A 66-year-old woman with an enlarged tongue

Article Type
Changed
Mon, 09/25/2017 - 14:42
Display Headline
A 66-year-old woman with an enlarged tongue

A 66-year-old woman was prompted by her dentist to seek medical attention for an unusually enlarged, smooth-appearing tongue (Figure 1). She also complained of fatigue, dyspnea on exertion, and tingling of her hands.

Figure 1.

Basic laboratory tests showed normocytic anemia and renal insufficiency. Her thyrotropin level was within normal limits. Serum protein electrophoresis showed a monoclonal M-spike, which prompted a bone marrow biopsy that was diagnostic of multiple myeloma.

Transthoracic echocardiography revealed diffuse hypokinesis with a restrictive filling pattern, myocardial thickening, and moderate mitral and tricuspid regurgitation, highly suggestive of an infiltrative cardiomyopathy. Biopsy of the right ventricle confirmed cardiac amyloidosis of the amyloid immunoglobulin light chain (AL) subtype.

The patient underwent chemotherapy, followed by autologous stem-cell transplantation. She achieved successful remission, and her cardiomyopathy was compensated.

AMYLOIDOSIS IS HETEROGENEOUS

Amyloidosis is a heterogeneous syndrome characterized by abnormal folding of proteins that deposit as insoluble fibrils in different tissues, impairing both structure and function. Virchow was the first to describe amyloid (from amylon, Greek for starch) as an abnormal material seen in postmortem examination of the liver. On Congo red staining, the extracellular proteins appear as salmon-red conglomerates, which also show apple-green birefringence under polarized light.

Amyloidosis can be localized but more often represents a systemic process, often associated with a plasma cell dyscrasia such as multiple myeloma.

Modern classification is based on the precursor protein,1 eg:

  • Light chains (AL)
  • Acute-phase protein (AA)
  • Beta-2-microglobulin (Aß2M)
  • Transthyretin (ATTR; occurring in senile systemic amyloidosis)
  • Other proteins (occurring in various forms of hereditary systemic amyloidosis).

AMYLOIDOSIS AND THE TONGUE

Macroglossia is defined as protrusion of the tongue beyond the alveolar ridge of the teeth at rest. When caused by amyloidosis, it is most often associated with the systemic AL variant and is present in 10% to 23% of patients with this subtype.2

On physical examination, tongue enlargement can present with lateral indentations, with a smooth contour or with nodular deposits. Less often, bullous lesions, vesicles, and ulcers can also be seen, particularly on the lips. Infiltration of salivary glands can result in xerostomia. Functional symptoms, such as hypogeusia, dysarthria, dysphagia, dysphonia, and, in advanced cases, upper-airway dysfunction can result from restricted mobility of the tongue and tethering to deeper structures.

Surgical management may be necessary if severe obstructive symptoms are present, but infiltrative lesions tend to recur.

AMYLOIDOSIS AND THE HEART

Cardiac involvement in amyloidosis is currently the primary determinant of prognosis.3 It is more often seen in the AL, senile, and hereditary forms. It usually manifests as diastolic heart failure, but angina, orthostatic hypotension, dysrhythmias, and syncope can also occur. Systolic dysfunction is typically a late finding in the course of the disease.

Although an electrocardiographic pattern of low voltage in the precordial and limb leads has been classically associated with cardiac amyloidosis, only 30% of patients with the senile and hereditary forms show this feature.4 Left ventricular hypertrophy on electrocardiography is thought to be uncommon, but it has been reported in 16% of patients with AL amyloidosis and biopsy-proven cardiac involvement.5 Cardiac troponin levels can be elevated, as seen in other infiltrative cardiomyopathies.3 The diagnosis can be established by endomyocardial biopsy or indirectly by cardiac imaging and a positive extracardiac biopsy.

Drug therapy is supportive and mainly involves diuretics, since angiotensin-converting enzyme inhibitors, beta-blockers, and calcium channel blockers may cause hypotension and exacerbate myocardial dysfunction. The specific treatment varies depending on the underlying cause of amyloidosis.

References
  1. Westermark P, Benson MD, Buxbaum JN, et al. A primer of amyloid nomenclature. Amyloid 2007; 14:179183.
  2. Kyle RA, Gertz MA. Primary systemic amyloidosis: clinical and laboratory features in 474 cases. Semin Hematol 1995; 32:4559.
  3. Kapoor P, Thenappan T, Singh E, Kumar S, Greipp PR. Cardiac amyloidosis: a practical approach to diagnosis and management. Am J Med 2011; 124:10061015.
  4. Ng B, Connors LH, Davidoff R, Skinner M, Falk RH. Senile systemic amyloidosis presenting with heart failure: a comparison with light chain-associated amyloidosis. Arch Intern Med 2005; 165:14251429.
  5. Murtagh B, Hammill SC, Gertz MA, Kyle RA, Tajik AJ, Grogan M. Electrocardiographic findings in primary systemic amyloidosis and biopsy-proven cardiac involvement. Am J Cardiol 2005; 95:535537.
Article PDF
Author and Disclosure Information

Jorge A. Brenes-Salazar, MD
Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN

Martha Grogan, MD
Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN

Martha Q. Lacy, MD
Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN

Address: Jorge A. Brenes-Salazar, MD, Mayo Clinic, 200 First St. SW, Rochester, MN 55905; e-mail: [email protected]

Issue
Cleveland Clinic Journal of Medicine - 80(11)
Publications
Topics
Page Number
696-697
Sections
Author and Disclosure Information

Jorge A. Brenes-Salazar, MD
Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN

Martha Grogan, MD
Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN

Martha Q. Lacy, MD
Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN

Address: Jorge A. Brenes-Salazar, MD, Mayo Clinic, 200 First St. SW, Rochester, MN 55905; e-mail: [email protected]

Author and Disclosure Information

Jorge A. Brenes-Salazar, MD
Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN

Martha Grogan, MD
Department of Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN

Martha Q. Lacy, MD
Department of Medicine, Division of Hematology, Mayo Clinic, Rochester, MN

Address: Jorge A. Brenes-Salazar, MD, Mayo Clinic, 200 First St. SW, Rochester, MN 55905; e-mail: [email protected]

Article PDF
Article PDF

A 66-year-old woman was prompted by her dentist to seek medical attention for an unusually enlarged, smooth-appearing tongue (Figure 1). She also complained of fatigue, dyspnea on exertion, and tingling of her hands.

Figure 1.

Basic laboratory tests showed normocytic anemia and renal insufficiency. Her thyrotropin level was within normal limits. Serum protein electrophoresis showed a monoclonal M-spike, which prompted a bone marrow biopsy that was diagnostic of multiple myeloma.

Transthoracic echocardiography revealed diffuse hypokinesis with a restrictive filling pattern, myocardial thickening, and moderate mitral and tricuspid regurgitation, highly suggestive of an infiltrative cardiomyopathy. Biopsy of the right ventricle confirmed cardiac amyloidosis of the amyloid immunoglobulin light chain (AL) subtype.

The patient underwent chemotherapy, followed by autologous stem-cell transplantation. She achieved successful remission, and her cardiomyopathy was compensated.

AMYLOIDOSIS IS HETEROGENEOUS

Amyloidosis is a heterogeneous syndrome characterized by abnormal folding of proteins that deposit as insoluble fibrils in different tissues, impairing both structure and function. Virchow was the first to describe amyloid (from amylon, Greek for starch) as an abnormal material seen in postmortem examination of the liver. On Congo red staining, the extracellular proteins appear as salmon-red conglomerates, which also show apple-green birefringence under polarized light.

Amyloidosis can be localized but more often represents a systemic process, often associated with a plasma cell dyscrasia such as multiple myeloma.

Modern classification is based on the precursor protein,1 eg:

  • Light chains (AL)
  • Acute-phase protein (AA)
  • Beta-2-microglobulin (Aß2M)
  • Transthyretin (ATTR; occurring in senile systemic amyloidosis)
  • Other proteins (occurring in various forms of hereditary systemic amyloidosis).

AMYLOIDOSIS AND THE TONGUE

Macroglossia is defined as protrusion of the tongue beyond the alveolar ridge of the teeth at rest. When caused by amyloidosis, it is most often associated with the systemic AL variant and is present in 10% to 23% of patients with this subtype.2

On physical examination, tongue enlargement can present with lateral indentations, with a smooth contour or with nodular deposits. Less often, bullous lesions, vesicles, and ulcers can also be seen, particularly on the lips. Infiltration of salivary glands can result in xerostomia. Functional symptoms, such as hypogeusia, dysarthria, dysphagia, dysphonia, and, in advanced cases, upper-airway dysfunction can result from restricted mobility of the tongue and tethering to deeper structures.

Surgical management may be necessary if severe obstructive symptoms are present, but infiltrative lesions tend to recur.

AMYLOIDOSIS AND THE HEART

Cardiac involvement in amyloidosis is currently the primary determinant of prognosis.3 It is more often seen in the AL, senile, and hereditary forms. It usually manifests as diastolic heart failure, but angina, orthostatic hypotension, dysrhythmias, and syncope can also occur. Systolic dysfunction is typically a late finding in the course of the disease.

Although an electrocardiographic pattern of low voltage in the precordial and limb leads has been classically associated with cardiac amyloidosis, only 30% of patients with the senile and hereditary forms show this feature.4 Left ventricular hypertrophy on electrocardiography is thought to be uncommon, but it has been reported in 16% of patients with AL amyloidosis and biopsy-proven cardiac involvement.5 Cardiac troponin levels can be elevated, as seen in other infiltrative cardiomyopathies.3 The diagnosis can be established by endomyocardial biopsy or indirectly by cardiac imaging and a positive extracardiac biopsy.

Drug therapy is supportive and mainly involves diuretics, since angiotensin-converting enzyme inhibitors, beta-blockers, and calcium channel blockers may cause hypotension and exacerbate myocardial dysfunction. The specific treatment varies depending on the underlying cause of amyloidosis.

A 66-year-old woman was prompted by her dentist to seek medical attention for an unusually enlarged, smooth-appearing tongue (Figure 1). She also complained of fatigue, dyspnea on exertion, and tingling of her hands.

Figure 1.

Basic laboratory tests showed normocytic anemia and renal insufficiency. Her thyrotropin level was within normal limits. Serum protein electrophoresis showed a monoclonal M-spike, which prompted a bone marrow biopsy that was diagnostic of multiple myeloma.

Transthoracic echocardiography revealed diffuse hypokinesis with a restrictive filling pattern, myocardial thickening, and moderate mitral and tricuspid regurgitation, highly suggestive of an infiltrative cardiomyopathy. Biopsy of the right ventricle confirmed cardiac amyloidosis of the amyloid immunoglobulin light chain (AL) subtype.

The patient underwent chemotherapy, followed by autologous stem-cell transplantation. She achieved successful remission, and her cardiomyopathy was compensated.

AMYLOIDOSIS IS HETEROGENEOUS

Amyloidosis is a heterogeneous syndrome characterized by abnormal folding of proteins that deposit as insoluble fibrils in different tissues, impairing both structure and function. Virchow was the first to describe amyloid (from amylon, Greek for starch) as an abnormal material seen in postmortem examination of the liver. On Congo red staining, the extracellular proteins appear as salmon-red conglomerates, which also show apple-green birefringence under polarized light.

Amyloidosis can be localized but more often represents a systemic process, often associated with a plasma cell dyscrasia such as multiple myeloma.

Modern classification is based on the precursor protein,1 eg:

  • Light chains (AL)
  • Acute-phase protein (AA)
  • Beta-2-microglobulin (Aß2M)
  • Transthyretin (ATTR; occurring in senile systemic amyloidosis)
  • Other proteins (occurring in various forms of hereditary systemic amyloidosis).

AMYLOIDOSIS AND THE TONGUE

Macroglossia is defined as protrusion of the tongue beyond the alveolar ridge of the teeth at rest. When caused by amyloidosis, it is most often associated with the systemic AL variant and is present in 10% to 23% of patients with this subtype.2

On physical examination, tongue enlargement can present with lateral indentations, with a smooth contour or with nodular deposits. Less often, bullous lesions, vesicles, and ulcers can also be seen, particularly on the lips. Infiltration of salivary glands can result in xerostomia. Functional symptoms, such as hypogeusia, dysarthria, dysphagia, dysphonia, and, in advanced cases, upper-airway dysfunction can result from restricted mobility of the tongue and tethering to deeper structures.

Surgical management may be necessary if severe obstructive symptoms are present, but infiltrative lesions tend to recur.

AMYLOIDOSIS AND THE HEART

Cardiac involvement in amyloidosis is currently the primary determinant of prognosis.3 It is more often seen in the AL, senile, and hereditary forms. It usually manifests as diastolic heart failure, but angina, orthostatic hypotension, dysrhythmias, and syncope can also occur. Systolic dysfunction is typically a late finding in the course of the disease.

Although an electrocardiographic pattern of low voltage in the precordial and limb leads has been classically associated with cardiac amyloidosis, only 30% of patients with the senile and hereditary forms show this feature.4 Left ventricular hypertrophy on electrocardiography is thought to be uncommon, but it has been reported in 16% of patients with AL amyloidosis and biopsy-proven cardiac involvement.5 Cardiac troponin levels can be elevated, as seen in other infiltrative cardiomyopathies.3 The diagnosis can be established by endomyocardial biopsy or indirectly by cardiac imaging and a positive extracardiac biopsy.

Drug therapy is supportive and mainly involves diuretics, since angiotensin-converting enzyme inhibitors, beta-blockers, and calcium channel blockers may cause hypotension and exacerbate myocardial dysfunction. The specific treatment varies depending on the underlying cause of amyloidosis.

