretion of proinflammatory cytokines (239,250).

 

The subsequent protracted lack of axis recovery due to a post-illness state of hypocortisolism in probably predisposed individuals may explain the underlying pathophysiologic mechanisms responsible for some post-viral infection sickness syndromes (232,251), such as long COVID syndrome (252).

 

AUTOIMMUNE DISORDERS  

 

In some cases, chronic stress can contribute to the development or exacerbation of autoimmune disorders. Without appropriate cortisol regulation in cases of chronic stress and chronic inflammation, the organism fails to downregulate inflammatory processes, contributing to a vicious cycle where stress, inflammation, and compromised HPA axis function may result in the development of chronic immune-mediated inflammatory diseases, such as rheumatoid arthritis. Stress can potentially trigger or worsen these conditions by dysregulating the immune response.

 

Cortisol diurnal secretion displays a circadian rhythm in healthy individuals, with the highest levels in the morning and a gradual decline throughout the day, reaching the lowest levels around midnight (253). During acute stress (254-256) or infection (237,257), the rhythm is disturbed and the afternoon and/or midnight cortisol levels do not drop. A disturbed circadian cortisol rhythm with abnormally high afternoon and night cortisol levels was found in patients with even mild or moderate COVID-19 compared to healthy controls (246). However, although not systematically studied, the adrenal response to chronic versus acute exposure to proinflammatory cytokines appears to follow a different pattern.

 

In patients with rheumatoid arthritis, symptoms follow circadian rhythms with impaired function due to pain and joint stiffness being most severe in the early morning (258,259) because of increased proinflammatory cytokines, such as TNF-a and IL-6, that occur during late night hours (260,261). This explains the inverse relation between diurnal variation of circulating IL-6 and glucocorticoid levels (262). Increased endogenous nocturnal secretion of cortisol could alleviate these morning symptoms, but this is not the case in most patients with rheumatoid arthritis (263-270). To clarify this issue, several studies have been carried out reporting that the optimal time for delivery of glucocorticoid treatment would be during the night in order to target the effects of nocturnal proinflammatory stimuli (271-274).

 

PSYCHOLOGICAL WELL-BEING

 

The immune system also plays a role in maintaining overall psychological well-being. Chronic stress and its impact on the immune system can increase susceptibility to mental health problems such as anxiety and depression. Conversely, psychological well-being can improve the human body’s immune responses, enhance resistance to diseases (including infectious diseases), and improve mental health (275,276). Changes in psychological status of patients with Alzheimer’s induced significant differences in their immune response (277).

In addition, long-term practice of meditation was shown to decrease stress reactivity and exert a favorable therapeutic effect in chronic inflammatory conditions characterized by neurogenic inflammation (278), while joyful activities such as singing were able to boost the immune response in cancer patients and family members (279).

 

ADRENAL MEDULLA

 

The chromaffin cells of the adrenal medulla play a role in stress response by secreting catecholamines and various biologically active peptides (238). As the stress response starts, rapidly augmented secretion of norepinephrine and epinephrine is initiated, followed by activation of the HPA axis and increased release of CRH and ACTH and secretion of glucocorticoids (280). CRH and norepinephrine stimulate the secretion of each other through CRH-R1 and α1-noradrenergic receptors, respectively (281).

 

Cytokines TNF-α, IL-1, and IFN-γ act directly on chromaffin cells (282-285). It has also been demonstrated that cytokines regulate the secretion of various peptides that are co-secreted with catecholamines, such as vasoactive intestinal peptide (VIP), galanin and secretogranin II, enkephalin and neuropeptide Y (283,285). IL-6 directly modulates the secretion of catecholamines and neuropeptides by chromaffin cells and therefore influences the adrenal stress response. It has been hypothesized that medullary peptides may serve as paracrine modulators of glucocorticoid production (286). It has also been shown that IL-6 increases intracellular Ca2+ concentration and induces catecholamine secretion in rat carotid body glomus cells, a finding which has finally confirmed the relations between IL-6 and catecholamine secretion (287). Furthermore, IL-10 is a critical target downstream of epinephrine and norepinephrine which limits inflammation (288). On the other hand, norepinephrine may act directly on macrophages and dendritic cells to suppress inflammatory cytokine secretion through primarily the β2-adrenergic receptors that are expressed by both innate and adaptive immune cells (289,290).

 

Although scientific advances of the last decade have shed light on the previously unrecognized major role of the catecholamines epinephrine and norepinephrine in controlling the immune system, much remains to be discovered, and further revelations will reveal new therapeutic targets in the management of inflammation.

 

ACKNOWLEDGEMENTS

 

This chapter is an update of a previous chapter and the authors would like to thank Professor Gregory Kaltsas, an author of the prior chapter.

 

REFERENCES

 