References
  1. Westermark P, Benson MD, Buxbaum JN, et al. A primer of amyloid nomenclature. Amyloid 2007; 14:179183.
  2. Kyle RA, Gertz MA. Primary systemic amyloidosis: clinical and laboratory features in 474 cases. Semin Hematol 1995; 32:4559.
  3. Kapoor P, Thenappan T, Singh E, Kumar S, Greipp PR. Cardiac amyloidosis: a practical approach to diagnosis and management. Am J Med 2011; 124:10061015.
  4. Ng B, Connors LH, Davidoff R, Skinner M, Falk RH. Senile systemic amyloidosis presenting with heart failure: a comparison with light chain-associated amyloidosis. Arch Intern Med 2005; 165:14251429.
  5. Murtagh B, Hammill SC, Gertz MA, Kyle RA, Tajik AJ, Grogan M. Electrocardiographic findings in primary systemic amyloidosis and biopsy-proven cardiac involvement. Am J Cardiol 2005; 95:535537.
References
  1. Westermark P, Benson MD, Buxbaum JN, et al. A primer of amyloid nomenclature. Amyloid 2007; 14:179183.
  2. Kyle RA, Gertz MA. Primary systemic amyloidosis: clinical and laboratory features in 474 cases. Semin Hematol 1995; 32:4559.
  3. Kapoor P, Thenappan T, Singh E, Kumar S, Greipp PR. Cardiac amyloidosis: a practical approach to diagnosis and management. Am J Med 2011; 124:10061015.
  4. Ng B, Connors LH, Davidoff R, Skinner M, Falk RH. Senile systemic amyloidosis presenting with heart failure: a comparison with light chain-associated amyloidosis. Arch Intern Med 2005; 165:14251429.
  5. Murtagh B, Hammill SC, Gertz MA, Kyle RA, Tajik AJ, Grogan M. Electrocardiographic findings in primary systemic amyloidosis and biopsy-proven cardiac involvement. Am J Cardiol 2005; 95:535537.
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Page Number
696-697
Page Number
696-697
Publications
Publications
Topics
Article Type
Display Headline
A 66-year-old woman with an enlarged tongue
Display Headline
A 66-year-old woman with an enlarged tongue
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Dyspnea after treatment of recurrent urinary tract infection

Article Type
Changed
Mon, 09/25/2017 - 14:40
Display Headline
Dyspnea after treatment of recurrent urinary tract infection

A 71-year-old woman came to the hospital because of generalized weakness, fatigue, and exertional dyspnea.

She had a history of anemia, recurrent urinary tract infections, and hyperactive bladder. She had been taking nitrofurantoin for a urinary tract infection and phenazopyridine for dysuria, and she noticed that her urine was dark-colored.

She was of northern European descent. She was unaware of any family history of blood-related disorders. She had been admitted to the hospital 6 weeks earlier for symptomatic anemia after taking nitrofurantoin for a urinary tract infection. At that time, she received 2 units of packed red blood cells and then was discharged. Follow-up blood work done 2 weeks later—including a glucose-6 phosphate dehydrogenase (G6PD) assay—was normal.

On physical examination, she was pale and weak. Her hemoglobin level was 5.5 g/dL (reference range 14.0–17.5), with normal white blood cell and platelet counts and an elevated reticulocyte count. A comprehensive metabolic panel showed elevated indirect bilirubin and lactate dehydrogenase levels. A direct Coombs test for autoimmune hemolytic anemia was negative, as was a haptoglobin assay to look for intravascular hemolytic anemia. G6PD levels were normal, yet a peripheral blood smear (Figure 1) showed features of G6PD deficiency.

What was the cause of her anemia?

Figure 1. A peripheral blood smear in our patient shows several “bite cells” with one or two bites (arrows). These are indicators of a Heinz body hemolytic anemia and suggest the possibility of glucose-6 phosphate dehydrogenase deficiency or an unstable hemoglobin. Heinz bodies, visible only after supravital staining (and not in this smear with conventional Wright-Giemsa stain), consist of clumps of denatured hemoglobin that attach to the inner surface of the red blood cell membrane. Heinz bodies, along with bits of the cell membrane, are pitted from the cells as they pass through the splenic cords, resulting in the formation of bite cells and shortened red cell survival (Wright-Giemsa, ×100).

GLUCOSE-6-PHOSPHATE DEHYDROGENASE DEFICIENCY

G6PD deficiency is an X-linked disorder1 that can present as hemolytic anemia. Symptoms of hemolysis can range from mild to severe on exposure to an inciting agent. Men are more commonly affected than women, and affected women are mostly heterozygous. The severity of hemolysis in heterozygous women depends on inactivation of the unaffected X chromosome in some cells.

When exposed to oxidizing agents, people with G6PD deficiency do not have enough nicotinamide adenine dinucleotide phosphate to protect red blood cells.2 This leads to oxidative denaturation of hemoglobin, formation of methemoglobin, and denaturation of globulin. These products are insoluble; they collect in red blood cells and are called Heinz bodies.3 When red blood cells containing Heinz bodies pass through the liver and spleen, the insoluble masses are taken up by macrophages, causing hemolysis and the formation of “bite cells”4 (so named because macrophages “bite” the Heinz bodies out of the red blood cells).

Patients with G6PD deficiency have all the clinical features of hemolytic anemia. On laboratory testing, the Coombs test is negative, the G6PD level is low, and the peripheral smear shows bite cells. The G6PD level is falsely normal or elevated during acute hemolysis because red blood cells deficient in G6PD are removed from circulation and replaced by young red blood cells. The G6PD level is also elevated after blood transfusion. Thus, the G6PD level should be tested 3 months after an acute event.

Hemolysis in G6PD is usually intermittent and self-limited. No treatment is needed except for avoidance of triggers and transfusion for symptomatic anemia. Of note, triggers include some of the drugs commonly used for urinary tract infections (sulfa drugs, nitrofurantoin, phenazopyridine) and antimalarials. Fava beans are also known to cause hemolytic crisis. A complete list of things to avoid can be found at www.g6pd.org/en/G6PDDeficiency/SafeUnsafe/DaEvitare_ISS-it.

There is no commercially available genetic testing kit for G6PD deficiency. Mutation analysis and G6PD gene sequencing are possible but are neither routinely done nor widely available.

BACK TO OUR PATIENT

Our patient’s hemolytic anemia was most likely drug-induced, secondary to a relative deficiency of G6PD. She had been taking nitrofurantoin and phenazopyridine; both of these are oxidizing agents and are known to cause acute hemolytic anemia in people with G6PD deficiency. The G6PD level can be normal after a recent blood transfusion and, as in our patient, during an acute episode of hemolysis.

Because of the strong suspicion of G6PD deficiency, both drugs were stopped when the patient was discharged from the hospital. She did not take either drug for 3 months. Her G6PD level was then retested and was found to be low, confirming the diagnosis. The patient was then advised not to take those drugs again. Since then, her hemoglobin level has remained stable and she has not needed any more blood transfusions.

References
  1. Mason PJ, Bautista JM, Gilsanz F. G6PD deficiency: the genotype-phenotype association. Blood Rev 2007; 21:267283.
  2. Arese P, De Flora A. Pathophysiology of hemolysis in glucose-6-phosphate dehydrogenase deficiency. Semin Hematol 1990; 27:140.
  3. Jacob HS. Mechanisms of Heinz body formation and attachment to red cell membrane. Semin Hematol 1970; 7:341354.
  4. Rifkind RA. Heinz body anemia: an ultrastructural study. II. Red cell sequestration and destruction. Blood 1965; 26:433448.
Article PDF
Author and Disclosure Information

Krishna B. Ghimire, MD
Assistant Professor, Des Moines University, College of Osteopathic Medicine, Mercy Medical Center–North Iowa, Mason City

Barsha Nepal, MBBS
Mason City, IA

Address: Krishna B. Ghimire, MD, College of Osteopathic Medicine, Mercy Medical Center–North Iowa, 1000 4th Street SW, Mason City, IA 50401; e-mail: [email protected]

Issue
Cleveland Clinic Journal of Medicine - 80(11)
Publications
Topics
Page Number
690-691
Sections
Author and Disclosure Information

Krishna B. Ghimire, MD
Assistant Professor, Des Moines University, College of Osteopathic Medicine, Mercy Medical Center–North Iowa, Mason City

Barsha Nepal, MBBS
Mason City, IA

Address: Krishna B. Ghimire, MD, College of Osteopathic Medicine, Mercy Medical Center–North Iowa, 1000 4th Street SW, Mason City, IA 50401; e-mail: [email protected]

Author and Disclosure Information

Krishna B. Ghimire, MD
Assistant Professor, Des Moines University, College of Osteopathic Medicine, Mercy Medical Center–North Iowa, Mason City

Barsha Nepal, MBBS
Mason City, IA

Address: Krishna B. Ghimire, MD, College of Osteopathic Medicine, Mercy Medical Center–North Iowa, 1000 4th Street SW, Mason City, IA 50401; e-mail: [email protected]

Article PDF
Article PDF

A 71-year-old woman came to the hospital because of generalized weakness, fatigue, and exertional dyspnea.

She had a history of anemia, recurrent urinary tract infections, and hyperactive bladder. She had been taking nitrofurantoin for a urinary tract infection and phenazopyridine for dysuria, and she noticed that her urine was dark-colored.

She was of northern European descent. She was unaware of any family history of blood-related disorders. She had been admitted to the hospital 6 weeks earlier for symptomatic anemia after taking nitrofurantoin for a urinary tract infection. At that time, she received 2 units of packed red blood cells and then was discharged. Follow-up blood work done 2 weeks later—including a glucose-6 phosphate dehydrogenase (G6PD) assay—was normal.

On physical examination, she was pale and weak. Her hemoglobin level was 5.5 g/dL (reference range 14.0–17.5), with normal white blood cell and platelet counts and an elevated reticulocyte count. A comprehensive metabolic panel showed elevated indirect bilirubin and lactate dehydrogenase levels. A direct Coombs test for autoimmune hemolytic anemia was negative, as was a haptoglobin assay to look for intravascular hemolytic anemia. G6PD levels were normal, yet a peripheral blood smear (Figure 1) showed features of G6PD deficiency.

What was the cause of her anemia?

Figure 1. A peripheral blood smear in our patient shows several “bite cells” with one or two bites (arrows). These are indicators of a Heinz body hemolytic anemia and suggest the possibility of glucose-6 phosphate dehydrogenase deficiency or an unstable hemoglobin. Heinz bodies, visible only after supravital staining (and not in this smear with conventional Wright-Giemsa stain), consist of clumps of denatured hemoglobin that attach to the inner surface of the red blood cell membrane. Heinz bodies, along with bits of the cell membrane, are pitted from the cells as they pass through the splenic cords, resulting in the formation of bite cells and shortened red cell survival (Wright-Giemsa, ×100).

GLUCOSE-6-PHOSPHATE DEHYDROGENASE DEFICIENCY

G6PD deficiency is an X-linked disorder1 that can present as hemolytic anemia. Symptoms of hemolysis can range from mild to severe on exposure to an inciting agent. Men are more commonly affected than women, and affected women are mostly heterozygous. The severity of hemolysis in heterozygous women depends on inactivation of the unaffected X chromosome in some cells.

When exposed to oxidizing agents, people with G6PD deficiency do not have enough nicotinamide adenine dinucleotide phosphate to protect red blood cells.2 This leads to oxidative denaturation of hemoglobin, formation of methemoglobin, and denaturation of globulin. These products are insoluble; they collect in red blood cells and are called Heinz bodies.3 When red blood cells containing Heinz bodies pass through the liver and spleen, the insoluble masses are taken up by macrophages, causing hemolysis and the formation of “bite cells”4 (so named because macrophages “bite” the Heinz bodies out of the red blood cells).

Patients with G6PD deficiency have all the clinical features of hemolytic anemia. On laboratory testing, the Coombs test is negative, the G6PD level is low, and the peripheral smear shows bite cells. The G6PD level is falsely normal or elevated during acute hemolysis because red blood cells deficient in G6PD are removed from circulation and replaced by young red blood cells. The G6PD level is also elevated after blood transfusion. Thus, the G6PD level should be tested 3 months after an acute event.

Hemolysis in G6PD is usually intermittent and self-limited. No treatment is needed except for avoidance of triggers and transfusion for symptomatic anemia. Of note, triggers include some of the drugs commonly used for urinary tract infections (sulfa drugs, nitrofurantoin, phenazopyridine) and antimalarials. Fava beans are also known to cause hemolytic crisis. A complete list of things to avoid can be found at www.g6pd.org/en/G6PDDeficiency/SafeUnsafe/DaEvitare_ISS-it.

There is no commercially available genetic testing kit for G6PD deficiency. Mutation analysis and G6PD gene sequencing are possible but are neither routinely done nor widely available.

BACK TO OUR PATIENT

Our patient’s hemolytic anemia was most likely drug-induced, secondary to a relative deficiency of G6PD. She had been taking nitrofurantoin and phenazopyridine; both of these are oxidizing agents and are known to cause acute hemolytic anemia in people with G6PD deficiency. The G6PD level can be normal after a recent blood transfusion and, as in our patient, during an acute episode of hemolysis.

Because of the strong suspicion of G6PD deficiency, both drugs were stopped when the patient was discharged from the hospital. She did not take either drug for 3 months. Her G6PD level was then retested and was found to be low, confirming the diagnosis. The patient was then advised not to take those drugs again. Since then, her hemoglobin level has remained stable and she has not needed any more blood transfusions.

A 71-year-old woman came to the hospital because of generalized weakness, fatigue, and exertional dyspnea.

She had a history of anemia, recurrent urinary tract infections, and hyperactive bladder. She had been taking nitrofurantoin for a urinary tract infection and phenazopyridine for dysuria, and she noticed that her urine was dark-colored.

She was of northern European descent. She was unaware of any family history of blood-related disorders. She had been admitted to the hospital 6 weeks earlier for symptomatic anemia after taking nitrofurantoin for a urinary tract infection. At that time, she received 2 units of packed red blood cells and then was discharged. Follow-up blood work done 2 weeks later—including a glucose-6 phosphate dehydrogenase (G6PD) assay—was normal.

On physical examination, she was pale and weak. Her hemoglobin level was 5.5 g/dL (reference range 14.0–17.5), with normal white blood cell and platelet counts and an elevated reticulocyte count. A comprehensive metabolic panel showed elevated indirect bilirubin and lactate dehydrogenase levels. A direct Coombs test for autoimmune hemolytic anemia was negative, as was a haptoglobin assay to look for intravascular hemolytic anemia. G6PD levels were normal, yet a peripheral blood smear (Figure 1) showed features of G6PD deficiency.

What was the cause of her anemia?

Figure 1. A peripheral blood smear in our patient shows several “bite cells” with one or two bites (arrows). These are indicators of a Heinz body hemolytic anemia and suggest the possibility of glucose-6 phosphate dehydrogenase deficiency or an unstable hemoglobin. Heinz bodies, visible only after supravital staining (and not in this smear with conventional Wright-Giemsa stain), consist of clumps of denatured hemoglobin that attach to the inner surface of the red blood cell membrane. Heinz bodies, along with bits of the cell membrane, are pitted from the cells as they pass through the splenic cords, resulting in the formation of bite cells and shortened red cell survival (Wright-Giemsa, ×100).