  1. Poland GA, Quill H, Togias A. Understanding the human immune system in the 21st century: the Human Immunology Project Consortium. Vaccine. 2013;31(28):2911-2912.
  2. Li Q, Wang B, Mu K, Zhang JA. The pathogenesis of thyroid autoimmune diseases: New T lymphocytes - Cytokines circuits beyond the Th1-Th2 paradigm. J Cell Physiol. 2019;234(3):2204-2216.
  3. Dunn GP, Old LJ, Schreiber RD. The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 2004;21(2):137-148.
  4. McLachlan SM, Rapoport B. Breaking tolerance to thyroid antigens: changing concepts in thyroid autoimmunity. Endocr Rev. 2014;35(1):59-105.
  5. Colaci M, Malatino L, Antonelli A, Fallahi P, Giuggioli D, Ferri C. Endocrine disorders associated with hepatitis C virus chronic infection. Rev Endocr Metab Disord. 2018;19(4):397-403.
  6. Wang B, Shao X, Song R, Xu D, Zhang JA. The Emerging Role of Epigenetics in Autoimmune Thyroid Diseases. Front Immunol. 2017;8:396.
  7. Boguslawska J, Godlewska M, Gajda E, Piekielko-Witkowska A. Cellular and molecular basis of thyroid autoimmunity. Eur Thyroid J. 2022;11(1).
  8. Coscia F, Taler-Vercic A, Chang VT, Sinn L, O'Reilly FJ, Izore T, Renko M, Berger I, Rappsilber J, Turk D, Lowe J. The structure of human thyroglobulin. Nature. 2020;578(7796):627-630.
  9. McLachlan SM, Rapoport B. Thyroid Autoantibodies Display both "Original Antigenic Sin" and Epitope Spreading. Front Immunol. 2017;8:1845.
  10. Godlewska M, Banga PJ. Thyroid peroxidase as a dual active site enzyme: Focus on biosynthesis, hormonogenesis and thyroid disorders of autoimmunity and cancer. Biochimie. 2019;160:34-45.
  11. Godlewska M, Gawel D, Buckle AM, Banga JP. Thyroid Peroxidase Revisited - What's New? Horm Metab Res. 2019;51(12):765-769.
  12. Jaume JC, Burek CL, Hoffman WH, Rose NR, McLachlan SM, Rapoport B. Thyroid peroxidase autoantibody epitopic 'fingerprints' in juvenile Hashimoto's thyroiditis: evidence for conservation over time and in families. Clin Exp Immunol. 1996;104(1):115-123.
  13. Morshed SA, Ando T, Latif R, Davies TF. Neutral antibodies to the TSH receptor are present in Graves disease and regulate selective signaling cascades. Endocrinology. 2010;151(11):5537-5549.
  14. Sanders J, Miguel RN, Bolton J, Bhardwaja A, Sanders P, Nakatake N, Evans M, Furmaniak J, Smith BR. Molecular interactions between the TSH receptor and a Thyroid-stimulating monoclonal autoantibody. Thyroid. 2007;17(8):699-706.
  15. Sanders P, Young S, Sanders J, Kabelis K, Baker S, Sullivan A, Evans M, Clark J, Wilmot J, Hu X, Roberts E, Powell M, Nunez Miguel R, Furmaniak J, Rees Smith B. Crystal structure of the TSH receptor (TSHR) bound to a blocking-type TSHR autoantibody. J Mol Endocrinol. 2011;46(2):81-99.
  16. Eleftheriadou AM, Mehl S, Renko K, Kasim RH, Schaefer JA, Minich WB, Schomburg L. Re-visiting autoimmunity to sodium-iodide symporter and pendrin in thyroid disease. Eur J Endocrinol. 2020;183(6):571-580.
  17. Fallahi P, Ferrari SM, Ragusa F, Ruffilli I, Elia G, Paparo SR, Antonelli A. Th1 Chemokines in Autoimmune Endocrine Disorders. J Clin Endocrinol Metab. 2020;105(4).
  18. Meeusen EN, Premier RR, Brandon MR. Tissue-specific migration of lymphocytes: a key role for Th1 and Th2 cells? Immunol Today. 1996;17(9):421-424.
  19. Davies TF, Andersen S, Latif R, Nagayama Y, Barbesino G, Brito M, Eckstein AK, Stagnaro-Green A, Kahaly GJ. Graves disease. Nat Rev Dis Primers. 2020;6(1):52.
  20. Wiersinga WM. Advances in treatment of active, moderate-to-severe Graves ophthalmopathy. Lancet Diabetes Endocrinol. 2017;5(2):134-142.
  21. Neumann S, Krieger CC, Gershengorn MC. Targeting TSH and IGF-1 Receptors to Treat Thyroid Eye Disease. Eur Thyroid J. 2020;9(Suppl 1):59-65.
  22. Taheri M, Eghtedarian R, Dinger ME, Ghafouri-Fard S. Dysregulation of non-coding RNAs in autoimmune thyroid disease. Exp Mol Pathol. 2020;117:104527.
  23. Liu Y, Cui X, Wang S, Liu J, Zhao N, Huang M, Qin J, Li Y, Shan Z, Teng W. Elevated MicroRNA-326 Levels Regulate the IL-23/IL-23R/Th17 Cell Axis in Hashimoto's Thyroiditis by Targeting a Disintegrin and Metalloprotease 17. Thyroid. 2020;30(9):1327-1337.
  24. Zheng L, Zhuang C, Wang X, Ming L. Serum miR-146a, miR-155, and miR-210 as potential markers of Graves disease. J Clin Lab Anal. 2018;32(2).
  25. Jiang X, Wang Y, Li X, He L, Yang Q, Wang W, Liu J, Zha B. Microarray profile of B cells from Graves disease patients reveals biomarkers of proliferation. Endocr Connect. 2020;9(5):405-417.
  26. Xiong S, Peng H, Ding X, Wang X, Wang L, Wu C, Wang S, Xu H, Liu Y. Circular RNA Expression Profiling and the Potential Role of hsa_circ_0089172 in Hashimoto's Thyroiditis via Sponging miR125a-3p. Mol Ther Nucleic Acids. 2019;17:38-48.
  27. Talebi S, Karimifar M, Heidari Z, Mohammadi H, Askari G. The effects of synbiotic supplementation on thyroid function and inflammation in hypothyroid patients: A randomized, double‑blind, placebo‑controlled trial. Complement Ther Med. 2020;48:102234.
  28. Biscarini F, Masetti G, Muller I, Verhasselt HL, Covelli D, Colucci G, Zhang L, Draman MS, Okosieme O, Taylor P, Daumerie C, Burlacu MC, Marino M, Ezra DG, Perros P, Plummer S, Eckstein A, Salvi M, Marchesi JR, Ludgate M. Gut Microbiome Associated With Graves Disease and Graves Orbitopathy: The INDIGO Multicenter European Study. J Clin Endocrinol Metab. 2023;108(8):2065-2077.
  29. Olivieri A, De Angelis S, Vaccari V, Valensise H, Magnani F, Stazi MA, Cotichini R, Gilardi E, Cordeddu V, Sorcini M, Boirivant M. Postpartum thyroiditis is associated with fluctuations in transforming growth factor-beta1 serum levels. J Clin Endocrinol Metab. 2003;88(3):1280-1284.
  30. Weetman AP, Bennett GL, Wong WL. Thyroid follicular cells produce interleukin-8. J Clin Endocrinol Metab. 1992;75(1):328-330.
  31. Premawardhana LD, Parkes AB, Ammari F, John R, Darke C, Adams H, Lazarus JH. Postpartum thyroiditis and long-term thyroid status: prognostic influence of thyroid peroxidase antibodies and ultrasound echogenicity. J Clin Endocrinol Metab. 2000;85(1):71-75.
  32. Papanicolaou DA. Euthyroid Sick Syndrome and the role of cytokines. Rev Endocr Metab Disord. 2000;1(1-2):43-48.
  33. Simons PJ, Delemarre FG, Drexhage HA. Antigen-presenting dendritic cells as regulators of the growth of thyrocytes: a role of interleukin-1beta and interleukin-6. Endocrinology. 1998;139(7):3148-3156.
  34. Asakawa H, Hanafusa T, Kobayashi T, Takai S, Kono N, Tarui S. Interferon-gamma reduces the thyroid peroxidase content of cultured human thyrocytes and inhibits its increase induced by thyrotropin. J Clin Endocrinol Metab. 1992;74(6):1331-1335.
  35. Ashizawa K, Yamashita S, Nagayama Y, Kimura H, Hirayu H, Izumi M, Nagataki S. Interferon-gamma inhibits thyrotropin-induced thyroidal peroxidase gene expression in cultured human thyrocytes. J Clin Endocrinol Metab. 1989;69(2):475-477.
  36. Poth M, Tseng YC, Wartofsky L. Inhibition of TSH activation of human cultured thyroid cells by tumor necrosis factor: an explanation for decreased thyroid function in systemic illness? Thyroid. 1991;1(3):235-240.
  37. Yamashita S, Kimura H, Ashizawa K, Nagayama Y, Hirayu H, Izumi M, Nagataki S. Interleukin-1 inhibits thyrotrophin-induced human thyroglobulin gene expression. J Endocrinol. 1989;122(1):177-183.
  38. van der Poll T, Romijn JA, Wiersinga WM, Sauerwein HP. Tumor necrosis factor: a putative mediator of the sick euthyroid syndrome in man. J Clin Endocrinol Metab. 1990;71(6):1567-1572.
  39. Chopra IJ, Sakane S, Teco GN. A study of the serum concentration of tumor necrosis factor-alpha in thyroidal and nonthyroidal illnesses. J Clin Endocrinol Metab. 1991;72(5):1113-1116.
  40. Girvent M, Maestro S, Hernandez R, Carajol I, Monne J, Sancho JJ, Gubern JM, Sitges-Serra A. Euthyroid sick syndrome, associated endocrine abnormalities, and outcome in elderly patients undergoing emergency operation. Surgery. 1998;123(5):560-567.
  41. Davis PJ. Cytokines and growth factors and thyroid hormone. Curr Opin Endocrinol Diabetes Obes. 2008;15(5):428.
  42. Baur A, Bauer K, Jarry H, Kohrle J. Effects of proinflammatory cytokines on anterior pituitary 5'-deiodinase type I and type II. J Endocrinol. 2000;167(3):505-515.
  43. Calikoglu M, Sahin G, Unlu A, Ozturk C, Tamer L, Ercan B, Kanik A, Atik U. Leptin and TNF-alpha levels in patients with chronic obstructive pulmonary disease and their relationship to nutritional parameters. Respiration. 2004;71(1):45-50.
  44. Park MC, Lee SW, Choi ST, Park YB, Lee SK. Serum leptin levels correlate with interleukin-6 levels and disease activity in patients with ankylosing spondylitis. Scand J Rheumatol. 2007;36(2):101-106.
  45. Lechan RM, Fekete C. Feedback regulation of thyrotropin-releasing hormone (TRH): mechanisms for the non-thyroidal illness syndrome. J Endocrinol Invest. 2004;27(6 Suppl):105-119.
  46. Harris M, Aschkenasi C, Elias CF, Chandrankunnel A, Nillni EA, Bjoorbaek C, Elmquist JK, Flier JS, Hollenberg AN. Transcriptional regulation of the thyrotropin-releasing hormone gene by leptin and melanocortin signaling. J Clin Invest. 2001;107(1):111-120.
  47. Harjai KJ, Licata AA. Effects of amiodarone on thyroid function. Ann Intern Med. 1997;126(1):63-73.
  48. Cohen-Lehman J, Dahl P, Danzi S, Klein I. Effects of amiodarone therapy on thyroid function. Nat Rev Endocrinol. 2010;6(1):34-41.
  49. Goundan PN, Lee SL. Thyroid effects of amiodarone: clinical update. Curr Opin Endocrinol Diabetes Obes. 2020;27(5):329-334.
  50. Newman CM, Price A, Davies DW, Gray TA, Weetman AP. Amiodarone and the thyroid: a practical guide to the management of thyroid dysfunction induced by amiodarone therapy. Heart. 1998;79(2):121-127.
  51. Narayana SK, Woods DR, Boos CJ. Management of amiodarone-related thyroid problems. Ther Adv Endocrinol Metab. 2011;2(3):115-126.
  52. Loh KC. Amiodarone-induced thyroid disorders: a clinical review. Postgrad Med J. 2000;76(893):133-140.
  53. Martino E, Bartalena L, Bogazzi F, Braverman LE. The effects of amiodarone on the thyroid. Endocr Rev. 2001;22(2):240-254.
  54. Bartalena L, Brogioni S, Grasso L, Bogazzi F, Burelli A, Martino E. Treatment of amiodarone-induced thyrotoxicosis, a difficult challenge: results of a prospective study. J Clin Endocrinol Metab. 1996;81(8):2930-2933.
  55. Mosher MC. Amiodarone-induced hypothyroidism and other adverse effects. Dimens Crit Care Nurs. 2011;30(2):87-93.
  56. Piga M, Serra A, Boi F, Tanda ML, Martino E, Mariotti S. Amiodarone-induced thyrotoxicosis. A review. Minerva Endocrinol. 2008;33(3):213-228.
  57. Kent WD, Hall SF, Isotalo PA, Houlden RL, George RL, Groome PA. Increased incidence of differentiated thyroid carcinoma and detection of subclinical disease. CMAJ. 2007;177(11):1357-1361.
  58. Cunha LL, Marcello MA, Ward LS. The role of the inflammatory microenvironment in thyroid carcinogenesis. Endocr Relat Cancer. 2014;21(3):R85-R103.
  59. Zeng R, Lyu Y, Zhang G, Shou T, Wang K, Niu H, Yan X. Positive effect of RORgammat on the prognosis of thyroid papillary carcinoma patients combined with Hashimoto's thyroiditis. Am J Transl Res. 2018;10(10):3011-3024.
  60. Kim EY, Kim WG, Kim WB, Kim TY, Kim JM, Ryu JS, Hong SJ, Gong G, Shong YK. Coexistence of chronic lymphocytic thyroiditis is associated with lower recurrence rates in patients with papillary thyroid carcinoma. Clin Endocrinol (Oxf). 2009;71(4):581-586.
  61. Zivancevic-Simonovic S, Mihaljevic O, Majstorovic I, Popovic S, Markovic S, Milosevic-Djordjevic O, Jovanovic Z, Mijatovic-Teodorovic L, Mihajlovic D, Colic M. Cytokine production in patients with papillary thyroid cancer and associated autoimmune Hashimoto thyroiditis. Cancer Immunol Immunother. 2015;64(8):1011-1019.
  62. Veit F, Graf D, Momberger S, Helmich-Kapp B, Ruschenburg I, Peters A, Kussmann J, Saeger W, Schmidt KW, Toetsch M, Nestler K, Mann K. Papillary Thyroid Cancer and Coexisting Autoimmune Thyroiditis. Horm Metab Res. 2017;49(11):869-872.
  63. Chowdhury S, Veyhl J, Jessa F, Polyakova O, Alenzi A, MacMillan C, Ralhan R, Walfish PG. Programmed death-ligand 1 overexpression is a prognostic marker for aggressive papillary thyroid cancer and its variants. Oncotarget. 2016;7(22):32318-32328.
  64. Dias Lopes NM, Mendonca Lens HH, Armani A, Marinello PC, Cecchini AL. Thyroid cancer and thyroid autoimmune disease: A review of molecular aspects and clinical outcomes. Pathol Res Pract. 2020;216(9):153098.
  65. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20(11):651-668.
  66. Na KJ, Choi H. Immune landscape of papillary thyroid cancer and immunotherapeutic implications. Endocr Relat Cancer. 2018;25(5):523-531.
  67. Ryder M, Ghossein RA, Ricarte-Filho JC, Knauf JA, Fagin JA. Increased density of tumor-associated macrophages is associated with decreased survival in advanced thyroid cancer. Endocr Relat Cancer. 2008;15(4):1069-1074.
  68. French JD, Kotnis GR, Said S, Raeburn CD, McIntyre RC, Jr., Klopper JP, Haugen BR. Programmed death-1+ T cells and regulatory T cells are enriched in tumor-involved lymph nodes and associated with aggressive features in papillary thyroid cancer. J Clin Endocrinol Metab. 2012;97(6):E934-943.
  69. Gogali F, Paterakis G, Rassidakis GZ, Kaltsas G, Liakou CI, Gousis P, Neonakis E, Manoussakis MN, Liapi C. Phenotypical analysis of lymphocytes with suppressive and regulatory properties (Tregs) and NK cells in the papillary carcinoma of thyroid. J Clin Endocrinol Metab. 2012;97(5):1474-1482.
  70. Garcia-Alvarez A, Hernando J, Carmona-Alonso A, Capdevila J. What is the status of immunotherapy in thyroid neoplasms? Front Endocrinol (Lausanne). 2022;13:929091.
  71. Mould RC, van Vloten JP, AuYeung AWK, Karimi K, Bridle BW. Immune responses in the thyroid cancer microenvironment: making immunotherapy a possible mission. Endocr Relat Cancer. 2017;24(12):T311-T329.
  72. Pan J, Ye F, Yu C, Zhu Q, Li J, Zhang Y, Tian H, Yao Y, Zhu M, Shen Y, Zhu F, Wang Y, Zhou X, Guo G, Wu Y. Papillary Thyroid Carcinoma Landscape and Its Immunological Link With Hashimoto Thyroiditis at Single-Cell Resolution. Front Cell Dev Biol. 2021;9:758339.
  73. Galdiero MR, Varricchi G, Marone G. The immune network in thyroid cancer. Oncoimmunology. 2016;5(6):e1168556.
  74. Ahn S, Kim TH, Kim SW, Ki CS, Jang HW, Kim JS, Kim JH, Choe JH, Shin JH, Hahn SY, Oh YL, Chung JH. Comprehensive screening for PD-L1 expression in thyroid cancer. Endocr Relat Cancer. 2017;24(2):97-106.
  75. Cameselle-Garcia S, Abdulkader-Sande S, Sanchez-Ares M, Rodriguez-Carnero G, Garcia-Gomez J, Gude-Sampedro F, Abdulkader-Nallib I, Cameselle-Teijeiro JM. PD-L1 expression and immune cells in anaplastic carcinoma and poorly differentiated carcinoma of the human thyroid gland: A retrospective study. Oncol Lett. 2021;22(1):553.
  76. Cantara S, Bertelli E, Occhini R, Regoli M, Brilli L, Pacini F, Castagna MG, Toti P. Blockade of the programmed death ligand 1 (PD-L1) as potential therapy for anaplastic thyroid cancer. Endocrine. 2019;64(1):122-129.
  77. Chintakuntlawar AV, Rumilla KM, Smith CY, Jenkins SM, Foote RL, Kasperbauer JL, Morris JC, Ryder M, Alsidawi S, Hilger C, Bible KC. Expression of PD-1 and PD-L1 in Anaplastic Thyroid Cancer Patients Treated With Multimodal Therapy: Results From a Retrospective Study. J Clin Endocrinol Metab. 2017;102(6):1943-1950.
  78. Zwaenepoel K, Jacobs J, De Meulenaere A, Silence K, Smits E, Siozopoulou V, Hauben E, Rolfo C, Rottey S, Pauwels P. CD70 and PD-L1 in anaplastic thyroid cancer - promising targets for immunotherapy. Histopathology. 2017;71(3):357-365.
  79. Wu H, Sun Y, Ye H, Yang S, Lee SL, de las Morenas A. Anaplastic thyroid cancer: outcome and the mutation/expression profiles of potential targets. Pathol Oncol Res. 2015;21(3):695-701.
  80. Strickler JH, Hanks BA, Khasraw M. Tumor Mutational Burden as a Predictor of Immunotherapy Response: Is More Always Better? Clin Cancer Res. 2021;27(5):1236-1241.
  81. Mehnert JM, Varga A, Brose MS, Aggarwal RR, Lin CC, Prawira A, de Braud F, Tamura K, Doi T, Piha-Paul SA, Gilbert J, Saraf S, Thanigaimani P, Cheng JD, Keam B. Safety and antitumor activity of the anti-PD-1 antibody pembrolizumab in patients with advanced, PD-L1-positive papillary or follicular thyroid cancer. BMC Cancer. 2019;19(1):196.
  82. Bluestone JA, Herold K, Eisenbarth G. Genetics, pathogenesis and clinical interventions in type 1 diabetes. Nature. 2010;464(7293):1293-1300.
  83. Erlich H, Valdes AM, Noble J, Carlson JA, Varney M, Concannon P, Mychaleckyj JC, Todd JA, Bonella P, Fear AL, Lavant E, Louey A, Moonsamy P, Type 1 Diabetes Genetics C. HLA DR-DQ haplotypes and genotypes and type 1 diabetes risk: analysis of the type 1 diabetes genetics consortium families. Diabetes. 2008;57(4):1084-1092.
  84. Willcox A, Richardson SJ, Bone AJ, Foulis AK, Morgan NG. Analysis of islet inflammation in human type 1 diabetes. Clin Exp Immunol. 2009;155(2):173-181.
  85. Vandamme C, Kinnunen T. B cell helper T cells and type 1 diabetes. Scand J Immunol. 2020;92(4):e12943.
  86. Pugliese A. Insulitis in the pathogenesis of type 1 diabetes. Pediatr Diabetes. 2016;17 Suppl 22(Suppl Suppl 22):31-36.
  87. Mitchell AM, Michels AW. Self-Antigens Targeted by Regulatory T Cells in Type 1 Diabetes. Int J Mol Sci. 2022;23(6).
  88. Atkinson MA, Bowman MA, Campbell L, Darrow BL, Kaufman DL, Maclaren NK. Cellular immunity to a determinant common to glutamate decarboxylase and coxsackie virus in insulin-dependent diabetes. J Clin Invest. 1994;94(5):2125-2129.
  89. Tough DF, Borrow P, Sprent J. Induction of bystander T cell proliferation by viruses and type I interferon in vivo. Science. 1996;272(5270):1947-1950.