GLUCOSE-6-PHOSPHATE DEHYDROGENASE DEFICIENCY

G6PD deficiency is an X-linked disorder1 that can present as hemolytic anemia. Symptoms of hemolysis can range from mild to severe on exposure to an inciting agent. Men are more commonly affected than women, and affected women are mostly heterozygous. The severity of hemolysis in heterozygous women depends on inactivation of the unaffected X chromosome in some cells.

When exposed to oxidizing agents, people with G6PD deficiency do not have enough nicotinamide adenine dinucleotide phosphate to protect red blood cells.2 This leads to oxidative denaturation of hemoglobin, formation of methemoglobin, and denaturation of globulin. These products are insoluble; they collect in red blood cells and are called Heinz bodies.3 When red blood cells containing Heinz bodies pass through the liver and spleen, the insoluble masses are taken up by macrophages, causing hemolysis and the formation of “bite cells”4 (so named because macrophages “bite” the Heinz bodies out of the red blood cells).

Patients with G6PD deficiency have all the clinical features of hemolytic anemia. On laboratory testing, the Coombs test is negative, the G6PD level is low, and the peripheral smear shows bite cells. The G6PD level is falsely normal or elevated during acute hemolysis because red blood cells deficient in G6PD are removed from circulation and replaced by young red blood cells. The G6PD level is also elevated after blood transfusion. Thus, the G6PD level should be tested 3 months after an acute event.

Hemolysis in G6PD is usually intermittent and self-limited. No treatment is needed except for avoidance of triggers and transfusion for symptomatic anemia. Of note, triggers include some of the drugs commonly used for urinary tract infections (sulfa drugs, nitrofurantoin, phenazopyridine) and antimalarials. Fava beans are also known to cause hemolytic crisis. A complete list of things to avoid can be found at www.g6pd.org/en/G6PDDeficiency/SafeUnsafe/DaEvitare_ISS-it.

There is no commercially available genetic testing kit for G6PD deficiency. Mutation analysis and G6PD gene sequencing are possible but are neither routinely done nor widely available.

BACK TO OUR PATIENT

Our patient’s hemolytic anemia was most likely drug-induced, secondary to a relative deficiency of G6PD. She had been taking nitrofurantoin and phenazopyridine; both of these are oxidizing agents and are known to cause acute hemolytic anemia in people with G6PD deficiency. The G6PD level can be normal after a recent blood transfusion and, as in our patient, during an acute episode of hemolysis.

Because of the strong suspicion of G6PD deficiency, both drugs were stopped when the patient was discharged from the hospital. She did not take either drug for 3 months. Her G6PD level was then retested and was found to be low, confirming the diagnosis. The patient was then advised not to take those drugs again. Since then, her hemoglobin level has remained stable and she has not needed any more blood transfusions.

References
  1. Mason PJ, Bautista JM, Gilsanz F. G6PD deficiency: the genotype-phenotype association. Blood Rev 2007; 21:267283.
  2. Arese P, De Flora A. Pathophysiology of hemolysis in glucose-6-phosphate dehydrogenase deficiency. Semin Hematol 1990; 27:140.
  3. Jacob HS. Mechanisms of Heinz body formation and attachment to red cell membrane. Semin Hematol 1970; 7:341354.
  4. Rifkind RA. Heinz body anemia: an ultrastructural study. II. Red cell sequestration and destruction. Blood 1965; 26:433448.
References
  1. Mason PJ, Bautista JM, Gilsanz F. G6PD deficiency: the genotype-phenotype association. Blood Rev 2007; 21:267283.
  2. Arese P, De Flora A. Pathophysiology of hemolysis in glucose-6-phosphate dehydrogenase deficiency. Semin Hematol 1990; 27:140.
  3. Jacob HS. Mechanisms of Heinz body formation and attachment to red cell membrane. Semin Hematol 1970; 7:341354.
  4. Rifkind RA. Heinz body anemia: an ultrastructural study. II. Red cell sequestration and destruction. Blood 1965; 26:433448.
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Page Number
690-691
Page Number
690-691
Publications
Publications
Topics
Article Type
Display Headline
Dyspnea after treatment of recurrent urinary tract infection
Display Headline
Dyspnea after treatment of recurrent urinary tract infection
Sections
Disallow All Ads
Alternative CME
Article PDF Media

Canagliflozin: Improving diabetes by making urine sweet

Article Type
Changed
Tue, 05/03/2022 - 15:52
Display Headline
Canagliflozin: Improving diabetes by making urine sweet

Glycosuria used to be a sign of uncontrolled diabetes and was something to be corrected, not a therapeutic mechanism. But now we have a new class of drugs that lower plasma glucose levels by increasing the renal excretion of glucose.

Here, we will review canagliflozin, the first in a new class of drugs for type 2 diabetes: how it works, who is a candidate for it, and what to watch out for.

THE NEED FOR NEW DIABETES DRUGS

Diabetes mellitus affects more than 25.8 million people in the United States—8.3% of the population—and this staggering number is rising.1 Among US residents age 65 and older, more than 10.9 million (26.9%) have diabetes.1 People with uncontrolled diabetes are at risk of microvascular complications such as retinopathy, nephropathy, and neuropathy, as well as cardiovascular disease. Diabetes is the leading cause of blindness, chronic kidney disease, and nontraumatic lower-limb amputation in the United States.1

Type 2 diabetes accounts for more than 90% of cases of diabetes in the United States, Europe, and Canada.2 It is characterized by insulin resistance, decreased beta-cell function, and progressive beta-cell decline.3

Current American Diabetes Association guidelines for the treatment of diabetes recommend a hemoglobin A1c target of less than 7.0%.4 Initial management includes lifestyle modifications such as changes in diet and an increase in exercise, as well as consideration of metformin treatment at the same time. If glucose levels remain uncontrolled despite these efforts, other drugs should be added.

A number of oral and injectable antihyperglycemic drugs are available to help achieve this goal, though none is without risk of adverse effects. Those available up to now include metformin, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, thiazolidinediones, gliptins, glucagon-like peptide-1 agonists, amylin analogues, colesevelam, dopamine agonists, and insulin.5 Most of the available antihyperglycemics target the liver, pancreas, gut, and muscle to improve insulin sensitivity, reduce insulin resistance, or stimulate insulin secretion.

Despite the abundance of agents, type 2 diabetes remains uncontrolled in many patients. Only 57.1% of participants with previously diagnosed diabetes in the 2003–2006 National Health and Nutrition Examination Survey were at the hemoglobin A1c goal of less than 7.0%.6 Possible reasons for failure include adverse effects such as hypoglycemia, weight gain, and gastrointestinal symptoms resulting in discontinued use, nonadherence to the prescribed regimen, and failure to increase the dosage or to add additional agents, including insulin, to optimize glycemic control as beta-cell function declines over time.

HOW THE KIDNEYS HANDLE GLUCOSE

In the kidney, glucose is filtered in the glomerulus and then is reabsorbed in the proximal tubule. Normally, the filtered glucose is all reabsorbed unless the glucose load exceeds the kidney’s absorptive capacity. Membrane proteins called sodium-glucose cotransporters reabsorb glucose at the proximal tubule and return it into the peripheral circulation. Glucose enters the tubular epithelial cell with sodium by passive cotransport via the sodiumglucose cotransporters, and then exits on the other side via the glucose transporter GLUT in the basolateral membrane.

Two sodium-glucose transporters that act in the proximal tubule of the kidney have been identified: SGLT1 and SGLT2. SGLT2 reabsorbs most of the glucose in the early segment of the proximal tubule, while SLGT1 reabsorbs the remaining glucose at the distal end.7 SGLT2 is responsible for more than 90% of renal tubular reabsorption of glucose and is found only in the proximal tubule, whereas SGLT1 is found mainly in the gastrointestinal tract.8

Patients with type 2 diabetes have a higher capacity for glucose reabsorption in the proximal tubule as a result of the up-regulation of SGLT2.9

SGLT2 INHIBITORS AND TYPE 2 DIABETES

Drugs that inhibit SGLT2 block reabsorption of glucose in the proximal tubule, lowering the renal threshold for glucose and thereby increasing urinary glucose excretion and lowering the serum glucose level in patients with hyperglycemia. This mechanism of action is insulin-independent.

On March 29, 2013, canagliflozin became the first SGLT2 inhibitor to be approved in the United States for the treatment of type 2 diabetes.10 However, it is not the first of its class to be introduced.

Dapagliflozin was the first SGLT2 inhibitor approved in Europe and has been available there since November 2012. However, the US Food and Drug Administration withheld its approval in the United States in January 2012 because of concerns of a possible association with cancer, specifically breast and bladder cancers, as well as possible liver injury.10 Canagliflozin does not appear to share this risk.

Several other SGLT2 inhibitors may soon be available. Empagliflozin is in phase III trials, and the manufacturer has filed for approval in the United States. Ipragliflozin is awaiting approval in Japan.

 

 

CANAGLIFLOZIN: PHARMACOKINETICS AND THERAPEUTIC EFFICACY

Canagliflozin reaches its peak plasma concentration within 1 to 2 hours of oral administration.11 Its half-life is 10.6 hours with a 100-mg dose and 13.1 hours with a 300-mg dose. A steady state is typically achieved in 4 to 5 days.11

Canagliflozin lowers fasting plasma glucose and hemoglobin A1c levels in a dose-dependent manner.10,11 These effects are independent of age, sex, body mass index, and race.12 Postprandial glucose levels are also lowered.

Other potential benefits of canagliflozin include lowering of the systolic blood pressure and, especially important in obese people with type 2 diabetes, weight loss.12 Aside from metformin, which occasionally results in modest weight loss, other oral drugs used in treating type 2 diabetes are weight-neutral or can cause weight gain.

Trials of canagliflozin

Nine phase III trials of canagliflozin have enrolled 10,285 patients, in one of the largest clinical trial programs in type 2 diabetes to date.10 Several of these trials evaluated canagliflozin as monotherapy, whereas others assessed its effect as an add-on therapy in combination with another antihyperglycemic agent such as a sulfonylurea, metformin, pioglitazone, or insulin. There has not yet been a trial directly comparing canagliflozin with metformin.

Four of the placebo-controlled trials evaluated canagliflozin as monotherapy, canagliflozin added to metformin alone, canagliflozin added to metformin plus glimepiride, and canagliflozin added to metformin plus pioglitazone.

When canagliflozin was used as monotherapy, hemoglobin A1c levels at 26 weeks were an absolute 0.91% lower in the canagliflozin 100 mg/day group than in the placebo group, and an absolute 1.16% lower in the canagliflozin 300 mg/day group than in the placebo group (P < .001 for both).12 Patients lost 2.8% of their body weight with canagliflozin 100 mg and 3.3% with canagliflozin 300 mg, compared with 0.6% with placebo. Systolic blood pressure fell by a mean of 3.7 mm Hg with the 100-mg dose and by a mean of 5.4 mm Hg with the 300-mg dose compared with placebo (P < .001 for both dose groups).12

When canagliflozin was added to metformin, with glimepiride as the comparator drug, there was a 5.2% weight reduction with the 100-mg dose, a 5.7% reduction with 300 mg, and a 1% gain with glimepiride. Hemoglobin A1c fell about equally in the three groups.11

When canagliflozin was added to metformin and a sulfonylurea, with sitagliptin as the comparator third drug, the 300-mg canagliflozin dosage group had a 2.8% weight reduction.11

WHAT ARE THE ADVERSE EFFECTS?

Overall, canagliflozin seems to be well tolerated. The most common adverse effects reported in the clinical trials were genital yeast infections, urinary tract infections, and increased urination.

Genital yeast infections were more common in women than in men, occurring in 10.4% of women who received canagliflozin 100 mg and in 11.4% of women who received 300 mg, compared with only 3.2% in the placebo group.11

Urinary tract infections occurred in 5.9% of the 100-mg group and in 4.3% of the 300-mg group, compared with 4.0% of the placebo group.11

Postural hypotension. Lowering of blood pressure and symptoms of postural hypotension were also reported, and these may be attributed to the drug’s mild osmotic diuretic effect. The risk of adverse effects of volume depletion was dose-dependent; in patients over age 75, they occurred in 4.9% of those taking 100 mg and in 8.7% of those taking 300 mg, compared with 2.6% of those in the placebo or active-comparator groups.11 Therefore, one should exercise particular caution when starting this drug in the elderly or in patients taking diuretics or multiple antihypertensive drugs.

Hypoglycemia. When canagliflozin was used as monotherapy, the incidence of hypoglycemia over 26 weeks was similar to that with placebo, occurring in 3.6% of the 100-mg group, 3.0% of the 300-mg group, and 2.6% of the placebo group.12 Canagliflozin was associated with fewer episodes of hypoglycemia than were sulfonylureas, and the number of episodes was similar to that in patients taking gliptins. There was a higher overall incidence of hypoglycemia when canagliflozin was used in combination with a sulfonylurea or with insulin than when it was used as monotherapy.11

Hyperkalemia. Patients with moderate renal impairment or who are on potassiumsparing drugs or drugs that interfere with the renin-angiotensin-aldosterone system may be at higher risk of hyperkalemia, so close monitoring of potassium is recommended. There was also a dose-dependent increase in serum phosphate and magnesium levels, more notably in patients with moderate renal impairment within the first 3 weeks of starting the drug.11

Patients on canagliflozin who are also taking digoxin, ritonavir, phenytoin, phenobarbital, or rifampin should be closely monitored because of the risk of drug-drug interactions.11 Specifically, there was an increase in mean peak digoxin concentrations when used with canagliflozin 300 mg, and the use of phenytoin, phenobarbital, and ritonavir decreased the efficacy of canagliflozin.

WHAT ARE THE CARDIOVASCULAR RISKS OR LONG-TERM CONCERNS?