  90. Katz JD, Benoist C, Mathis D. T helper cell subsets in insulin-dependent diabetes. Science. 1995;268(5214):1185-1188.
  91. Han S, Donelan W, Wang H, Reeves W, Yang LJ. Novel autoantigens in type 1 diabetes. Am J Transl Res. 2013;5(4):379-392.
  92. Ziegler AG, Nepom GT. Prediction and pathogenesis in type 1 diabetes. Immunity. 2010;32(4):468-478.
  93. Burton AR, Vincent E, Arnold PY, Lennon GP, Smeltzer M, Li CS, Haskins K, Hutton J, Tisch RM, Sercarz EE, Santamaria P, Workman CJ, Vignali DA. On the pathogenicity of autoantigen-specific T-cell receptors. Diabetes. 2008;57(5):1321-1330.
  94. Serreze DV, Fleming SA, Chapman HD, Richard SD, Leiter EH, Tisch RM. B lymphocytes are critical antigen-presenting cells for the initiation of T cell-mediated autoimmune diabetes in nonobese diabetic mice. J Immunol. 1998;161(8):3912-3918.
  95. Sutherland AP, Van Belle T, Wurster AL, Suto A, Michaud M, Zhang D, Grusby MJ, von Herrath M. Interleukin-21 is required for the development of type 1 diabetes in NOD mice. Diabetes. 2009;58(5):1144-1155.
  96. Lechleitner M, Koch T, Herold M, Hoppichler F. Relationship of tumor necrosis factor-alpha plasma levels to metabolic control in type 1 diabetes. Diabetes Care. 1999;22(10):1749.
  97. Farhan J, Al-Shobaili HA, Zafar U, Al Salloom A, Meki AR, Rasheed Z. Interleukin-6: a possible inflammatory link between vitiligo and type 1 diabetes. Br J Biomed Sci. 2014;71(4):151-157.
  98. Zhang J, Huang Z, Sun R, Tian Z, Wei H. IFN-gamma induced by IL-12 administration prevents diabetes by inhibiting pathogenic IL-17 production in NOD mice. J Autoimmun. 2012;38(1):20-28.
  99. Arif S, Moore F, Marks K, Bouckenooghe T, Dayan CM, Planas R, Vives-Pi M, Powrie J, Tree T, Marchetti P, Huang GC, Gurzov EN, Pujol-Borrell R, Eizirik DL, Peakman M. Peripheral and islet interleukin-17 pathway activation characterizes human autoimmune diabetes and promotes cytokine-mediated beta-cell death. Diabetes. 2011;60(8):2112-2119.
  100. Jacobsen LM, Newby BN, Perry DJ, Posgai AL, Haller MJ, Brusko TM. Immune Mechanisms and Pathways Targeted in Type 1 Diabetes. Curr Diab Rep. 2018;18(10):90.
  101. Smigoc Schweiger D. Recent Advances in Immune-based Therapies for Type 1 Diabetes. Horm Res Paediatr. 2022.
  102. An Anti-CD3 Antibody, Teplizumab, in Relatives at Risk for Type 1 Diabetes. N Engl J Med. 2020;382(6):586.
  103. Eizirik DL, Szymczak F, Mallone R. Why does the immune system destroy pancreatic beta-cells but not alpha-cells in type 1 diabetes? Nat Rev Endocrinol. 2023;19(7):425-434.
  104. Bosco D, Armanet M, Morel P, Niclauss N, Sgroi A, Muller YD, Giovannoni L, Parnaud G, Berney T. Unique arrangement of alpha- and beta-cells in human islets of Langerhans. Diabetes. 2010;59(5):1202-1210.
  105. Campbell JE, Newgard CB. Mechanisms controlling pancreatic islet cell function in insulin secretion. Nat Rev Mol Cell Biol. 2021;22(2):142-158.
  106. Gromada J, Chabosseau P, Rutter GA. The alpha-cell in diabetes mellitus. Nat Rev Endocrinol. 2018;14(12):694-704.
  107. Brissova M, Haliyur R, Saunders D, Shrestha S, Dai C, Blodgett DM, Bottino R, Campbell-Thompson M, Aramandla R, Poffenberger G, Lindner J, Pan FC, von Herrath MG, Greiner DL, Shultz LD, Sanyoura M, Philipson LH, Atkinson M, Harlan DM, Levy SE, Prasad N, Stein R, Powers AC. alpha Cell Function and Gene Expression Are Compromised in Type 1 Diabetes. Cell Rep. 2018;22(10):2667-2676.
  108. Shapira SN, Naji A, Atkinson MA, Powers AC, Kaestner KH. Understanding islet dysfunction in type 2 diabetes through multidimensional pancreatic phenotyping: The Human Pancreas Analysis Program. Cell Metab. 2022;34(12):1906-1913.
  109. Fasolino M, Schwartz GW, Patil AR, Mongia A, Golson ML, Wang YJ, Morgan A, Liu C, Schug J, Liu J, Wu M, Traum D, Kondo A, May CL, Goldman N, Wang W, Feldman M, Moore JH, Japp AS, Betts MR, Consortium H, Faryabi RB, Naji A, Kaestner KH, Vahedi G. Single-cell multi-omics analysis of human pancreatic islets reveals novel cellular states in type 1 diabetes. Nat Metab. 2022;4(2):284-299.
  110. Kaestner KH, Powers AC, Naji A, Consortium H, Atkinson MA. NIH Initiative to Improve Understanding of the Pancreas, Islet, and Autoimmunity in Type 1 Diabetes: The Human Pancreas Analysis Program (HPAP). Diabetes. 2019;68(7):1394-1402.
  111. Doliba NM, Rozo AV, Roman J, Qin W, Traum D, Gao L, Liu J, Manduchi E, Liu C, Golson ML, Vahedi G, Naji A, Matschinsky FM, Atkinson MA, Powers AC, Brissova M, Kaestner KH, Stoffers DA, Consortium H. alpha Cell dysfunction in islets from nondiabetic, glutamic acid decarboxylase autoantibody-positive individuals. J Clin Invest. 2022;132(11).
  112. Colli ML, Ramos-Rodriguez M, Nakayasu ES, Alvelos MI, Lopes M, Hill JLE, Turatsinze JV, Coomans de Brachene A, Russell MA, Raurell-Vila H, Castela A, Juan-Mateu J, Webb-Robertson BM, Krogvold L, Dahl-Jorgensen K, Marselli L, Marchetti P, Richardson SJ, Morgan NG, Metz TO, Pasquali L, Eizirik DL. An integrated multi-omics approach identifies the landscape of interferon-alpha-mediated responses of human pancreatic beta cells. Nat Commun. 2020;11(1):2584.
  113. Chen CW, Guan BJ, Alzahrani MR, Gao Z, Gao L, Bracey S, Wu J, Mbow CA, Jobava R, Haataja L, Zalavadia AH, Schaffer AE, Lee H, LaFramboise T, Bederman I, Arvan P, Mathews CE, Gerling IC, Kaestner KH, Tirosh B, Engin F, Hatzoglou M. Adaptation to chronic ER stress enforces pancreatic beta-cell plasticity. Nat Commun. 2022;13(1):4621.
  114. Thorel F, Damond N, Chera S, Wiederkehr A, Thorens B, Meda P, Wollheim CB, Herrera PL. Normal glucagon signaling and beta-cell function after near-total alpha-cell ablation in adult mice. Diabetes. 2011;60(11):2872-2882.
  115. Shaw JE, Sicree RA, Zimmet PZ. Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 2010;87(1):4-14.
  116. Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, Malanda B. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271-281.
  117. Donath MY, Storling J, Berchtold LA, Billestrup N, Mandrup-Poulsen T. Cytokines and beta-cell biology: from concept to clinical translation. Endocr Rev. 2008;29(3):334-350.
  118. Varma V, Yao-Borengasser A, Rasouli N, Nolen GT, Phanavanh B, Starks T, Gurley C, Simpson P, McGehee RE, Jr., Kern PA, Peterson CA. Muscle inflammatory response and insulin resistance: synergistic interaction between macrophages and fatty acids leads to impaired insulin action. Am J Physiol Endocrinol Metab. 2009;296(6):E1300-1310.
  119. Ehses JA, Boni-Schnetzler M, Faulenbach M, Donath MY. Macrophages, cytokines and beta-cell death in Type 2 diabetes. Biochem Soc Trans. 2008;36(Pt 3):340-342.
  120. Shoelson SE, Lee J, Goldfine AB. Inflammation and insulin resistance. J Clin Invest. 2006;116(7):1793-1801.
  121. Sell H, Habich C, Eckel J. Adaptive immunity in obesity and insulin resistance. Nat Rev Endocrinol. 2012;8(12):709-716.
  122. Nikolajczyk BS, Jagannathan-Bogdan M, Shin H, Gyurko R. State of the union between metabolism and the immune system in type 2 diabetes. Genes Immun. 2011;12(4):239-250.
  123. Hotamisligil GS. The role of TNFalpha and TNF receptors in obesity and insulin resistance. J Intern Med. 1999;245(6):621-625.
  124. Hotamisligil GS, Arner P, Caro JF, Atkinson RL, Spiegelman BM. Increased adipose tissue expression of tumor necrosis factor-alpha in human obesity and insulin resistance. J Clin Invest. 1995;95(5):2409-2415.
  125. Hotamisligil GS, Shargill NS, Spiegelman BM. Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance. Science. 1993;259(5091):87-91.
  126. Kershaw EE, Flier JS. Adipose tissue as an endocrine organ. J Clin Endocrinol Metab. 2004;89(6):2548-2556.
  127. Halberg N, Wernstedt-Asterholm I, Scherer PE. The adipocyte as an endocrine cell. Endocrinol Metab Clin North Am. 2008;37(3):753-768, x-xi.
  128. Gao Z, He Q, Peng B, Chiao PJ, Ye J. Regulation of nuclear translocation of HDAC3 by IkappaBalpha is required for tumor necrosis factor inhibition of peroxisome proliferator-activated receptor gamma function. J Biol Chem. 2006;281(7):4540-4547.
  129. Yuan M, Konstantopoulos N, Lee J, Hansen L, Li ZW, Karin M, Shoelson SE. Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta. Science. 2001;293(5535):1673-1677.
  130. Aguirre V, Uchida T, Yenush L, Davis R, White MF. The c-Jun NH(2)-terminal kinase promotes insulin resistance during association with insulin receptor substrate-1 and phosphorylation of Ser(307). J Biol Chem. 2000;275(12):9047-9054.
  131. Ye J. Regulation of PPARgamma function by TNF-alpha. Biochem Biophys Res Commun. 2008;374(3):405-408.
  132. Ruan H, Miles PD, Ladd CM, Ross K, Golub TR, Olefsky JM, Lodish HF. Profiling gene transcription in vivo reveals adipose tissue as an immediate target of tumor necrosis factor-alpha: implications for insulin resistance. Diabetes. 2002;51(11):3176-3188.