Dose-dependent increases in low-density lipoprotein cholesterol (LDL-C) may be seen with canagliflozin. Mean changes from baseline compared with placebo were 4.4 mg/dL (4.5%) with canagliflozin 100 mg and 8.3 mg/dL (8%) with canagliflozin 300 mg.11

There was also an increase in non-high-density lipoprotein cholesterol (non-HDL-C).12 Compared with placebo, mean non-HDL-C levels rose by 2.1 mg/dL (1.5%) with canagliflozin 100 mg and 5.1 mg/dL (3.6%) with 300 mg.11

In the 26-week canagliflozin monotherapy trial, archived blood samples in a small subgroup of patients (n = 349) were measured for apolipoprotein-B, which was found to increase by 1.2% with canagliflozin 100 mg and 3.5% with canagliflozin 300 mg, compared with 0.9% in the placebo group.12

Although small, the increase in LDL-C seen with this drug could be a concern, as diabetic patients are already at higher risk of cardiovascular events. The mechanism of this increase is not yet known, though it may be related to metabolic changes from urinary glucose excretion.12

The Canagliflozin Cardiovascular Assessment Study (CANVAS) is a randomized placebo-controlled trial in more than 4,000 patients with type 2 diabetes who have a history of or are at high risk of cardiovascular events. Currently under way, it is evaluating the occurrence of major adverse cardiovascular events (the primary end point) in patients randomized to receive canagliflozin 100 mg, canagliflozin 300 mg, or placebo once daily for up to 4 years. Secondary end points will be the drug’s effects on fasting plasma insulin and glucose, progression of albuminuria, body weight, blood pressure, HDL-C, LDL-C, bone mineral density, markers of bone turnover, and body composition.10 This trial will run for 9 years, to be completed in 2018.13

The CANVAS investigators have already reported that within the first month of treatment, 13 patients taking canagliflozin suffered a major cardiovascular event, including stroke (one of which was fatal) compared with just one patient taking placebo. These events were not seen after the first month. The hazard ratio for major adverse cardiovascular events within the first 30 days was 6.49, but this dropped to 0.89 after the first 30 days.10

Additional issues that should be addressed in long-term postmarketing studies include possible relationships with cancers and pancreatitis and the safety of the drug in pregnancy and in children with diabetes.10

 

 

WHO IS A CANDIDATE FOR THIS DRUG?

Canagliflozin is approved for use as monotherapy in addition to lifestyle modifications. It is also approved for use with other antihyperglycemic drugs, including metformin.

Obese patients with type 2 diabetes and normal kidney function may have the greatest benefit. Because of canagliflozin’s insulinin-dependent mechanism of action, patients with both early and late type 2 diabetes may benefit from its ability to lower hemoglobin A1c and blood glucose.14

Although it can be used in patients with moderate (but not severe) kidney disease, canagliflozin does not appear to be as effective in these patients, who had higher rates of adverse effects.11 It is not indicated for patients with type 1 diabetes, type 2 diabetes with ketonuria, or end-stage renal disease (estimated glomerular filtration rate < 45 mL/min or receiving dialysis).11 It also is not yet recommended for use in pregnant women or patients under age 18.

The recommended starting dose of canagliflozin is 100 mg once daily, taken with breakfast. This can be increased to 300 mg once daily if tolerated. However, patients with an estimated glomerular filtration rate of 45 to 60 mL/min should not exceed the 100- mg dose. No dose adjustment is required in patients with mild to moderate hepatic impairment. It is not recommended, however, in patients with severe hepatic impairment.11

Comment. Although canagliflozin is approved as monotherapy, metformin remains my choice for first-line oral therapy. Because canagliflozin is more expensive and its long-term affects are still relatively unknown, I prefer to use it as an adjunct, and believe it will be a useful addition, especially in obese patients who are seeking to lose weight.

WHAT IS THE COST OF THIS DRUG?

The suggested price is $10.53 per tablet (AmerisourceBergen), which is comparable to that of other newer drugs for type 2 diabetes.

THE BOTTOM LINE

The availability of canagliflozin as an additional oral antihyperglycemic option may prove helpful in managing patients with type 2 diabetes who experience adverse effects with other antihyperglycemic drugs.

As with any new drug, questions remain about the long-term risks of canagliflozin. However, it seems to be well tolerated, especially in patients with normal kidney function, and poses a low risk of hypoglycemia. The slight increase in LDL-C may prompt more aggressive lipid management. Whether blood pressure-lowering and weight loss will offset this increase in LDL-C is yet to be determined. Ongoing studies will help to further elucidate whether there is an increased risk of cardiovascular events.

Finally, canagliflozin distinguishes itself from other oral diabetes drugs by its added benefit of weight loss, an appealing side effect, especially in the growing population of obese individuals with type 2 diabetes mellitus.

References
  1. Centers for Disease Control and Prevention (CDC). Diabetes data and trends. www.cdc.gov/diabetes/statistics/. Accessed September 6, 2013.
  2. National Diabetes Information Clearinghouse (NDIC), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). National diabetes statistics, 2011. www.diabetes.niddk.nih.gov/dm/pubs/statistics/. Accessed September 6, 2013.
  3. Campbell RK. Fate of the beta-cell in the pathophysiology of type 2 diabetes. J Am Pharm Assoc (2003). 2009; 49(suppl 1):S10S15.
  4. American Diabetes Association. Executive summary: standards of medical care in diabetes—2012. Diabetes Care 2012; 35(suppl 1):S4S10.
  5. Blonde L. Current antihyperglycemic treatment strategies for patients with type 2 diabetes mellitus. Cleve Clin J Med 2009; 76(suppl 5):S4S11.
  6. Cheung BM, Ong KL, Cherny SS, Sham PC, Tso AW, Lam KS. Diabetes prevalence and therapeutic target achievement in the United States, 1999 to 2006. Am J Med 2009; 122:443453.
  7. Vallon V. The proximal tubule in the pathophysiology of the diabetic kidney. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1009R1022.
  8. DeFronzo RA, Davidson JA, Del Prato S. The role of the kidneys in glucose homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab 2012; 14:514.
  9. Pfister M, Whaley JM, Zhang L, List JF. Inhibition of SGLT2: a novel strategy for treatment of type 2 diabetes mellitus. Clin Pharmacol Ther 2011; 89:621625.
  10. Food and Drug Administration (FDA). FDA Briefing Document. NDA 204042. Invokana (canagliflozin) Tablets. www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM334550.pdf. Accessed September 6, 2013.
  11. INVOKANA (canagliflozin) tablets, for oral use. Prescribing Information. Janssen Pharmaceuticals, Inc. www.janssenpharmaceuticalsinc.com/assets/invokana_prescribing_info.pdf. Accessed September 6, 2013.
  12. Stenlöf K, Cefalu WT, Kim KA, et al. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes Metab 2013; 15:372382.
  13. US National Institutes of Health. ClinicalTrials.gov. CANVAS—CA Nagliflozin cardio Vascular Assessment Study. http://clinicaltrials.gov/show/NCT01032629. Accessed September 6, 2013.
  14. Devineni D, Morrow L, Hompesch M, et al. Canagliflozin improves glycaemic control over 28 days in subjects with type 2 diabetes not optimally controlled on insulin. Diabetes Obes Metab 2012; 14:539545.
Article PDF
Author and Disclosure Information

Mary Vouyiouklis, MD
Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic

Address: Mary Vouyiouklis, MD, Department of Endocrinology, Diabetes, and Metabolism, X20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Issue
Cleveland Clinic Journal of Medicine - 80(11)
Publications
Topics
Page Number
683-687
Sections
Author and Disclosure Information

Mary Vouyiouklis, MD
Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic

Address: Mary Vouyiouklis, MD, Department of Endocrinology, Diabetes, and Metabolism, X20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Author and Disclosure Information

Mary Vouyiouklis, MD
Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic

Address: Mary Vouyiouklis, MD, Department of Endocrinology, Diabetes, and Metabolism, X20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail [email protected]

Article PDF
Article PDF
Related Articles

Glycosuria used to be a sign of uncontrolled diabetes and was something to be corrected, not a therapeutic mechanism. But now we have a new class of drugs that lower plasma glucose levels by increasing the renal excretion of glucose.

Here, we will review canagliflozin, the first in a new class of drugs for type 2 diabetes: how it works, who is a candidate for it, and what to watch out for.

THE NEED FOR NEW DIABETES DRUGS

Diabetes mellitus affects more than 25.8 million people in the United States—8.3% of the population—and this staggering number is rising.1 Among US residents age 65 and older, more than 10.9 million (26.9%) have diabetes.1 People with uncontrolled diabetes are at risk of microvascular complications such as retinopathy, nephropathy, and neuropathy, as well as cardiovascular disease. Diabetes is the leading cause of blindness, chronic kidney disease, and nontraumatic lower-limb amputation in the United States.1

Type 2 diabetes accounts for more than 90% of cases of diabetes in the United States, Europe, and Canada.2 It is characterized by insulin resistance, decreased beta-cell function, and progressive beta-cell decline.3

Current American Diabetes Association guidelines for the treatment of diabetes recommend a hemoglobin A1c target of less than 7.0%.4 Initial management includes lifestyle modifications such as changes in diet and an increase in exercise, as well as consideration of metformin treatment at the same time. If glucose levels remain uncontrolled despite these efforts, other drugs should be added.

A number of oral and injectable antihyperglycemic drugs are available to help achieve this goal, though none is without risk of adverse effects. Those available up to now include metformin, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, thiazolidinediones, gliptins, glucagon-like peptide-1 agonists, amylin analogues, colesevelam, dopamine agonists, and insulin.5 Most of the available antihyperglycemics target the liver, pancreas, gut, and muscle to improve insulin sensitivity, reduce insulin resistance, or stimulate insulin secretion.

Despite the abundance of agents, type 2 diabetes remains uncontrolled in many patients. Only 57.1% of participants with previously diagnosed diabetes in the 2003–2006 National Health and Nutrition Examination Survey were at the hemoglobin A1c goal of less than 7.0%.6 Possible reasons for failure include adverse effects such as hypoglycemia, weight gain, and gastrointestinal symptoms resulting in discontinued use, nonadherence to the prescribed regimen, and failure to increase the dosage or to add additional agents, including insulin, to optimize glycemic control as beta-cell function declines over time.

HOW THE KIDNEYS HANDLE GLUCOSE

In the kidney, glucose is filtered in the glomerulus and then is reabsorbed in the proximal tubule. Normally, the filtered glucose is all reabsorbed unless the glucose load exceeds the kidney’s absorptive capacity. Membrane proteins called sodium-glucose cotransporters reabsorb glucose at the proximal tubule and return it into the peripheral circulation. Glucose enters the tubular epithelial cell with sodium by passive cotransport via the sodiumglucose cotransporters, and then exits on the other side via the glucose transporter GLUT in the basolateral membrane.

Two sodium-glucose transporters that act in the proximal tubule of the kidney have been identified: SGLT1 and SGLT2. SGLT2 reabsorbs most of the glucose in the early segment of the proximal tubule, while SLGT1 reabsorbs the remaining glucose at the distal end.7 SGLT2 is responsible for more than 90% of renal tubular reabsorption of glucose and is found only in the proximal tubule, whereas SGLT1 is found mainly in the gastrointestinal tract.8

Patients with type 2 diabetes have a higher capacity for glucose reabsorption in the proximal tubule as a result of the up-regulation of SGLT2.9

SGLT2 INHIBITORS AND TYPE 2 DIABETES

Drugs that inhibit SGLT2 block reabsorption of glucose in the proximal tubule, lowering the renal threshold for glucose and thereby increasing urinary glucose excretion and lowering the serum glucose level in patients with hyperglycemia. This mechanism of action is insulin-independent.

On March 29, 2013, canagliflozin became the first SGLT2 inhibitor to be approved in the United States for the treatment of type 2 diabetes.10 However, it is not the first of its class to be introduced.

Dapagliflozin was the first SGLT2 inhibitor approved in Europe and has been available there since November 2012. However, the US Food and Drug Administration withheld its approval in the United States in January 2012 because of concerns of a possible association with cancer, specifically breast and bladder cancers, as well as possible liver injury.10 Canagliflozin does not appear to share this risk.

Several other SGLT2 inhibitors may soon be available. Empagliflozin is in phase III trials, and the manufacturer has filed for approval in the United States. Ipragliflozin is awaiting approval in Japan.

 

 

CANAGLIFLOZIN: PHARMACOKINETICS AND THERAPEUTIC EFFICACY

Canagliflozin reaches its peak plasma concentration within 1 to 2 hours of oral administration.11 Its half-life is 10.6 hours with a 100-mg dose and 13.1 hours with a 300-mg dose. A steady state is typically achieved in 4 to 5 days.11

Canagliflozin lowers fasting plasma glucose and hemoglobin A1c levels in a dose-dependent manner.10,11 These effects are independent of age, sex, body mass index, and race.12 Postprandial glucose levels are also lowered.

Other potential benefits of canagliflozin include lowering of the systolic blood pressure and, especially important in obese people with type 2 diabetes, weight loss.12 Aside from metformin, which occasionally results in modest weight loss, other oral drugs used in treating type 2 diabetes are weight-neutral or can cause weight gain.

Trials of canagliflozin

Nine phase III trials of canagliflozin have enrolled 10,285 patients, in one of the largest clinical trial programs in type 2 diabetes to date.10 Several of these trials evaluated canagliflozin as monotherapy, whereas others assessed its effect as an add-on therapy in combination with another antihyperglycemic agent such as a sulfonylurea, metformin, pioglitazone, or insulin. There has not yet been a trial directly comparing canagliflozin with metformin.

Four of the placebo-controlled trials evaluated canagliflozin as monotherapy, canagliflozin added to metformin alone, canagliflozin added to metformin plus glimepiride, and canagliflozin added to metformin plus pioglitazone.

When canagliflozin was used as monotherapy, hemoglobin A1c levels at 26 weeks were an absolute 0.91% lower in the canagliflozin 100 mg/day group than in the placebo group, and an absolute 1.16% lower in the canagliflozin 300 mg/day group than in the placebo group (P < .001 for both).12 Patients lost 2.8% of their body weight with canagliflozin 100 mg and 3.3% with canagliflozin 300 mg, compared with 0.6% with placebo. Systolic blood pressure fell by a mean of 3.7 mm Hg with the 100-mg dose and by a mean of 5.4 mm Hg with the 300-mg dose compared with placebo (P < .001 for both dose groups).12

When canagliflozin was added to metformin, with glimepiride as the comparator drug, there was a 5.2% weight reduction with the 100-mg dose, a 5.7% reduction with 300 mg, and a 1% gain with glimepiride. Hemoglobin A1c fell about equally in the three groups.11

When canagliflozin was added to metformin and a sulfonylurea, with sitagliptin as the comparator third drug, the 300-mg canagliflozin dosage group had a 2.8% weight reduction.11

WHAT ARE THE ADVERSE EFFECTS?

Overall, canagliflozin seems to be well tolerated. The most common adverse effects reported in the clinical trials were genital yeast infections, urinary tract infections, and increased urination.

Genital yeast infections were more common in women than in men, occurring in 10.4% of women who received canagliflozin 100 mg and in 11.4% of women who received 300 mg, compared with only 3.2% in the placebo group.11

Urinary tract infections occurred in 5.9% of the 100-mg group and in 4.3% of the 300-mg group, compared with 4.0% of the placebo group.11

Postural hypotension. Lowering of blood pressure and symptoms of postural hypotension were also reported, and these may be attributed to the drug’s mild osmotic diuretic effect. The risk of adverse effects of volume depletion was dose-dependent; in patients over age 75, they occurred in 4.9% of those taking 100 mg and in 8.7% of those taking 300 mg, compared with 2.6% of those in the placebo or active-comparator groups.11 Therefore, one should exercise particular caution when starting this drug in the elderly or in patients taking diuretics or multiple antihypertensive drugs.

Hypoglycemia. When canagliflozin was used as monotherapy, the incidence of hypoglycemia over 26 weeks was similar to that with placebo, occurring in 3.6% of the 100-mg group, 3.0% of the 300-mg group, and 2.6% of the placebo group.12 Canagliflozin was associated with fewer episodes of hypoglycemia than were sulfonylureas, and the number of episodes was similar to that in patients taking gliptins. There was a higher overall incidence of hypoglycemia when canagliflozin was used in combination with a sulfonylurea or with insulin than when it was used as monotherapy.11

Hyperkalemia. Patients with moderate renal impairment or who are on potassiumsparing drugs or drugs that interfere with the renin-angiotensin-aldosterone system may be at higher risk of hyperkalemia, so close monitoring of potassium is recommended. There was also a dose-dependent increase in serum phosphate and magnesium levels, more notably in patients with moderate renal impairment within the first 3 weeks of starting the drug.11

Patients on canagliflozin who are also taking digoxin, ritonavir, phenytoin, phenobarbital, or rifampin should be closely monitored because of the risk of drug-drug interactions.11 Specifically, there was an increase in mean peak digoxin concentrations when used with canagliflozin 300 mg, and the use of phenytoin, phenobarbital, and ritonavir decreased the efficacy of canagliflozin.

WHAT ARE THE CARDIOVASCULAR RISKS OR LONG-TERM CONCERNS?

Dose-dependent increases in low-density lipoprotein cholesterol (LDL-C) may be seen with canagliflozin. Mean changes from baseline compared with placebo were 4.4 mg/dL (4.5%) with canagliflozin 100 mg and 8.3 mg/dL (8%) with canagliflozin 300 mg.11

There was also an increase in non-high-density lipoprotein cholesterol (non-HDL-C).12 Compared with placebo, mean non-HDL-C levels rose by 2.1 mg/dL (1.5%) with canagliflozin 100 mg and 5.1 mg/dL (3.6%) with 300 mg.11

In the 26-week canagliflozin monotherapy trial, archived blood samples in a small subgroup of patients (n = 349) were measured for apolipoprotein-B, which was found to increase by 1.2% with canagliflozin 100 mg and 3.5% with canagliflozin 300 mg, compared with 0.9% in the placebo group.12

Although small, the increase in LDL-C seen with this drug could be a concern, as diabetic patients are already at higher risk of cardiovascular events. The mechanism of this increase is not yet known, though it may be related to metabolic changes from urinary glucose excretion.12

The Canagliflozin Cardiovascular Assessment Study (CANVAS) is a randomized placebo-controlled trial in more than 4,000 patients with type 2 diabetes who have a history of or are at high risk of cardiovascular events. Currently under way, it is evaluating the occurrence of major adverse cardiovascular events (the primary end point) in patients randomized to receive canagliflozin 100 mg, canagliflozin 300 mg, or placebo once daily for up to 4 years. Secondary end points will be the drug’s effects on fasting plasma insulin and glucose, progression of albuminuria, body weight, blood pressure, HDL-C, LDL-C, bone mineral density, markers of bone turnover, and body composition.10 This trial will run for 9 years, to be completed in 2018.13

The CANVAS investigators have already reported that within the first month of treatment, 13 patients taking canagliflozin suffered a major cardiovascular event, including stroke (one of which was fatal) compared with just one patient taking placebo. These events were not seen after the first month. The hazard ratio for major adverse cardiovascular events within the first 30 days was 6.49, but this dropped to 0.89 after the first 30 days.10

Additional issues that should be addressed in long-term postmarketing studies include possible relationships with cancers and pancreatitis and the safety of the drug in pregnancy and in children with diabetes.10

 

 

WHO IS A CANDIDATE FOR THIS DRUG?

Canagliflozin is approved for use as monotherapy in addition to lifestyle modifications. It is also approved for use with other antihyperglycemic drugs, including metformin.

Obese patients with type 2 diabetes and normal kidney function may have the greatest benefit. Because of canagliflozin’s insulinin-dependent mechanism of action, patients with both early and late type 2 diabetes may benefit from its ability to lower hemoglobin A1c and blood glucose.14

Although it can be used in patients with moderate (but not severe) kidney disease, canagliflozin does not appear to be as effective in these patients, who had higher rates of adverse effects.11 It is not indicated for patients with type 1 diabetes, type 2 diabetes with ketonuria, or end-stage renal disease (estimated glomerular filtration rate < 45 mL/min or receiving dialysis).11 It also is not yet recommended for use in pregnant women or patients under age 18.

The recommended starting dose of canagliflozin is 100 mg once daily, taken with breakfast. This can be increased to 300 mg once daily if tolerated. However, patients with an estimated glomerular filtration rate of 45 to 60 mL/min should not exceed the 100- mg dose. No dose adjustment is required in patients with mild to moderate hepatic impairment. It is not recommended, however, in patients with severe hepatic impairment.11

Comment. Although canagliflozin is approved as monotherapy, metformin remains my choice for first-line oral therapy. Because canagliflozin is more expensive and its long-term affects are still relatively unknown, I prefer to use it as an adjunct, and believe it will be a useful addition, especially in obese patients who are seeking to lose weight.

WHAT IS THE COST OF THIS DRUG?

The suggested price is $10.53 per tablet (AmerisourceBergen), which is comparable to that of other newer drugs for type 2 diabetes.

THE BOTTOM LINE

The availability of canagliflozin as an additional oral antihyperglycemic option may prove helpful in managing patients with type 2 diabetes who experience adverse effects with other antihyperglycemic drugs.

As with any new drug, questions remain about the long-term risks of canagliflozin. However, it seems to be well tolerated, especially in patients with normal kidney function, and poses a low risk of hypoglycemia. The slight increase in LDL-C may prompt more aggressive lipid management. Whether blood pressure-lowering and weight loss will offset this increase in LDL-C is yet to be determined. Ongoing studies will help to further elucidate whether there is an increased risk of cardiovascular events.

Finally, canagliflozin distinguishes itself from other oral diabetes drugs by its added benefit of weight loss, an appealing side effect, especially in the growing population of obese individuals with type 2 diabetes mellitus.

Glycosuria used to be a sign of uncontrolled diabetes and was something to be corrected, not a therapeutic mechanism. But now we have a new class of drugs that lower plasma glucose levels by increasing the renal excretion of glucose.

Here, we will review canagliflozin, the first in a new class of drugs for type 2 diabetes: how it works, who is a candidate for it, and what to watch out for.

THE NEED FOR NEW DIABETES DRUGS

Diabetes mellitus affects more than 25.8 million people in the United States—8.3% of the population—and this staggering number is rising.1 Among US residents age 65 and older, more than 10.9 million (26.9%) have diabetes.1 People with uncontrolled diabetes are at risk of microvascular complications such as retinopathy, nephropathy, and neuropathy, as well as cardiovascular disease. Diabetes is the leading cause of blindness, chronic kidney disease, and nontraumatic lower-limb amputation in the United States.1

Type 2 diabetes accounts for more than 90% of cases of diabetes in the United States, Europe, and Canada.2 It is characterized by insulin resistance, decreased beta-cell function, and progressive beta-cell decline.3

Current American Diabetes Association guidelines for the treatment of diabetes recommend a hemoglobin A1c target of less than 7.0%.4 Initial management includes lifestyle modifications such as changes in diet and an increase in exercise, as well as consideration of metformin treatment at the same time. If glucose levels remain uncontrolled despite these efforts, other drugs should be added.

A number of oral and injectable antihyperglycemic drugs are available to help achieve this goal, though none is without risk of adverse effects. Those available up to now include metformin, sulfonylureas, meglitinides, alpha-glucosidase inhibitors, thiazolidinediones, gliptins, glucagon-like peptide-1 agonists, amylin analogues, colesevelam, dopamine agonists, and insulin.5 Most of the available antihyperglycemics target the liver, pancreas, gut, and muscle to improve insulin sensitivity, reduce insulin resistance, or stimulate insulin secretion.

Despite the abundance of agents, type 2 diabetes remains uncontrolled in many patients. Only 57.1% of participants with previously diagnosed diabetes in the 2003–2006 National Health and Nutrition Examination Survey were at the hemoglobin A1c goal of less than 7.0%.6 Possible reasons for failure include adverse effects such as hypoglycemia, weight gain, and gastrointestinal symptoms resulting in discontinued use, nonadherence to the prescribed regimen, and failure to increase the dosage or to add additional agents, including insulin, to optimize glycemic control as beta-cell function declines over time.

HOW THE KIDNEYS HANDLE GLUCOSE

In the kidney, glucose is filtered in the glomerulus and then is reabsorbed in the proximal tubule. Normally, the filtered glucose is all reabsorbed unless the glucose load exceeds the kidney’s absorptive capacity. Membrane proteins called sodium-glucose cotransporters reabsorb glucose at the proximal tubule and return it into the peripheral circulation. Glucose enters the tubular epithelial cell with sodium by passive cotransport via the sodiumglucose cotransporters, and then exits on the other side via the glucose transporter GLUT in the basolateral membrane.

Two sodium-glucose transporters that act in the proximal tubule of the kidney have been identified: SGLT1 and SGLT2. SGLT2 reabsorbs most of the glucose in the early segment of the proximal tubule, while SLGT1 reabsorbs the remaining glucose at the distal end.7 SGLT2 is responsible for more than 90% of renal tubular reabsorption of glucose and is found only in the proximal tubule, whereas SGLT1 is found mainly in the gastrointestinal tract.8

Patients with type 2 diabetes have a higher capacity for glucose reabsorption in the proximal tubule as a result of the up-regulation of SGLT2.9

SGLT2 INHIBITORS AND TYPE 2 DIABETES

Drugs that inhibit SGLT2 block reabsorption of glucose in the proximal tubule, lowering the renal threshold for glucose and thereby increasing urinary glucose excretion and lowering the serum glucose level in patients with hyperglycemia. This mechanism of action is insulin-independent.

On March 29, 2013, canagliflozin became the first SGLT2 inhibitor to be approved in the United States for the treatment of type 2 diabetes.10 However, it is not the first of its class to be introduced.

Dapagliflozin was the first SGLT2 inhibitor approved in Europe and has been available there since November 2012. However, the US Food and Drug Administration withheld its approval in the United States in January 2012 because of concerns of a possible association with cancer, specifically breast and bladder cancers, as well as possible liver injury.10 Canagliflozin does not appear to share this risk.

Several other SGLT2 inhibitors may soon be available. Empagliflozin is in phase III trials, and the manufacturer has filed for approval in the United States. Ipragliflozin is awaiting approval in Japan.

 

 

CANAGLIFLOZIN: PHARMACOKINETICS AND THERAPEUTIC EFFICACY

Canagliflozin reaches its peak plasma concentration within 1 to 2 hours of oral administration.11 Its half-life is 10.6 hours with a 100-mg dose and 13.1 hours with a 300-mg dose. A steady state is typically achieved in 4 to 5 days.11

Canagliflozin lowers fasting plasma glucose and hemoglobin A1c levels in a dose-dependent manner.10,11 These effects are independent of age, sex, body mass index, and race.12 Postprandial glucose levels are also lowered.

Other potential benefits of canagliflozin include lowering of the systolic blood pressure and, especially important in obese people with type 2 diabetes, weight loss.12 Aside from metformin, which occasionally results in modest weight loss, other oral drugs used in treating type 2 diabetes are weight-neutral or can cause weight gain.

Trials of canagliflozin

Nine phase III trials of canagliflozin have enrolled 10,285 patients, in one of the largest clinical trial programs in type 2 diabetes to date.10 Several of these trials evaluated canagliflozin as monotherapy, whereas others assessed its effect as an add-on therapy in combination with another antihyperglycemic agent such as a sulfonylurea, metformin, pioglitazone, or insulin. There has not yet been a trial directly comparing canagliflozin with metformin.

Four of the placebo-controlled trials evaluated canagliflozin as monotherapy, canagliflozin added to metformin alone, canagliflozin added to metformin plus glimepiride, and canagliflozin added to metformin plus pioglitazone.

When canagliflozin was used as monotherapy, hemoglobin A1c levels at 26 weeks were an absolute 0.91% lower in the canagliflozin 100 mg/day group than in the placebo group, and an absolute 1.16% lower in the canagliflozin 300 mg/day group than in the placebo group (P < .001 for both).12 Patients lost 2.8% of their body weight with canagliflozin 100 mg and 3.3% with canagliflozin 300 mg, compared with 0.6% with placebo. Systolic blood pressure fell by a mean of 3.7 mm Hg with the 100-mg dose and by a mean of 5.4 mm Hg with the 300-mg dose compared with placebo (P < .001 for both dose groups).12

When canagliflozin was added to metformin, with glimepiride as the comparator drug, there was a 5.2% weight reduction with the 100-mg dose, a 5.7% reduction with 300 mg, and a 1% gain with glimepiride. Hemoglobin A1c fell about equally in the three groups.11

When canagliflozin was added to metformin and a sulfonylurea, with sitagliptin as the comparator third drug, the 300-mg canagliflozin dosage group had a 2.8% weight reduction.11

WHAT ARE THE ADVERSE EFFECTS?

Overall, canagliflozin seems to be well tolerated. The most common adverse effects reported in the clinical trials were genital yeast infections, urinary tract infections, and increased urination.