  133. Berbudi A, Rahmadika N, Tjahjadi AI, Ruslami R. Type 2 Diabetes and its Impact on the Immune System. Curr Diabetes Rev. 2020;16(5):442-449.
  134. Duncan BB, Schmidt MI, Offenbacher S, Wu KK, Savage PJ, Heiss G. Factor VIII and other hemostasis variables are related to incident diabetes in adults. The Atherosclerosis Risk in Communities (ARIC) Study. Diabetes Care. 1999;22(5):767-772.
  135. Schmidt MI, Duncan BB, Sharrett AR, Lindberg G, Savage PJ, Offenbacher S, Azambuja MI, Tracy RP, Heiss G. Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. Lancet. 1999;353(9165):1649-1652.
  136. Festa A, D'Agostino R, Jr., Tracy RP, Haffner SM. Elevated levels of acute-phase proteins and plasminogen activator inhibitor-1 predict the development of type 2 diabetes: the insulin resistance atherosclerosis study. Diabetes. 2002;51(4):1131-1137.
  137. Herder C, Baumert J, Thorand B, Koenig W, de Jager W, Meisinger C, Illig T, Martin S, Kolb H. Chemokines as risk factors for type 2 diabetes: results from the MONICA/KORA Augsburg study, 1984-2002. Diabetologia. 2006;49(5):921-929.
  138. Lee CC, Adler AI, Sandhu MS, Sharp SJ, Forouhi NG, Erqou S, Luben R, Bingham S, Khaw KT, Wareham NJ. Association of C-reactive protein with type 2 diabetes: prospective analysis and meta-analysis. Diabetologia. 2009;52(6):1040-1047.
  139. Lin JD, Chao TC, Weng HF, Lin KD. The roles of cytokines and retinoic acid in the regulation of human thyroid cancer cell growth. Cytokine. 1998;10(7):536-539.
  140. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab. 2001;280(5):E745-751.
  141. Kirwan JP, Hauguel-De Mouzon S, Lepercq J, Challier JC, Huston-Presley L, Friedman JE, Kalhan SC, Catalano PM. TNF-alpha is a predictor of insulin resistance in human pregnancy. Diabetes. 2002;51(7):2207-2213.
  142. Vilcek J, Lee TH. Tumor necrosis factor. New insights into the molecular mechanisms of its multiple actions. J Biol Chem. 1991;266(12):7313-7316.
  143. Cornelius P, Lee MD, Marlowe M, Pekala PH. Monokine regulation of glucose transporter mRNA in L6 myotubes. Biochem Biophys Res Commun. 1989;165(1):429-436.
  144. Stephens JM, Pekala PH. Transcriptional repression of the GLUT4 and C/EBP genes in 3T3-L1 adipocytes by tumor necrosis factor-alpha. J Biol Chem. 1991;266(32):21839-21845.
  145. Meigs JB, Larson MG, Fox CS, Keaney JF, Jr., Vasan RS, Benjamin EJ. Association of oxidative stress, insulin resistance, and diabetes risk phenotypes: the Framingham Offspring Study. Diabetes Care. 2007;30(10):2529-2535.
  146. Rosen P, Nawroth PP, King G, Moller W, Tritschler HJ, Packer L. The role of oxidative stress in the onset and progression of diabetes and its complications: a summary of a Congress Series sponsored by UNESCO-MCBN, the American Diabetes Association and the German Diabetes Society. Diabetes Metab Res Rev. 2001;17(3):189-212.
  147. Hasnain SZ, Borg DJ, Harcourt BE, Tong H, Sheng YH, Ng CP, Das I, Wang R, Chen AC, Loudovaris T, Kay TW, Thomas HE, Whitehead JP, Forbes JM, Prins JB, McGuckin MA. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med. 2014;20(12):1417-1426.
  148. Huma ZE, Sanchez J, Lim HD, Bridgford JL, Huang C, Parker BJ, Pazhamalil JG, Porebski BT, Pfleger KDG, Lane JR, Canals M, Stone MJ. Key determinants of selective binding and activation by the monocyte chemoattractant proteins at the chemokine receptor CCR2. Sci Signal. 2017;10(480).
  149. Catalan V, Gomez-Ambrosi J, Ramirez B, Rotellar F, Pastor C, Silva C, Rodriguez A, Gil MJ, Cienfuegos JA, Fruhbeck G. Proinflammatory cytokines in obesity: impact of type 2 diabetes mellitus and gastric bypass. Obes Surg. 2007;17(11):1464-1474.
  150. Franca CN, Izar MCO, Hortencio MNS, do Amaral JB, Ferreira CES, Tuleta ID, Fonseca FAH. Monocyte subtypes and the CCR2 chemokine receptor in cardiovascular disease. Clin Sci (Lond). 2017;131(12):1215-1224.
  151. Gokulakrishnan K, Ranjani H, Weber MB, Pandey GK, Anjana RM, Balasubramanyam M, Prabhakaran D, Tandon N, Narayan KM, Mohan V. Effect of lifestyle improvement program on the biomarkers of adiposity, inflammation and gut hormones in overweight/obese Asian Indians with prediabetes. Acta Diabetol. 2017;54(9):843-852.
  152. Townsend SA, Newsome PN. Review article: new treatments in non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2017;46(5):494-507.
  153. Chistiakov DA, Melnichenko AA, Grechko AV, Myasoedova VA, Orekhov AN. Potential of anti-inflammatory agents for treatment of atherosclerosis. Exp Mol Pathol. 2018;104(2):114-124.
  154. Di Muzio C, Cipriani P, Ruscitti P. Rheumatoid Arthritis Treatment Options and Type 2 Diabetes: Unravelling the Association. BioDrugs. 2022;36(6):673-685.
  155. Larsen CM, Faulenbach M, Vaag A, Volund A, Ehses JA, Seifert B, Mandrup-Poulsen T, Donath MY. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;356(15):1517-1526.
  156. Malozowski S, Sahlroot JT. Interleukin-1-receptor antagonist in type 2 diabetes mellitus. N Engl J Med. 2007;357(3):302-303; author reply 303.
  157. Larsen CM, Faulenbach M, Vaag A, Ehses JA, Donath MY, Mandrup-Poulsen T. Sustained effects of interleukin-1 receptor antagonist treatment in type 2 diabetes. Diabetes Care. 2009;32(9):1663-1668.
  158. Ridker PM, Howard CP, Walter V, Everett B, Libby P, Hensen J, Thuren T, Group CPI. Effects of interleukin-1beta inhibition with canakinumab on hemoglobin A1c, lipids, C-reactive protein, interleukin-6, and fibrinogen: a phase IIb randomized, placebo-controlled trial. Circulation. 2012;126(23):2739-2748.
  159. Everett BM, Donath MY, Pradhan AD, Thuren T, Pais P, Nicolau JC, Glynn RJ, Libby P, Ridker PM. Anti-Inflammatory Therapy With Canakinumab for the Prevention and Management of Diabetes. J Am Coll Cardiol. 2018;71(21):2392-2401.
  160. Otsuka Y, Kiyohara C, Kashiwado Y, Sawabe T, Nagano S, Kimoto Y, Ayano M, Mitoma H, Akahoshi M, Arinobu Y, Niiro H, Akashi K, Horiuchi T. Effects of tumor necrosis factor inhibitors and tocilizumab on the glycosylated hemoglobin levels in patients with rheumatoid arthritis; an observational study. PLoS One. 2018;13(4):e0196368.
  161. Ogata A, Morishima A, Hirano T, Hishitani Y, Hagihara K, Shima Y, Narazaki M, Tanaka T. Improvement of HbA1c during treatment with humanised anti-interleukin 6 receptor antibody, tocilizumab. Ann Rheum Dis. 2011;70(6):1164-1165.
  162. Castaneda S, Remuzgo-Martinez S, Lopez-Mejias R, Genre F, Calvo-Alen J, Llorente I, Aurrecoechea E, Ortiz AM, Triguero A, Blanco R, Llorca J, Gonzalez-Gay MA. Rapid beneficial effect of the IL-6 receptor blockade on insulin resistance and insulin sensitivity in non-diabetic patients with rheumatoid arthritis. Clin Exp Rheumatol. 2019;37(3):465-473.
  163. Makrilakis K, Fragiadaki K, Smith J, Sfikakis PP, Kitas GD. Interrelated reduction of chemerin and plasminogen activator inhibitor-1 serum levels in rheumatoid arthritis after interleukin-6 receptor blockade. Clin Rheumatol. 2015;34(3):419-427.
  164. Tournadre A, Pereira B, Dutheil F, Giraud C, Courteix D, Sapin V, Frayssac T, Mathieu S, Malochet-Guinamand S, Soubrier M. Changes in body composition and metabolic profile during interleukin 6 inhibition in rheumatoid arthritis. J Cachexia Sarcopenia Muscle. 2017;8(4):639-646.
  165. Genovese MC, Burmester GR, Hagino O, Thangavelu K, Iglesias-Rodriguez M, John GS, Gonzalez-Gay MA, Mandrup-Poulsen T, Fleischmann R. Interleukin-6 receptor blockade or TNFalpha inhibition for reducing glycaemia in patients with RA and diabetes: post hoc analyses of three randomised, controlled trials. Arthritis Res Ther. 2020;22(1):206.
  166. Genovese MC, Fleischmann R, Kivitz AJ, Rell-Bakalarska M, Martincova R, Fiore S, Rohane P, van Hoogstraten H, Garg A, Fan C, van Adelsberg J, Weinstein SP, Graham NM, Stahl N, Yancopoulos GD, Huizinga TW, van der Heijde D. Sarilumab Plus Methotrexate in Patients With Active Rheumatoid Arthritis and Inadequate Response to Methotrexate: Results of a Phase III Study. Arthritis Rheumatol. 2015;67(6):1424-1437.
  167. Burmester GR, Lin Y, Patel R, van Adelsberg J, Mangan EK, Graham NM, van Hoogstraten H, Bauer D, Ignacio Vargas J, Lee EB. Efficacy and safety of sarilumab monotherapy versus adalimumab monotherapy for the treatment of patients with active rheumatoid arthritis (MONARCH): a randomised, double-blind, parallel-group phase III trial. Ann Rheum Dis. 2017;76(5):840-847.
  168. Fleischmann R, van Adelsberg J, Lin Y, Castelar-Pinheiro GD, Brzezicki J, Hrycaj P, Graham NM, van Hoogstraten H, Bauer D, Burmester GR. Sarilumab and Nonbiologic Disease-Modifying Antirheumatic Drugs in Patients With Active Rheumatoid Arthritis and Inadequate Response or Intolerance to Tumor Necrosis Factor Inhibitors. Arthritis Rheumatol. 2017;69(2):277-290.
  169. Gonzalez-Gay MA, De Matias JM, Gonzalez-Juanatey C, Garcia-Porrua C, Sanchez-Andrade A, Martin J, Llorca J. Anti-tumor necrosis factor-alpha blockade improves insulin resistance in patients with rheumatoid arthritis. Clin Exp Rheumatol. 2006;24(1):83-86.