Genital yeast infections were more common in women than in men, occurring in 10.4% of women who received canagliflozin 100 mg and in 11.4% of women who received 300 mg, compared with only 3.2% in the placebo group.11

Urinary tract infections occurred in 5.9% of the 100-mg group and in 4.3% of the 300-mg group, compared with 4.0% of the placebo group.11

Postural hypotension. Lowering of blood pressure and symptoms of postural hypotension were also reported, and these may be attributed to the drug’s mild osmotic diuretic effect. The risk of adverse effects of volume depletion was dose-dependent; in patients over age 75, they occurred in 4.9% of those taking 100 mg and in 8.7% of those taking 300 mg, compared with 2.6% of those in the placebo or active-comparator groups.11 Therefore, one should exercise particular caution when starting this drug in the elderly or in patients taking diuretics or multiple antihypertensive drugs.

Hypoglycemia. When canagliflozin was used as monotherapy, the incidence of hypoglycemia over 26 weeks was similar to that with placebo, occurring in 3.6% of the 100-mg group, 3.0% of the 300-mg group, and 2.6% of the placebo group.12 Canagliflozin was associated with fewer episodes of hypoglycemia than were sulfonylureas, and the number of episodes was similar to that in patients taking gliptins. There was a higher overall incidence of hypoglycemia when canagliflozin was used in combination with a sulfonylurea or with insulin than when it was used as monotherapy.11

Hyperkalemia. Patients with moderate renal impairment or who are on potassiumsparing drugs or drugs that interfere with the renin-angiotensin-aldosterone system may be at higher risk of hyperkalemia, so close monitoring of potassium is recommended. There was also a dose-dependent increase in serum phosphate and magnesium levels, more notably in patients with moderate renal impairment within the first 3 weeks of starting the drug.11

Patients on canagliflozin who are also taking digoxin, ritonavir, phenytoin, phenobarbital, or rifampin should be closely monitored because of the risk of drug-drug interactions.11 Specifically, there was an increase in mean peak digoxin concentrations when used with canagliflozin 300 mg, and the use of phenytoin, phenobarbital, and ritonavir decreased the efficacy of canagliflozin.

WHAT ARE THE CARDIOVASCULAR RISKS OR LONG-TERM CONCERNS?

Dose-dependent increases in low-density lipoprotein cholesterol (LDL-C) may be seen with canagliflozin. Mean changes from baseline compared with placebo were 4.4 mg/dL (4.5%) with canagliflozin 100 mg and 8.3 mg/dL (8%) with canagliflozin 300 mg.11

There was also an increase in non-high-density lipoprotein cholesterol (non-HDL-C).12 Compared with placebo, mean non-HDL-C levels rose by 2.1 mg/dL (1.5%) with canagliflozin 100 mg and 5.1 mg/dL (3.6%) with 300 mg.11

In the 26-week canagliflozin monotherapy trial, archived blood samples in a small subgroup of patients (n = 349) were measured for apolipoprotein-B, which was found to increase by 1.2% with canagliflozin 100 mg and 3.5% with canagliflozin 300 mg, compared with 0.9% in the placebo group.12

Although small, the increase in LDL-C seen with this drug could be a concern, as diabetic patients are already at higher risk of cardiovascular events. The mechanism of this increase is not yet known, though it may be related to metabolic changes from urinary glucose excretion.12

The Canagliflozin Cardiovascular Assessment Study (CANVAS) is a randomized placebo-controlled trial in more than 4,000 patients with type 2 diabetes who have a history of or are at high risk of cardiovascular events. Currently under way, it is evaluating the occurrence of major adverse cardiovascular events (the primary end point) in patients randomized to receive canagliflozin 100 mg, canagliflozin 300 mg, or placebo once daily for up to 4 years. Secondary end points will be the drug’s effects on fasting plasma insulin and glucose, progression of albuminuria, body weight, blood pressure, HDL-C, LDL-C, bone mineral density, markers of bone turnover, and body composition.10 This trial will run for 9 years, to be completed in 2018.13

The CANVAS investigators have already reported that within the first month of treatment, 13 patients taking canagliflozin suffered a major cardiovascular event, including stroke (one of which was fatal) compared with just one patient taking placebo. These events were not seen after the first month. The hazard ratio for major adverse cardiovascular events within the first 30 days was 6.49, but this dropped to 0.89 after the first 30 days.10

Additional issues that should be addressed in long-term postmarketing studies include possible relationships with cancers and pancreatitis and the safety of the drug in pregnancy and in children with diabetes.10

 

 

WHO IS A CANDIDATE FOR THIS DRUG?

Canagliflozin is approved for use as monotherapy in addition to lifestyle modifications. It is also approved for use with other antihyperglycemic drugs, including metformin.

Obese patients with type 2 diabetes and normal kidney function may have the greatest benefit. Because of canagliflozin’s insulinin-dependent mechanism of action, patients with both early and late type 2 diabetes may benefit from its ability to lower hemoglobin A1c and blood glucose.14

Although it can be used in patients with moderate (but not severe) kidney disease, canagliflozin does not appear to be as effective in these patients, who had higher rates of adverse effects.11 It is not indicated for patients with type 1 diabetes, type 2 diabetes with ketonuria, or end-stage renal disease (estimated glomerular filtration rate < 45 mL/min or receiving dialysis).11 It also is not yet recommended for use in pregnant women or patients under age 18.

The recommended starting dose of canagliflozin is 100 mg once daily, taken with breakfast. This can be increased to 300 mg once daily if tolerated. However, patients with an estimated glomerular filtration rate of 45 to 60 mL/min should not exceed the 100- mg dose. No dose adjustment is required in patients with mild to moderate hepatic impairment. It is not recommended, however, in patients with severe hepatic impairment.11

Comment. Although canagliflozin is approved as monotherapy, metformin remains my choice for first-line oral therapy. Because canagliflozin is more expensive and its long-term affects are still relatively unknown, I prefer to use it as an adjunct, and believe it will be a useful addition, especially in obese patients who are seeking to lose weight.

WHAT IS THE COST OF THIS DRUG?

The suggested price is $10.53 per tablet (AmerisourceBergen), which is comparable to that of other newer drugs for type 2 diabetes.

THE BOTTOM LINE

The availability of canagliflozin as an additional oral antihyperglycemic option may prove helpful in managing patients with type 2 diabetes who experience adverse effects with other antihyperglycemic drugs.

As with any new drug, questions remain about the long-term risks of canagliflozin. However, it seems to be well tolerated, especially in patients with normal kidney function, and poses a low risk of hypoglycemia. The slight increase in LDL-C may prompt more aggressive lipid management. Whether blood pressure-lowering and weight loss will offset this increase in LDL-C is yet to be determined. Ongoing studies will help to further elucidate whether there is an increased risk of cardiovascular events.

Finally, canagliflozin distinguishes itself from other oral diabetes drugs by its added benefit of weight loss, an appealing side effect, especially in the growing population of obese individuals with type 2 diabetes mellitus.

References
  1. Centers for Disease Control and Prevention (CDC). Diabetes data and trends. www.cdc.gov/diabetes/statistics/. Accessed September 6, 2013.
  2. National Diabetes Information Clearinghouse (NDIC), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). National diabetes statistics, 2011. www.diabetes.niddk.nih.gov/dm/pubs/statistics/. Accessed September 6, 2013.
  3. Campbell RK. Fate of the beta-cell in the pathophysiology of type 2 diabetes. J Am Pharm Assoc (2003). 2009; 49(suppl 1):S10S15.
  4. American Diabetes Association. Executive summary: standards of medical care in diabetes—2012. Diabetes Care 2012; 35(suppl 1):S4S10.
  5. Blonde L. Current antihyperglycemic treatment strategies for patients with type 2 diabetes mellitus. Cleve Clin J Med 2009; 76(suppl 5):S4S11.
  6. Cheung BM, Ong KL, Cherny SS, Sham PC, Tso AW, Lam KS. Diabetes prevalence and therapeutic target achievement in the United States, 1999 to 2006. Am J Med 2009; 122:443453.
  7. Vallon V. The proximal tubule in the pathophysiology of the diabetic kidney. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1009R1022.
  8. DeFronzo RA, Davidson JA, Del Prato S. The role of the kidneys in glucose homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab 2012; 14:514.
  9. Pfister M, Whaley JM, Zhang L, List JF. Inhibition of SGLT2: a novel strategy for treatment of type 2 diabetes mellitus. Clin Pharmacol Ther 2011; 89:621625.
  10. Food and Drug Administration (FDA). FDA Briefing Document. NDA 204042. Invokana (canagliflozin) Tablets. www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM334550.pdf. Accessed September 6, 2013.
  11. INVOKANA (canagliflozin) tablets, for oral use. Prescribing Information. Janssen Pharmaceuticals, Inc. www.janssenpharmaceuticalsinc.com/assets/invokana_prescribing_info.pdf. Accessed September 6, 2013.
  12. Stenlöf K, Cefalu WT, Kim KA, et al. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes Metab 2013; 15:372382.
  13. US National Institutes of Health. ClinicalTrials.gov. CANVAS—CA Nagliflozin cardio Vascular Assessment Study. http://clinicaltrials.gov/show/NCT01032629. Accessed September 6, 2013.
  14. Devineni D, Morrow L, Hompesch M, et al. Canagliflozin improves glycaemic control over 28 days in subjects with type 2 diabetes not optimally controlled on insulin. Diabetes Obes Metab 2012; 14:539545.
References
  1. Centers for Disease Control and Prevention (CDC). Diabetes data and trends. www.cdc.gov/diabetes/statistics/. Accessed September 6, 2013.
  2. National Diabetes Information Clearinghouse (NDIC), National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK). National diabetes statistics, 2011. www.diabetes.niddk.nih.gov/dm/pubs/statistics/. Accessed September 6, 2013.
  3. Campbell RK. Fate of the beta-cell in the pathophysiology of type 2 diabetes. J Am Pharm Assoc (2003). 2009; 49(suppl 1):S10S15.
  4. American Diabetes Association. Executive summary: standards of medical care in diabetes—2012. Diabetes Care 2012; 35(suppl 1):S4S10.
  5. Blonde L. Current antihyperglycemic treatment strategies for patients with type 2 diabetes mellitus. Cleve Clin J Med 2009; 76(suppl 5):S4S11.
  6. Cheung BM, Ong KL, Cherny SS, Sham PC, Tso AW, Lam KS. Diabetes prevalence and therapeutic target achievement in the United States, 1999 to 2006. Am J Med 2009; 122:443453.
  7. Vallon V. The proximal tubule in the pathophysiology of the diabetic kidney. Am J Physiol Regul Integr Comp Physiol 2011; 300:R1009R1022.
  8. DeFronzo RA, Davidson JA, Del Prato S. The role of the kidneys in glucose homeostasis: a new path towards normalizing glycaemia. Diabetes Obes Metab 2012; 14:514.
  9. Pfister M, Whaley JM, Zhang L, List JF. Inhibition of SGLT2: a novel strategy for treatment of type 2 diabetes mellitus. Clin Pharmacol Ther 2011; 89:621625.
  10. Food and Drug Administration (FDA). FDA Briefing Document. NDA 204042. Invokana (canagliflozin) Tablets. www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM334550.pdf. Accessed September 6, 2013.
  11. INVOKANA (canagliflozin) tablets, for oral use. Prescribing Information. Janssen Pharmaceuticals, Inc. www.janssenpharmaceuticalsinc.com/assets/invokana_prescribing_info.pdf. Accessed September 6, 2013.
  12. Stenlöf K, Cefalu WT, Kim KA, et al. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes Metab 2013; 15:372382.
  13. US National Institutes of Health. ClinicalTrials.gov. CANVAS—CA Nagliflozin cardio Vascular Assessment Study. http://clinicaltrials.gov/show/NCT01032629. Accessed September 6, 2013.
  14. Devineni D, Morrow L, Hompesch M, et al. Canagliflozin improves glycaemic control over 28 days in subjects with type 2 diabetes not optimally controlled on insulin. Diabetes Obes Metab 2012; 14:539545.
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Page Number
683-687
Page Number
683-687
Publications
Publications
Topics
Article Type
Display Headline
Canagliflozin: Improving diabetes by making urine sweet
Display Headline
Canagliflozin: Improving diabetes by making urine sweet
Sections
Inside the Article

KEY POINTS

  • Type 2 diabetes is ubiquitous and, despite an abundance of agents, often remains uncontrolled.
  • Canagliflozin and other drugs of its class cause glucose to be spilled in the urine by reducing the amount reabsorbed by the kidney.
  • In clinical trials, canagliflozin lowered hemoglobin A1c levels by approximately 1 absolute percentage point.
  • Beyond the adverse effects to be expected from the mechanism of action of the drug (ie, genital yeast infections, urinary tract infections, and hypotension caused by osmotic diuresis), canagliflozin seems to increase plasma levels of low-density lipoprotein cholesterol. This may be worrisome, as diabetic patients are already at increased risk of cardiovascular disease.
Disallow All Ads
Alternative CME
Article PDF Media

When people with diabetes go to surgery

Article Type
Changed
Tue, 05/03/2022 - 15:52
Display Headline
When people with diabetes go to surgery

Over the past decade, recommendations about the ideal glucose target in hospitalized diabetic patients have fluctuated. The controversy has extended to diabetic patients in various types of intensive care units and to those headed to the operating room. Although proposals exist on how to manage diabetes in the operating room, including intraoperative insulin infusions, outcomes probably depend more on how glucose is managed during the patient’s postoperative stay in the hospital. For patients who are less critically ill and less medically complex, continuous insulin infusions are used infrequently, and insulin is often prescribed by algorithm or, archaically, by some form of “catch-up” sliding scale. Studies indicate that even the fairly loose glucose target of 70 to 180 mg/dL is achieved consistently in only a few patients.1

In view of a number of observations, including the link between hyperglycemia and postoperative wound infections, studies were designed to test the hypothesis that aggressively keeping glucose levels quite low in critically ill and postoperative diabetic patients would be beneficial. Instead, most of these studies found that overly tight glucose control in these settings led to untoward outcomes—and not only as the result of hypoglycemic episodes. Aiming for a modest serum glucose target of 150 to 200 mg/dL can significantly reduce the postoperative death rate, but the beneficial reduction is no greater if the target is less than 150 mg/dL.

With a looser glucose target, pre- and perioperative management of insulin-dependent diabetic patients can be simplified. Dobri and Lansang discuss the key practical principles of managing insulin before the patient goes to the operating suite. They emphasize relevant pearls of insulin physiology and discuss several scenarios we often encounter.

In fact, the principles they review are equally useful to remember when we admit diabetic patients to the hospital with orders to keep them “npo” while planning and awaiting tests or other procedures. A key take-home point is that severely insulinopenic patients require some exogenous basal insulin, even when not eating.

References
  1. Lopes R, Albrecht A, Williams J, et al. Postoperative glucose control following coronary artery bypass graft surgery: predictors and clinical outcomes. J Am Coll Cardiol 2013; 61:e1601.
Article PDF
Author and Disclosure Information
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Publications
Topics
Page Number
682
Sections
Author and Disclosure Information
Author and Disclosure Information
Article PDF
Article PDF

Over the past decade, recommendations about the ideal glucose target in hospitalized diabetic patients have fluctuated. The controversy has extended to diabetic patients in various types of intensive care units and to those headed to the operating room. Although proposals exist on how to manage diabetes in the operating room, including intraoperative insulin infusions, outcomes probably depend more on how glucose is managed during the patient’s postoperative stay in the hospital. For patients who are less critically ill and less medically complex, continuous insulin infusions are used infrequently, and insulin is often prescribed by algorithm or, archaically, by some form of “catch-up” sliding scale. Studies indicate that even the fairly loose glucose target of 70 to 180 mg/dL is achieved consistently in only a few patients.1

In view of a number of observations, including the link between hyperglycemia and postoperative wound infections, studies were designed to test the hypothesis that aggressively keeping glucose levels quite low in critically ill and postoperative diabetic patients would be beneficial. Instead, most of these studies found that overly tight glucose control in these settings led to untoward outcomes—and not only as the result of hypoglycemic episodes. Aiming for a modest serum glucose target of 150 to 200 mg/dL can significantly reduce the postoperative death rate, but the beneficial reduction is no greater if the target is less than 150 mg/dL.

With a looser glucose target, pre- and perioperative management of insulin-dependent diabetic patients can be simplified. Dobri and Lansang discuss the key practical principles of managing insulin before the patient goes to the operating suite. They emphasize relevant pearls of insulin physiology and discuss several scenarios we often encounter.

In fact, the principles they review are equally useful to remember when we admit diabetic patients to the hospital with orders to keep them “npo” while planning and awaiting tests or other procedures. A key take-home point is that severely insulinopenic patients require some exogenous basal insulin, even when not eating.

Over the past decade, recommendations about the ideal glucose target in hospitalized diabetic patients have fluctuated. The controversy has extended to diabetic patients in various types of intensive care units and to those headed to the operating room. Although proposals exist on how to manage diabetes in the operating room, including intraoperative insulin infusions, outcomes probably depend more on how glucose is managed during the patient’s postoperative stay in the hospital. For patients who are less critically ill and less medically complex, continuous insulin infusions are used infrequently, and insulin is often prescribed by algorithm or, archaically, by some form of “catch-up” sliding scale. Studies indicate that even the fairly loose glucose target of 70 to 180 mg/dL is achieved consistently in only a few patients.1

In view of a number of observations, including the link between hyperglycemia and postoperative wound infections, studies were designed to test the hypothesis that aggressively keeping glucose levels quite low in critically ill and postoperative diabetic patients would be beneficial. Instead, most of these studies found that overly tight glucose control in these settings led to untoward outcomes—and not only as the result of hypoglycemic episodes. Aiming for a modest serum glucose target of 150 to 200 mg/dL can significantly reduce the postoperative death rate, but the beneficial reduction is no greater if the target is less than 150 mg/dL.

With a looser glucose target, pre- and perioperative management of insulin-dependent diabetic patients can be simplified. Dobri and Lansang discuss the key practical principles of managing insulin before the patient goes to the operating suite. They emphasize relevant pearls of insulin physiology and discuss several scenarios we often encounter.

In fact, the principles they review are equally useful to remember when we admit diabetic patients to the hospital with orders to keep them “npo” while planning and awaiting tests or other procedures. A key take-home point is that severely insulinopenic patients require some exogenous basal insulin, even when not eating.

References
  1. Lopes R, Albrecht A, Williams J, et al. Postoperative glucose control following coronary artery bypass graft surgery: predictors and clinical outcomes. J Am Coll Cardiol 2013; 61:e1601.
References
  1. Lopes R, Albrecht A, Williams J, et al. Postoperative glucose control following coronary artery bypass graft surgery: predictors and clinical outcomes. J Am Coll Cardiol 2013; 61:e1601.
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Page Number
682
Page Number
682
Publications
Publications
Topics
Article Type
Display Headline
When people with diabetes go to surgery
Display Headline
When people with diabetes go to surgery
Sections
Disallow All Ads
Alternative CME
Article PDF Media

How should we manage insulin therapy before surgery?

Article Type
Changed
Tue, 05/03/2022 - 15:52
Display Headline
How should we manage insulin therapy before surgery?

Continuing at least part of the basal insulin is the reasonable, physiologic approach to controlling glucose levels before surgery in patients with diabetes. The process involves three basic steps:

  • Ascertaining the type of diabetes
  • Adjusting the basal insulin dosage
  • Stopping the prandial insulin.

The steps are the same whether the surgery is major or minor. These recommendations are based on general principles of insulin action, data from large databases of surgical inpatients, and expert clinical experience translated into standardized protocols.1,2

WHY CONTINUE THE INSULIN?

Stopping or decreasing insulin because of a fear of hypoglycemia is not appropriate, as the resulting hyperglycemia can lead to delayed wound healing, wound infection, fluid and electrolyte shifts, diabetic ketoacidosis, and hyperosmolar states.

Insulin inhibits both gluconeogenesis and conversion of glycogen to glucose, processes that occur regardless of food intake. It also inhibits degradation of fats to fatty acids and of fatty acids to ketones. This is why inadequate insulin dosing can lead to uncontrolled hyperglycemia and even ketoacidosis, and thus why long-acting insulin is needed in a fasting state.

STEP 1: ASCERTAIN THE TYPE OF DIABETES

Does the patient have type 1 or type 2 diabetes, and does that even matter?

The type of diabetes should not matter, since ideally the insulin should be dosed the same for both types. However, the consequences of inappropriate insulin management may be different.

Usually, the type of diabetes can be ascertained by the history. If the patient was diagnosed at age 40 or later and was on oral medication for years before insulin was started, then he or she most likely has type 2. If the patient was younger than 40 at the time of diagnosis, was lean, and was started on insulin within a year of diagnosis, then he or she likely has type 1.

If this information is not available or is unreliable and the patient has been on insulin for many years, we recommend viewing the patient as being insulinopenic, ie, not producing enough insulin endogenously and thus requiring insulin at all times.

Though checking for antibody markers of type 1 diabetes might give a more definitive answer, it is not practical before surgery.

In the setting of surgical stress, withholding the basal insulin preoperatively and just giving a small dose of fast-acting (see Table 1 for the different classes of insulin) or shortacting insulin as part of a sliding scale (ie, insulin given only when the blood glucose reaches a certain high level) can send a patient with type 1 diabetes into diabetic ketoacidosis by the end of the day. This is less likely to occur in a patient with type 2 diabetes with some endogenous insulin secretion.

STEP 2: ADJUST THE BASAL INSULIN

Basal insulin is the insulin that the healthy person’s body produces when fasting. For a diabetic patient already on insulin, basal insulin is insulin injected to prevent ketogenesis, glycogenolysis, and gluconeogenesis in the fasting state.

If the basal insulin is long-acting

Long-acting insulins have a relatively peakless profile and, when properly dosed, should not result in hypoglycemia when a patient is fasting.

Preoperatively, the patient should take it as close as possible to the usual time of injection. This could be at home either at bedtime the night before surgery or the morning of surgery. If there is concern for hypoglycemia, the injection can be given when the patient is at the hospital.

  • If the patient does not tend to have hypoglycemic episodes and the total daily basal insulin dose is roughly the same as the total daily mealtime (prandial) dose (eg, 50% basal, 50% prandial ratio), the full dose of basal insulin can be given.3

Example: If the patient is on insulin glargine 30 U at bedtime and insulin lispro 10 U with each meal and does not have hypoglycemic episodes, then insulin glargine 30 U should be taken at bedtime.

  • If the patient has hypoglycemic episodes at home, then the basal insulin can be reduced by 25%.3

Example: If the patient is on insulin glargine 30 U at bedtime and insulin glulisine 10 U with each meal (appropriate proportion of doses, similar to the example above) but has hypoglycemic episodes at home on this regimen, then only 22 U of insulin glargine should be taken at bedtime.

  • If the patient’s regimen has disproportionately more basal insulin than mealtime insulin, then the total daily doses can be added and half can be given as the basal insulin.

Example: If the patient is on insulin detemir 30 U every morning at 6 am and insulin aspart 6 U with each meal and has no hypoglycemic episodes, then 24 U of insulin detemir should be taken in the morning (ie, half of the total of 30 + 6 + 6 + 6).

  • In the less common scenario of diabetes managed only with basal insulin (no other diabetes injections or oral agents), then half of the dose can be given.
  • If the patient is on twice-daily long-acting basal insulin, then both the dose the night before surgery and the dose the morning of surgery should be adjusted.

 

 

If the basal insulin is intermediate-acting

The intermediate-acting insulin neutral protamine Hagedorn (NPH) is usually given twice a day because of its profile (Table 1).

  • On the night before surgery, the full dose of NPH insulin should be taken, unless the patient will now skip a nighttime meal because of taking nothing by mouth, in which case the dose can be decreased by 25%.1
  • On the morning of surgery, since the patient will be skipping breakfast and probably also lunch, the dose should be reduced by 50%.3,4

Special situation: Premixed insulins

Premixed insulins (70/30, 75/25) are a combination of intermediate-acting insulin and either fast-acting or short-acting insulin. In other words, they are combinations of basal and prandial insulin. Their use is thus not ideal in the preoperative period. There are two options in these situations.

One option is to switch to a regimen that includes long-acting insulin. If the patient is admitted for surgery, then the hospital staff can change the insulin regimen to long-acting basal insulin. A quick formula for conversion is to add all the premixed insulin doses and give half as basal insulin on the morning of surgery, similar to the scenario above for the patient with long-acting basal insulin that was out of proportion to the prandial insulin injections.

For example, if the usual regimen is insulin 70/30 NPH/Regular, 60 U with breakfast, 30 U with dinner, then the patient can take 45 U of insulin glargine (which is half of 60 + 30) in the morning or evening before surgery.

Another option is to adjust the dose of pre-mixed insulin. Sometimes it is not feasible or economical to change the patient’s premixed insulin just before surgery. In these situations, the patient can take half of the morning dose, followed by dextrose-containing intravenous fluids and blood glucose checks.

We recommend preoperatively giving at least part of the patient’s previous basal insulin, regardless of the type of diabetes, the type of surgery, or the fasting period.

STEP 3: STOP THE PRANDIAL INSULIN

Prandial insulin—given before each meal to cover the carbohydrates to be consumed—should be stopped the morning of surgery.3,4

WHAT ABOUT SLIDING SCALE INSULIN?

Using a sliding scale alone has no known benefit. Although it can be a quick fix to correct a high glucose level, it should be added to the basal insulin and not used as the sole insulin therapy. If a sliding scale is used, fast-acting insulin (aspart, glulisine, lispro) is preferred over regular insulin because of the more rapid onset and shorter duration of action.

Patients already using a supplemental insulin scale can apply it to correct a blood glucose above 200 mg/dL on the morning of surgery.

MAINTENANCE FLUIDS

As long as glucose levels are not very elevated (ie, > 200 mg/dL), after 12 hours on a nothing-by-mouth regimen, provide dextrose in the IV fluid to prevent hypoglycemia (eg, the patient received long-acting insulin and the glucose levels are running low) or to prevent starvation ketosis, which may result in ketones in the blood or urine. We recommend 5% dextrose in half-normal (0.45%) saline at 50 to 75 mL/hour as maintenance fluid; the infusion rate should be lower if fluid overload is a concern.

POSTOPERATIVE INSULIN MANAGEMENT

Once patients are discharged and can go back to their previous routine, they can restart their usual insulin regimen the same evening. The prandial insulin will be resumed when the regular diet is reintroduced, and the doses will be adjusted according to food intake.

References
  1. Joshi GP, Chung F, Vann MA, et al; Society for Ambulatory Anesthesia. Society for Ambulatory Anesthesia consensus statement on perioperative blood glucose management in diabetic patients undergoing ambulatory surgery. Anesth Analg 2010; 111:13781387.
  2. DiNardo M, Donihi AC, Forte P, Gieraltowski L, Korytkowski M. Standardized glycemic management and perioperative glycemic outcomes in patients with diabetes mellitus who undergo same-day surgery. Endocr Pract 2011; 17:404411.
  3. Vann MA. Perioperative management of ambulatory surgical patients with diabetes mellitus. Curr Opin Anaesthesiol 2009; 22:718724.
  4. Meneghini LF. Perioperative management of diabetes: translating evidence into practice. Cleve Clin J Med 2009; 76(suppl 4):S53S59.
Article PDF
Author and Disclosure Information

Georgiana A. Dobri, MD
Department of Endocrinology, Diabetes, and Metabolism, Endocrinology and Metabolism Institute, Cleveland Clinic

M. Cecilia Lansang, MD, MPH
Director, Inpatient Diabetes Services, Department of Endocrinology, Diabetes, and Metabolism, Endocrinology and Metabolism Institute, Cleveland Clinic; Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Georgiana A. Dobri, MD, Department of Endocrinology, Diabetes, and Metabolism, F20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail: [email protected]

Issue
Cleveland Clinic Journal of Medicine - 80(11)
Publications
Topics
Page Number
702-704
Sections
Author and Disclosure Information

Georgiana A. Dobri, MD
Department of Endocrinology, Diabetes, and Metabolism, Endocrinology and Metabolism Institute, Cleveland Clinic

M. Cecilia Lansang, MD, MPH
Director, Inpatient Diabetes Services, Department of Endocrinology, Diabetes, and Metabolism, Endocrinology and Metabolism Institute, Cleveland Clinic; Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Georgiana A. Dobri, MD, Department of Endocrinology, Diabetes, and Metabolism, F20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail: [email protected]

Author and Disclosure Information

Georgiana A. Dobri, MD
Department of Endocrinology, Diabetes, and Metabolism, Endocrinology and Metabolism Institute, Cleveland Clinic

M. Cecilia Lansang, MD, MPH
Director, Inpatient Diabetes Services, Department of Endocrinology, Diabetes, and Metabolism, Endocrinology and Metabolism Institute, Cleveland Clinic; Associate Professor of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH

Address: Georgiana A. Dobri, MD, Department of Endocrinology, Diabetes, and Metabolism, F20, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195; e-mail: [email protected]

Article PDF
Article PDF
Related Articles

Continuing at least part of the basal insulin is the reasonable, physiologic approach to controlling glucose levels before surgery in patients with diabetes. The process involves three basic steps:

  • Ascertaining the type of diabetes
  • Adjusting the basal insulin dosage
  • Stopping the prandial insulin.