  170. Solomon DH, Massarotti E, Garg R, Liu J, Canning C, Schneeweiss S. Association between disease-modifying antirheumatic drugs and diabetes risk in patients with rheumatoid arthritis and psoriasis. JAMA. 2011;305(24):2525-2531.
  171. Lin C, Ji H, Cai X, Yang W, Lv F, Ji L. The association between the biological disease-modifying anti-rheumatic drugs and the incidence of diabetes: A systematic review and meta-analysis. Pharmacol Res. 2020;161:105216.
  172. Tessaro FHG, Ayala TS, Nolasco EL, Bella LM, Martins JO. Insulin Influences LPS-Induced TNF-alpha and IL-6 Release Through Distinct Pathways in Mouse Macrophages from Different Compartments. Cell Physiol Biochem. 2017;42(5):2093-2104.
  173. Kumar M, Roe K, Nerurkar PV, Orillo B, Thompson KS, Verma S, Nerurkar VR. Reduced immune cell infiltration and increased pro-inflammatory mediators in the brain of Type 2 diabetic mouse model infected with West Nile virus. J Neuroinflammation. 2014;11:80.
  174. Martinez N, Ketheesan N, Martens GW, West K, Lien E, Kornfeld H. Defects in early cell recruitment contribute to the increased susceptibility to respiratory Klebsiella pneumoniae infection in diabetic mice. Microbes Infect. 2016;18(10):649-655.
  175. Gupta S, Maratha A, Siednienko J, Natarajan A, Gajanayake T, Hoashi S, Miggin S. Analysis of inflammatory cytokine and TLR expression levels in Type 2 Diabetes with complications. Sci Rep. 2017;7(1):7633.
  176. Chao WC, Yen CL, Wu YH, Chen SY, Hsieh CY, Chang TC, Ou HY, Shieh CC. Increased resistin may suppress reactive oxygen species production and inflammasome activation in type 2 diabetic patients with pulmonary tuberculosis infection. Microbes Infect. 2015;17(3):195-204.
  177. Stegenga ME, van der Crabben SN, Blumer RM, Levi M, Meijers JC, Serlie MJ, Tanck MW, Sauerwein HP, van der Poll T. Hyperglycemia enhances coagulation and reduces neutrophil degranulation, whereas hyperinsulinemia inhibits fibrinolysis during human endotoxemia. Blood. 2008;112(1):82-89.
  178. Joshi MB, Lad A, Bharath Prasad AS, Balakrishnan A, Ramachandra L, Satyamoorthy K. High glucose modulates IL-6 mediated immune homeostasis through impeding neutrophil extracellular trap formation. FEBS Lett. 2013;587(14):2241-2246.
  179. Restrepo BI, Twahirwa M, Rahbar MH, Schlesinger LS. Phagocytosis via complement or Fc-gamma receptors is compromised in monocytes from type 2 diabetes patients with chronic hyperglycemia. PLoS One. 2014;9(3):e92977.
  180. Berrou J, Fougeray S, Venot M, Chardiny V, Gautier JF, Dulphy N, Toubert A, Peraldi MN. Natural killer cell function, an important target for infection and tumor protection, is impaired in type 2 diabetes. PLoS One. 2013;8(4):e62418.
  181. Mauriello CT, Hair PS, Rohn RD, Rister NS, Krishna NK, Cunnion KM. Hyperglycemia inhibits complement-mediated immunological control of S. aureus in a rat model of peritonitis. J Diabetes Res. 2014;2014:762051.
  182. Ponzetti M, Rucci N. Updates on Osteoimmunology: What's New on the Cross-Talk Between Bone and Immune System. Front Endocrinol (Lausanne). 2019;10:236.
  183. Huang F, Wong P, Li J, Lv Z, Xu L, Zhu G, He M, Luo Y. Osteoimmunology: The correlation between osteoclasts and the Th17/Treg balance in osteoporosis. J Cell Mol Med. 2022;26(13):3591-3597.
  184. Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Front Immunol. 2018;9:657.
  185. Ahmad SS, Ahmed F, Ali R, Ghoneim MM, Alshehri S, Najmi AK, Ahmad S, Ahmad MZ, Ahmad J, Khan MA. Immunology of osteoporosis: relevance of inflammatory targets for the development of novel interventions. Immunotherapy. 2022;14(10):815-831.
  186. Srivastava RK, Sapra L. The Rising Era of "Immunoporosis": Role of Immune System in the Pathophysiology of Osteoporosis. J Inflamm Res. 2022;15:1667-1698.
  187. Saxena Y, Routh S, Mukhopadhaya A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front Immunol. 2021;12:687037.
  188. Zhang W, Gao R, Rong X, Zhu S, Cui Y, Liu H, Li M. Immunoporosis: Role of immune system in the pathophysiology of different types of osteoporosis. Front Endocrinol (Lausanne). 2022;13:965258.
  189. Xiong J, Piemontese M, Onal M, Campbell J, Goellner JJ, Dusevich V, Bonewald L, Manolagas SC, O'Brien CA. Osteocytes, not Osteoblasts or Lining Cells, are the Main Source of the RANKL Required for Osteoclast Formation in Remodeling Bone. PLoS One. 2015;10(9):e0138189.
  190. Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21(2):115-137.
  191. Walsh MC, Choi Y. Biology of the RANKL-RANK-OPG System in Immunity, Bone, and Beyond. Front Immunol. 2014;5:511.
  192. Pietschmann P, Mechtcheriakova D, Meshcheryakova A, Foger-Samwald U, Ellinger I. Immunology of Osteoporosis: A Mini-Review. Gerontology. 2016;62(2):128-137.
  193. Lorenzo J. Cytokines and Bone: Osteoimmunology. Handb Exp Pharmacol. 2020;262:177-230.
  194. Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J. Regulation of Osteoclast Differentiation by Cytokine Networks. Immune Netw. 2018;18(1):e8.
  195. Rauner M, Sipos W, Pietschmann P. Osteoimmunology. Int Arch Allergy Immunol. 2007;143(1):31-48.
  196. Weitzmann MN, Cenci S, Rifas L, Brown C, Pacifici R. Interleukin-7 stimulates osteoclast formation by up-regulating the T-cell production of soluble osteoclastogenic cytokines. Blood. 2000;96(5):1873-1878.
  197. Titanji K, Vunnava A, Sheth AN, Delille C, Lennox JL, Sanford SE, Foster A, Knezevic A, Easley KA, Weitzmann MN, Ofotokun I. Dysregulated B cell expression of RANKL and OPG correlates with loss of bone mineral density in HIV infection. PLoS Pathog. 2014;10(10):e1004497.
  198. Manolagas SC, Jilka RL, Girasole G, Passeri G, Bellido T. Estrogen, cytokines, and the control of osteoclast formation and bone resorption in vitro and in vivo. Osteoporos Int. 1993;3 Suppl 1:114-116.
  199. Klein B, Wijdenes J, Zhang XG, Jourdan M, Boiron JM, Brochier J, Liautard J, Merlin M, Clement C, Morel-Fournier B, et al. Murine anti-interleukin-6 monoclonal antibody therapy for a patient with plasma cell leukemia. Blood. 1991;78(5):1198-1204.
  200. Roodman GD, Kurihara N, Ohsaki Y, Kukita A, Hosking D, Demulder A, Smith JF, Singer FR. Interleukin 6. A potential autocrine/paracrine factor in Paget's disease of bone. J Clin Invest. 1992;89(1):46-52.
  201. Kotake S, Sato K, Kim KJ, Takahashi N, Udagawa N, Nakamura I, Yamaguchi A, Kishimoto T, Suda T, Kashiwazaki S. Interleukin-6 and soluble interleukin-6 receptors in the synovial fluids from rheumatoid arthritis patients are responsible for osteoclast-like cell formation. J Bone Miner Res. 1996;11(1):88-95.
  202. Devlin RD, Bone HG, 3rd, Roodman GD. Interleukin-6: a potential mediator of the massive osteolysis in patients with Gorham-Stout disease. J Clin Endocrinol Metab. 1996;81(5):1893-1897.
  203. Martin TJ, Allan EH, Evely RS, Reid IR. Leukaemia inhibitory factor and bone cell function. Ciba Found Symp. 1992;167:141-150; discussion 150-145.
  204. Wei S, Kitaura H, Zhou P, Ross FP, Teitelbaum SL. IL-1 mediates TNF-induced osteoclastogenesis. J Clin Invest. 2005;115(2):282-290.
  205. Ruscitti P, Cipriani P, Carubbi F, Liakouli V, Zazzeroni F, Di Benedetto P, Berardicurti O, Alesse E, Giacomelli R. The role of IL-1beta in the bone loss during rheumatic diseases. Mediators Inflamm. 2015;2015:782382.
  206. D'Amelio P. The immune system and postmenopausal osteoporosis. Immunol Invest. 2013;42(7):544-554.
  207. Cenci S, Toraldo G, Weitzmann MN, Roggia C, Gao Y, Qian WP, Sierra O, Pacifici R. Estrogen deficiency induces bone loss by increasing T cell proliferation and lifespan through IFN-gamma-induced class II transactivator. Proc Natl Acad Sci U S A. 2003;100(18):10405-10410.
  208. Miyaura C, Onoe Y, Inada M, Maki K, Ikuta K, Ito M, Suda T. Increased B-lymphopoiesis by interleukin 7 induces bone loss in mice with intact ovarian function: similarity to estrogen deficiency. Proc Natl Acad Sci U S A. 1997;94(17):9360-9365.
  209. Jabbar S, Drury J, Fordham JN, Datta HK, Francis RM, Tuck SP. Osteoprotegerin, RANKL and bone turnover in postmenopausal osteoporosis. J Clin Pathol. 2011;64(4):354-357.
  210. Fischer V, Haffner-Luntzer M. Interaction between bone and immune cells: Implications for postmenopausal osteoporosis. Semin Cell Dev Biol. 2022;123:14-21.
  211. Zha L, He L, Liang Y, Qin H, Yu B, Chang L, Xue L. TNF-alpha contributes to postmenopausal osteoporosis by synergistically promoting RANKL-induced osteoclast formation. Biomed Pharmacother. 2018;102:369-374.
  212. Onal M, Xiong J, Chen X, Thostenson JD, Almeida M, Manolagas SC, O'Brien CA. Receptor activator of nuclear factor kappaB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J Biol Chem. 2012;287(35):29851-29860.
  213. Wu D, Cline-Smith A, Shashkova E, Perla A, Katyal A, Aurora R. T-Cell Mediated Inflammation in Postmenopausal Osteoporosis. Front Immunol. 2021;12:687551.
  214. Lai N, Zhang Z, Wang B, Miao X, Guo Y, Yao C, Wang Z, Wang L, Ma R, Li X, Jiang G. Regulatory effect of traditional Chinese medicinal formula Zuo-Gui-Wan on the Th17/Treg paradigm in mice with bone loss induced by estrogen deficiency. J Ethnopharmacol. 2015;166:228-239.
  215. Duque G, Troen BR. Understanding the mechanisms of senile osteoporosis: new facts for a major geriatric syndrome. J Am Geriatr Soc. 2008;56(5):935-941.
  216. De Maeyer RPH, Chambers ES. The impact of ageing on monocytes and macrophages. Immunol Lett. 2021;230:1-10.
  217. Li CJ, Xiao Y, Sun YC, He WZ, Liu L, Huang M, He C, Huang M, Chen KX, Hou J, Feng X, Su T, Guo Q, Huang Y, Peng H, Yang M, Liu GH, Luo XH. Senescent immune cells release grancalcin to promote skeletal aging. Cell Metab. 2022;34(1):184-185.
  218. Sandmand M, Bruunsgaard H, Kemp K, Andersen-Ranberg K, Pedersen AN, Skinhoj P, Pedersen BK. Is ageing associated with a shift in the balance between Type 1 and Type 2 cytokines in humans? Clin Exp Immunol. 2002;127(1):107-114.
  219. Siddiqi A, Monson JP, Wood DF, Besser GM, Burrin JM. Serum cytokines in thyrotoxicosis. J Clin Endocrinol Metab. 1999;84(2):435-439.
  220. Sun L, Zhu LL, Lu P, Yuen T, Li J, Ma R, Baliram R, Moonga SS, Liu P, Zallone A, New MI, Davies TF, Zaidi M. Genetic confirmation for a central role for TNFalpha in the direct action of thyroid stimulating hormone on the skeleton. Proc Natl Acad Sci U S A. 2013;110(24):9891-9896.
  221. Baliram R, Sun L, Cao J, Li J, Latif R, Huber AK, Yuen T, Blair HC, Zaidi M, Davies TF. Hyperthyroid-associated osteoporosis is exacerbated by the loss of TSH signaling. J Clin Invest. 2012;122(10):3737-3741.
  222. Grey A, Mitnick MA, Shapses S, Ellison A, Gundberg C, Insogna K. Circulating levels of interleukin-6 and tumor necrosis factor-alpha are elevated in primary hyperparathyroidism and correlate with markers of bone resorption--a clinical research center study. J Clin Endocrinol Metab. 1996;81(10):3450-3454.
  223. Grey A, Mitnick MA, Masiukiewicz U, Sun BH, Rudikoff S, Jilka RL, Manolagas SC, Insogna K. A role for interleukin-6 in parathyroid hormone-induced bone resorption in vivo. Endocrinology. 1999;140(10):4683-4690.
  224. Hory BG, Roussanne MC, Rostand S, Bourdeau A, Drueke TB, Gogusev J. Absence of response to human parathyroid hormone in athymic mice grafted with human parathyroid adenoma, hyperplasia or parathyroid cells maintained in culture. J Endocrinol Invest. 2000;23(5):273-279.
  225. Bedi B, Li JY, Tawfeek H, Baek KH, Adams J, Vangara SS, Chang MK, Kneissel M, Weitzmann MN, Pacifici R. Silencing of parathyroid hormone (PTH) receptor 1 in T cells blunts the bone anabolic activity of PTH. Proc Natl Acad Sci U S A. 2012;109(12):E725-733.
  226. Weitzmann MN. Bone and the Immune System. Toxicol Pathol. 2017;45(7):911-924.
  227. Song L, Cao L, Liu R, Ma H, Li Y, Shang Q, Zheng Z, Zhang L, Zhang W, Han Y, Zhang X, Yang H, Wang Y, Melino G, Shao C, Shi Y. The critical role of T cells in glucocorticoid-induced osteoporosis. Cell Death Dis. 2020;12(1):45.
  228. Ponnapakkam T, Katikaneni R, Nichols T, Tobin G, Sakon J, Matsushita O, Gensure RC. Prevention of chemotherapy-induced osteoporosis by cyclophosphamide with a long-acting form of parathyroid hormone. J Endocrinol Invest. 2011;34(11):e392-397.
  229. Wang W, Gao Y, Liu H, Feng W, Li X, Guo J, Li M. Eldecalcitol, an active vitamin D analog, effectively prevents cyclophosphamide-induced osteoporosis in rats. Exp Ther Med. 2019;18(3):1571-1580.
  230. Zhao D, Wang C, Zhao Y, Shu B, Jia Y, Liu S, Wang H, Chang J, Dai W, Lu S, Shi Q, Yang Y, Zhang Y, Wang Y. Cyclophosphamide causes osteoporosis in C57BL/6 male mice: suppressive effects of cyclophosphamide on osteoblastogenesis and osteoclastogenesis. Oncotarget. 2017;8(58):98163-98183.
  231. Chen X, Wang S, Chen G, Wang Z, Kan J. The immunomodulatory effects of Carapax Trionycis ultrafine powder on cyclophosphamide-induced immunosuppression in Balb/c mice. J Sci Food Agric. 2021;101(5):2014-2026.
  232. Filippa MG, Tektonidou MG, Mantzou A, Kaltsas GA, Chrousos GP, Sfikakis PP, Yavropoulou MP. Adrenocortical dysfunction in rheumatoid arthritis: Alpha narrative review and future directions. Eur J Clin Invest. 2022;52(1):e13635.
  233. Nicolaides NC, Kyratzi E, Lamprokostopoulou A, Chrousos GP, Charmandari E. Stress, the stress system and the role of glucocorticoids. Neuroimmunomodulation. 2015;22(1-2):6-19.
  234. Laflamme N, Lacroix S, Rivest S. An essential role of interleukin-1beta in mediating NF-kappaB activity and COX-2 transcription in cells of the blood-brain barrier in response to a systemic and localized inflammation but not during endotoxemia. J Neurosci. 1999;19(24):10923-10930.
  235. Nadeau S, Rivest S. Regulation of the gene encoding tumor necrosis factor alpha (TNF-alpha) in the rat brain and pituitary in response in different models of systemic immune challenge. J Neuropathol Exp Neurol. 1999;58(1):61-77.
  236. Perlstein RS, Whitnall MH, Abrams JS, Mougey EH, Neta R. Synergistic roles of interleukin-6, interleukin-1, and tumor necrosis factor in the adrenocorticotropin response to bacterial lipopolysaccharide in vivo. Endocrinology. 1993;132(3):946-952.
  237. Chrousos GP, Kaltsas G. Post-SARS sickness syndrome manifestations and endocrinopathy: how, why, and so what? Clin Endocrinol (Oxf). 2005;63(4):363-365.
  238. Chrousos GP, Gold PW. The concepts of stress and stress system disorders. Overview of physical and behavioral homeostasis. JAMA. 1992;267(9):1244-1252.
  239. Papanicolaou DA, Tsigos C, Oldfield EH, Chrousos GP. Acute glucocorticoid deficiency is associated with plasma elevations of interleukin-6: does the latter participate in the symptomatology of the steroid withdrawal syndrome and adrenal insufficiency? J Clin Endocrinol Metab. 1996;81(6):2303-2306.
  240. Bethin KE, Vogt SK, Muglia LJ. Interleukin-6 is an essential, corticotropin-releasing hormone-independent stimulator of the adrenal axis during immune system activation. Proc Natl Acad Sci U S A. 2000;97(16):9317-9322.
  241. Silverman MN, Miller AH, Biron CA, Pearce BD. Characterization of an interleukin-6- and adrenocorticotropin-dependent, immune-to-adrenal pathway during viral infection. Endocrinology. 2004;145(8):3580-3589.
  242. Franchimont D, Bouma G, Galon J, Wolkersdorfer GW, Haidan A, Chrousos GP, Bornstein SR. Adrenal cortical activation in murine colitis. Gastroenterology. 2000;119(6):1560-1568.
  243. Path G, Bornstein SR, Ehrhart-Bornstein M, Scherbaum WA. Interleukin-6 and the interleukin-6 receptor in the human adrenal gland: expression and effects on steroidogenesis. J Clin Endocrinol Metab. 1997;82(7):2343-2349.
  244. Path G, Scherbaum WA, Bornstein SR. The role of interleukin-6 in the human adrenal gland. Eur J Clin Invest. 2000;30 Suppl 3:91-95.
  245. Filippa MG, Tektonidou MG, Mantzou A, Kaltsas GA, Chrousos GP, Sfikakis PP, Yavropoulou MP. Adrenocortical dysfunction in rheumatoid arthritis: Alpha narrative review and future directions. Eur J Clin Invest. 2021:e13635.
  246. Yavropoulou MP, Filippa MG, Mantzou A, Ntziora F, Mylona M, Tektonidou MG, Vlachogiannis NI, Paraskevis D, Kaltsas GA, Chrousos GP, Sfikakis PP. Alterations in cortisol and interleukin-6 secretion in patients with COVID-19 suggestive of neuroendocrine-immune adaptations. Endocrine. 2022;75(2):317-327.
  247. de Kloet ER. Stress in the brain. Eur J Pharmacol. 2000;405(1-3):187-198.
  248. Lightman SL, Wiles CC, Atkinson HC, Henley DE, Russell GM, Leendertz JA, McKenna MA, Spiga F, Wood SA, Conway-Campbell BL. The significance of glucocorticoid pulsatility. Eur J Pharmacol. 2008;583(2-3):255-262.
  249. Stavreva DA, Wiench M, John S, Conway-Campbell BL, McKenna MA, Pooley JR, Johnson TA, Voss TC, Lightman SL, Hager GL. Ultradian hormone stimulation induces glucocorticoid receptor-mediated pulses of gene transcription. Nat Cell Biol. 2009;11(9):1093-1102.
  250. Hochberg Z, Pacak K, Chrousos GP. Endocrine withdrawal syndromes. Endocr Rev. 2003;24(4):523-538.
  251. Yavropoulou MP, Filippa MG, Panopoulos S, Spanos E, Spanos G, Tektonidou MG, Sfikakis PP. Impaired adrenal cortex reserve in patients with rheumatic and musculoskeletal diseases who relapse upon tapering of low glucocorticoid dose. Clin Exp Rheumatol. 2022.
  252. Yavropoulou MP, Tsokos GC, Chrousos GP, Sfikakis PP. Protracted stress-induced hypocortisolemia may account for the clinical and immune manifestations of Long COVID. Clin Immunol. 2022;245:109133.
  253. Kirschbaum C, Hellhammer DH. Salivary cortisol in psychobiological research: an overview. Neuropsychobiology. 1989;22(3):150-169.
  254. Sephton SE, Sapolsky RM, Kraemer HC, Spiegel D. Diurnal cortisol rhythm as a predictor of breast cancer survival. J Natl Cancer Inst. 2000;92(12):994-1000.
  255. Pervanidou P, Kolaitis G, Charitaki S, Margeli A, Ferentinos S, Bakoula C, Lazaropoulou C, Papassotiriou I, Tsiantis J, Chrousos GP. Elevated morning serum interleukin (IL)-6 or evening salivary cortisol concentrations predict posttraumatic stress disorder in children and adolescents six months after a motor vehicle accident. Psychoneuroendocrinology. 2007;32(8-10):991-999.
  256. Bougea AM, Spandideas N, Alexopoulos EC, Thomaides T, Chrousos GP, Darviri C. Effect of the emotional freedom technique on perceived stress, quality of life, and cortisol salivary levels in tension-type headache sufferers: a randomized controlled trial. Explore (NY). 2013;9(2):91-99.
  257. Lopez-Acevo CA, Arrendondo-Loza E, Salinas-Carmona MC, Rendon A, Martinez-Castilla AM, Vazquez-Marmolejo AV, Munoz-Maldonado G, Rosas-Taraco AG. Cortisol and perceived stress are associated with cytokines levels in patients infected with influenza B virus. Cytokine. 2021;138:155400.
  258. Cutolo M, Villaggio B, Otsa K, Aakre O, Sulli A, Seriolo B. Altered circadian rhythms in rheumatoid arthritis patients play a role in the disease's symptoms. Autoimmun Rev. 2005;4(8):497-502.
  259. Straub RH, Cutolo M. Circadian rhythms in rheumatoid arthritis: implications for pathophysiology and therapeutic management. Arthritis Rheum. 2007;56(2):399-408.
  260. Cutolo M, Masi AT. Circadian rhythms and arthritis. Rheum Dis Clin North Am. 2005;31(1):115-129, ix-x.
  261. Cutolo M. Circadian rhythms and rheumatoid arthritis. Joint Bone Spine. 2019;86(3):327-333.
  262. Sothern RB, Roitman-Johnson B, Kanabrocki EL, Yager JG, Fuerstenberg RK, Weatherbee JA, Young MR, Nemchausky BM, Scheving LE. Circadian characteristics of interleukin-6 in blood and urine of clinically healthy men. In Vivo. 1995;9(4):331-339.
  263. Chikanza IC, Petrou P, Kingsley G, Chrousos G, Panayi GS. Defective hypothalamic response to immune and inflammatory stimuli in patients with rheumatoid arthritis. Arthritis Rheum. 1992;35(11):1281-1288.
  264. Gudbjornsson B, Skogseid B, Oberg K, Wide L, Hallgren R. Intact adrenocorticotropic hormone secretion but impaired cortisol response in patients with active rheumatoid arthritis. Effect of glucocorticoids. J Rheumatol. 1996;23(4):596-602.
  265. Crofford LJ, Kalogeras KT, Mastorakos G, Magiakou MA, Wells J, Kanik KS, Gold PW, Chrousos GP, Wilder RL. Circadian relationships between interleukin (IL)-6 and hypothalamic-pituitary-adrenal axis hormones: failure of IL-6 to cause sustained hypercortisolism in patients with early untreated rheumatoid arthritis. J Clin Endocrinol Metab. 1997;82(4):1279-1283.
  266. Gutierrez MA, Garcia ME, Rodriguez JA, Mardonez G, Jacobelli S, Rivero S. Hypothalamic-pituitary-adrenal axis function in patients with active rheumatoid arthritis: a controlled study using insulin hypoglycemia stress test and prolactin stimulation. J Rheumatol. 1999;26(2):277-281.
  267. Cutolo M, Foppiani L, Prete C, Ballarino P, Sulli A, Villaggio B, Seriolo B, Giusti M, Accardo S. Hypothalamic-pituitary-adrenocortical axis function in premenopausal women with rheumatoid arthritis not treated with glucocorticoids. J Rheumatol. 1999;26(2):282-288.
  268. Cutolo M, Foppiani L, Minuto F. Hypothalamic-pituitary-adrenal axis impairment in the pathogenesis of rheumatoid arthritis and polymyalgia rheumatica. J Endocrinol Invest. 2002;25(10 Suppl):19-23.
  269. Straub RH, Paimela L, Peltomaa R, Scholmerich J, Leirisalo-Repo M. Inadequately low serum levels of steroid hormones in relation to interleukin-6 and tumor necrosis factor in untreated patients with early rheumatoid arthritis and reactive arthritis. Arthritis Rheum. 2002;46(3):654-662.
  270. Imrich R, Vigas M, Rovensky J, Aldag JC, Masi AT. Adrenal plasma steroid relations in glucocorticoid-naive premenopausal rheumatoid arthritis patients during insulin-induced hypoglycemia test compared to matched normal control females. Endocr Regul. 2009;43(2):65-73.
  271. De Silva M, Binder A, Hazleman BL. The timing of prednisolone dosage and its effect on morning stiffness in rheumatoid arthritis. Ann Rheum Dis. 1984;43(6):790-793.
  272. Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, Jeka S, Krueger K, Szechinski J, Alten R. Efficacy of modified-release versus standard prednisone to reduce duration of morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet. 2008;371(9608):205-214.
  273. Buttgereit F, Doering G, Schaeffler A, Witte S, Sierakowski S, Gromnica-Ihle E, Jeka S, Krueger K, Szechinski J, Alten R. Targeting pathophysiological rhythms: prednisone chronotherapy shows sustained efficacy in rheumatoid arthritis. Ann Rheum Dis. 2010;69(7):1275-1280.
  274. Buttgereit F, Mehta D, Kirwan J, Szechinski J, Boers M, Alten RE, Supronik J, Szombati I, Romer U, Witte S, Saag KG. Low-dose prednisone chronotherapy for rheumatoid arthritis: a randomised clinical trial (CAPRA-2). Ann Rheum Dis. 2013;72(2):204-210.
  275. Nicolaides NC, Chrousos GP. Impact of Stress on Health in Childhood and Adolescence. Horm Res Paediatr. 2023;96(1):5-7.
  276. Tzelepi I, Bacopoulou F, Chrousos GP, Sotiropoulou L, Vlachakis D, Darviri C. Mindfulness and Academic Performance of College and University Students: A Systematic Review. Adv Exp Med Biol. 2023;1425:207-215.
  277. Redwine L, Mills PJ, Sada M, Dimsdale J, Patterson T, Grant I. Differential immune cell chemotaxis responses to acute psychological stress in Alzheimer caregivers compared to non-caregiver controls. Psychosom Med. 2004;66(5):770-775.
  278. Rosenkranz MA, Lutz A, Perlman DM, Bachhuber DR, Schuyler BS, MacCoon DG, Davidson RJ. Reduced stress and inflammatory responsiveness in experienced meditators compared to a matched healthy control group. Psychoneuroendocrinology. 2016;68:117-125.
  279. Fancourt D, Perkins R, Ascenso S, Carvalho LA, Steptoe A, Williamon A. Effects of Group Drumming Interventions on Anxiety, Depression, Social Resilience and Inflammatory Immune Response among Mental Health Service Users. PLoS One. 2016;11(3):e0151136.
  280. Sapolsky RM, Romero LM, Munck AU. How do glucocorticoids influence stress responses? Integrating permissive, suppressive, stimulatory, and preparative actions. Endocr Rev. 2000;21(1):55-89.
  281. Kiss A, Aguilera G. Participation of alpha 1-adrenergic receptors in the secretion of hypothalamic corticotropin-releasing hormone during stress. Neuroendocrinology. 1992;56(2):153-160.
  282. Bunn SJ, Ait-Ali D, Eiden LE. Immune-neuroendocrine integration at the adrenal gland: cytokine control of the adrenomedullary transcriptome. J Mol Neurosci. 2012;48(2):413-419.
  283. Ait-Ali D, Turquier V, Tanguy Y, Thouennon E, Ghzili H, Mounien L, Derambure C, Jegou S, Salier JP, Vaudry H, Eiden LE, Anouar Y. Tumor necrosis factor (TNF)-alpha persistently activates nuclear factor-kappaB signaling through the type 2 TNF receptor in chromaffin cells: implications for long-term regulation of neuropeptide gene expression in inflammation. Endocrinology. 2008;149(6):2840-2852.
  284. Douglas SA, Bunn SJ. Interferon-alpha signalling in bovine adrenal chromaffin cells: involvement of signal-transducer and activator of transcription 1 and 2, extracellular signal-regulated protein kinases 1/2 and serine 31 phosphorylation of tyrosine hydroxylase. J Neuroendocrinol. 2009;21(3):200-207.
  285. Rosmaninho-Salgado J, Araujo IM, Alvaro AR, Mendes AF, Ferreira L, Grouzmann E, Mota A, Duarte EP, Cavadas C. Regulation of catecholamine release and tyrosine hydroxylase in human adrenal chromaffin cells by interleukin-1beta: role of neuropeptide Y and nitric oxide. J Neurochem. 2009;109(3):911-922.
  286. Jenkins DE, Sreenivasan D, Carman F, Samal B, Eiden LE, Bunn SJ. Interleukin-6-mediated signaling in adrenal medullary chromaffin cells. J Neurochem. 2016;139(6):1138-1150.
  287. Fan J, Zhang B, Shu HF, Zhang XY, Wang X, Kuang F, Liu L, Peng ZW, Wu R, Zhou Z, Wang BR. Interleukin-6 increases intracellular Ca2+ concentration and induces catecholamine secretion in rat carotid body glomus cells. J Neurosci Res. 2009;87(12):2757-2762.
  288. Sharma D, Farrar JD. Adrenergic regulation of immune cell function and inflammation. Semin Immunopathol. 2020;42(6):709-717.
  289. Takenaka MC, Guereschi MG, Basso AS. Neuroimmune interactions: dendritic cell modulation by the sympathetic nervous system. Semin Immunopathol. 2017;39(2):165-176.
  290. Guyot M, Simon T, Panzolini C, Ceppo F, Daoudlarian D, Murris E, Macia E, Abelanet S, Sridhar A, Vervoordeldonk MJ, Glaichenhaus N, Blancou P. Apical splenic nerve electrical stimulation discloses an anti-inflammatory pathway relying on adrenergic and nicotinic receptors in myeloid cells. Brain Behav Immun. 2019;80:238-246.