The steps are the same whether the surgery is major or minor. These recommendations are based on general principles of insulin action, data from large databases of surgical inpatients, and expert clinical experience translated into standardized protocols.1,2

WHY CONTINUE THE INSULIN?

Stopping or decreasing insulin because of a fear of hypoglycemia is not appropriate, as the resulting hyperglycemia can lead to delayed wound healing, wound infection, fluid and electrolyte shifts, diabetic ketoacidosis, and hyperosmolar states.

Insulin inhibits both gluconeogenesis and conversion of glycogen to glucose, processes that occur regardless of food intake. It also inhibits degradation of fats to fatty acids and of fatty acids to ketones. This is why inadequate insulin dosing can lead to uncontrolled hyperglycemia and even ketoacidosis, and thus why long-acting insulin is needed in a fasting state.

STEP 1: ASCERTAIN THE TYPE OF DIABETES

Does the patient have type 1 or type 2 diabetes, and does that even matter?

The type of diabetes should not matter, since ideally the insulin should be dosed the same for both types. However, the consequences of inappropriate insulin management may be different.

Usually, the type of diabetes can be ascertained by the history. If the patient was diagnosed at age 40 or later and was on oral medication for years before insulin was started, then he or she most likely has type 2. If the patient was younger than 40 at the time of diagnosis, was lean, and was started on insulin within a year of diagnosis, then he or she likely has type 1.

If this information is not available or is unreliable and the patient has been on insulin for many years, we recommend viewing the patient as being insulinopenic, ie, not producing enough insulin endogenously and thus requiring insulin at all times.

Though checking for antibody markers of type 1 diabetes might give a more definitive answer, it is not practical before surgery.

In the setting of surgical stress, withholding the basal insulin preoperatively and just giving a small dose of fast-acting (see Table 1 for the different classes of insulin) or shortacting insulin as part of a sliding scale (ie, insulin given only when the blood glucose reaches a certain high level) can send a patient with type 1 diabetes into diabetic ketoacidosis by the end of the day. This is less likely to occur in a patient with type 2 diabetes with some endogenous insulin secretion.

STEP 2: ADJUST THE BASAL INSULIN

Basal insulin is the insulin that the healthy person’s body produces when fasting. For a diabetic patient already on insulin, basal insulin is insulin injected to prevent ketogenesis, glycogenolysis, and gluconeogenesis in the fasting state.

If the basal insulin is long-acting

Long-acting insulins have a relatively peakless profile and, when properly dosed, should not result in hypoglycemia when a patient is fasting.

Preoperatively, the patient should take it as close as possible to the usual time of injection. This could be at home either at bedtime the night before surgery or the morning of surgery. If there is concern for hypoglycemia, the injection can be given when the patient is at the hospital.

  • If the patient does not tend to have hypoglycemic episodes and the total daily basal insulin dose is roughly the same as the total daily mealtime (prandial) dose (eg, 50% basal, 50% prandial ratio), the full dose of basal insulin can be given.3

Example: If the patient is on insulin glargine 30 U at bedtime and insulin lispro 10 U with each meal and does not have hypoglycemic episodes, then insulin glargine 30 U should be taken at bedtime.

  • If the patient has hypoglycemic episodes at home, then the basal insulin can be reduced by 25%.3

Example: If the patient is on insulin glargine 30 U at bedtime and insulin glulisine 10 U with each meal (appropriate proportion of doses, similar to the example above) but has hypoglycemic episodes at home on this regimen, then only 22 U of insulin glargine should be taken at bedtime.

  • If the patient’s regimen has disproportionately more basal insulin than mealtime insulin, then the total daily doses can be added and half can be given as the basal insulin.

Example: If the patient is on insulin detemir 30 U every morning at 6 am and insulin aspart 6 U with each meal and has no hypoglycemic episodes, then 24 U of insulin detemir should be taken in the morning (ie, half of the total of 30 + 6 + 6 + 6).

  • In the less common scenario of diabetes managed only with basal insulin (no other diabetes injections or oral agents), then half of the dose can be given.
  • If the patient is on twice-daily long-acting basal insulin, then both the dose the night before surgery and the dose the morning of surgery should be adjusted.

 

 

If the basal insulin is intermediate-acting

The intermediate-acting insulin neutral protamine Hagedorn (NPH) is usually given twice a day because of its profile (Table 1).

  • On the night before surgery, the full dose of NPH insulin should be taken, unless the patient will now skip a nighttime meal because of taking nothing by mouth, in which case the dose can be decreased by 25%.1
  • On the morning of surgery, since the patient will be skipping breakfast and probably also lunch, the dose should be reduced by 50%.3,4

Special situation: Premixed insulins

Premixed insulins (70/30, 75/25) are a combination of intermediate-acting insulin and either fast-acting or short-acting insulin. In other words, they are combinations of basal and prandial insulin. Their use is thus not ideal in the preoperative period. There are two options in these situations.

One option is to switch to a regimen that includes long-acting insulin. If the patient is admitted for surgery, then the hospital staff can change the insulin regimen to long-acting basal insulin. A quick formula for conversion is to add all the premixed insulin doses and give half as basal insulin on the morning of surgery, similar to the scenario above for the patient with long-acting basal insulin that was out of proportion to the prandial insulin injections.

For example, if the usual regimen is insulin 70/30 NPH/Regular, 60 U with breakfast, 30 U with dinner, then the patient can take 45 U of insulin glargine (which is half of 60 + 30) in the morning or evening before surgery.

Another option is to adjust the dose of pre-mixed insulin. Sometimes it is not feasible or economical to change the patient’s premixed insulin just before surgery. In these situations, the patient can take half of the morning dose, followed by dextrose-containing intravenous fluids and blood glucose checks.

We recommend preoperatively giving at least part of the patient’s previous basal insulin, regardless of the type of diabetes, the type of surgery, or the fasting period.

STEP 3: STOP THE PRANDIAL INSULIN

Prandial insulin—given before each meal to cover the carbohydrates to be consumed—should be stopped the morning of surgery.3,4

WHAT ABOUT SLIDING SCALE INSULIN?

Using a sliding scale alone has no known benefit. Although it can be a quick fix to correct a high glucose level, it should be added to the basal insulin and not used as the sole insulin therapy. If a sliding scale is used, fast-acting insulin (aspart, glulisine, lispro) is preferred over regular insulin because of the more rapid onset and shorter duration of action.

Patients already using a supplemental insulin scale can apply it to correct a blood glucose above 200 mg/dL on the morning of surgery.

MAINTENANCE FLUIDS

As long as glucose levels are not very elevated (ie, > 200 mg/dL), after 12 hours on a nothing-by-mouth regimen, provide dextrose in the IV fluid to prevent hypoglycemia (eg, the patient received long-acting insulin and the glucose levels are running low) or to prevent starvation ketosis, which may result in ketones in the blood or urine. We recommend 5% dextrose in half-normal (0.45%) saline at 50 to 75 mL/hour as maintenance fluid; the infusion rate should be lower if fluid overload is a concern.

POSTOPERATIVE INSULIN MANAGEMENT

Once patients are discharged and can go back to their previous routine, they can restart their usual insulin regimen the same evening. The prandial insulin will be resumed when the regular diet is reintroduced, and the doses will be adjusted according to food intake.

Continuing at least part of the basal insulin is the reasonable, physiologic approach to controlling glucose levels before surgery in patients with diabetes. The process involves three basic steps:

  • Ascertaining the type of diabetes
  • Adjusting the basal insulin dosage
  • Stopping the prandial insulin.

The steps are the same whether the surgery is major or minor. These recommendations are based on general principles of insulin action, data from large databases of surgical inpatients, and expert clinical experience translated into standardized protocols.1,2

WHY CONTINUE THE INSULIN?

Stopping or decreasing insulin because of a fear of hypoglycemia is not appropriate, as the resulting hyperglycemia can lead to delayed wound healing, wound infection, fluid and electrolyte shifts, diabetic ketoacidosis, and hyperosmolar states.

Insulin inhibits both gluconeogenesis and conversion of glycogen to glucose, processes that occur regardless of food intake. It also inhibits degradation of fats to fatty acids and of fatty acids to ketones. This is why inadequate insulin dosing can lead to uncontrolled hyperglycemia and even ketoacidosis, and thus why long-acting insulin is needed in a fasting state.

STEP 1: ASCERTAIN THE TYPE OF DIABETES

Does the patient have type 1 or type 2 diabetes, and does that even matter?

The type of diabetes should not matter, since ideally the insulin should be dosed the same for both types. However, the consequences of inappropriate insulin management may be different.

Usually, the type of diabetes can be ascertained by the history. If the patient was diagnosed at age 40 or later and was on oral medication for years before insulin was started, then he or she most likely has type 2. If the patient was younger than 40 at the time of diagnosis, was lean, and was started on insulin within a year of diagnosis, then he or she likely has type 1.

If this information is not available or is unreliable and the patient has been on insulin for many years, we recommend viewing the patient as being insulinopenic, ie, not producing enough insulin endogenously and thus requiring insulin at all times.

Though checking for antibody markers of type 1 diabetes might give a more definitive answer, it is not practical before surgery.

In the setting of surgical stress, withholding the basal insulin preoperatively and just giving a small dose of fast-acting (see Table 1 for the different classes of insulin) or shortacting insulin as part of a sliding scale (ie, insulin given only when the blood glucose reaches a certain high level) can send a patient with type 1 diabetes into diabetic ketoacidosis by the end of the day. This is less likely to occur in a patient with type 2 diabetes with some endogenous insulin secretion.

STEP 2: ADJUST THE BASAL INSULIN

Basal insulin is the insulin that the healthy person’s body produces when fasting. For a diabetic patient already on insulin, basal insulin is insulin injected to prevent ketogenesis, glycogenolysis, and gluconeogenesis in the fasting state.

If the basal insulin is long-acting

Long-acting insulins have a relatively peakless profile and, when properly dosed, should not result in hypoglycemia when a patient is fasting.

Preoperatively, the patient should take it as close as possible to the usual time of injection. This could be at home either at bedtime the night before surgery or the morning of surgery. If there is concern for hypoglycemia, the injection can be given when the patient is at the hospital.

  • If the patient does not tend to have hypoglycemic episodes and the total daily basal insulin dose is roughly the same as the total daily mealtime (prandial) dose (eg, 50% basal, 50% prandial ratio), the full dose of basal insulin can be given.3

Example: If the patient is on insulin glargine 30 U at bedtime and insulin lispro 10 U with each meal and does not have hypoglycemic episodes, then insulin glargine 30 U should be taken at bedtime.

  • If the patient has hypoglycemic episodes at home, then the basal insulin can be reduced by 25%.3

Example: If the patient is on insulin glargine 30 U at bedtime and insulin glulisine 10 U with each meal (appropriate proportion of doses, similar to the example above) but has hypoglycemic episodes at home on this regimen, then only 22 U of insulin glargine should be taken at bedtime.

  • If the patient’s regimen has disproportionately more basal insulin than mealtime insulin, then the total daily doses can be added and half can be given as the basal insulin.

Example: If the patient is on insulin detemir 30 U every morning at 6 am and insulin aspart 6 U with each meal and has no hypoglycemic episodes, then 24 U of insulin detemir should be taken in the morning (ie, half of the total of 30 + 6 + 6 + 6).

  • In the less common scenario of diabetes managed only with basal insulin (no other diabetes injections or oral agents), then half of the dose can be given.
  • If the patient is on twice-daily long-acting basal insulin, then both the dose the night before surgery and the dose the morning of surgery should be adjusted.

 

 

If the basal insulin is intermediate-acting

The intermediate-acting insulin neutral protamine Hagedorn (NPH) is usually given twice a day because of its profile (Table 1).

  • On the night before surgery, the full dose of NPH insulin should be taken, unless the patient will now skip a nighttime meal because of taking nothing by mouth, in which case the dose can be decreased by 25%.1
  • On the morning of surgery, since the patient will be skipping breakfast and probably also lunch, the dose should be reduced by 50%.3,4

Special situation: Premixed insulins

Premixed insulins (70/30, 75/25) are a combination of intermediate-acting insulin and either fast-acting or short-acting insulin. In other words, they are combinations of basal and prandial insulin. Their use is thus not ideal in the preoperative period. There are two options in these situations.

One option is to switch to a regimen that includes long-acting insulin. If the patient is admitted for surgery, then the hospital staff can change the insulin regimen to long-acting basal insulin. A quick formula for conversion is to add all the premixed insulin doses and give half as basal insulin on the morning of surgery, similar to the scenario above for the patient with long-acting basal insulin that was out of proportion to the prandial insulin injections.

For example, if the usual regimen is insulin 70/30 NPH/Regular, 60 U with breakfast, 30 U with dinner, then the patient can take 45 U of insulin glargine (which is half of 60 + 30) in the morning or evening before surgery.

Another option is to adjust the dose of pre-mixed insulin. Sometimes it is not feasible or economical to change the patient’s premixed insulin just before surgery. In these situations, the patient can take half of the morning dose, followed by dextrose-containing intravenous fluids and blood glucose checks.

We recommend preoperatively giving at least part of the patient’s previous basal insulin, regardless of the type of diabetes, the type of surgery, or the fasting period.

STEP 3: STOP THE PRANDIAL INSULIN

Prandial insulin—given before each meal to cover the carbohydrates to be consumed—should be stopped the morning of surgery.3,4

WHAT ABOUT SLIDING SCALE INSULIN?

Using a sliding scale alone has no known benefit. Although it can be a quick fix to correct a high glucose level, it should be added to the basal insulin and not used as the sole insulin therapy. If a sliding scale is used, fast-acting insulin (aspart, glulisine, lispro) is preferred over regular insulin because of the more rapid onset and shorter duration of action.

Patients already using a supplemental insulin scale can apply it to correct a blood glucose above 200 mg/dL on the morning of surgery.

MAINTENANCE FLUIDS

As long as glucose levels are not very elevated (ie, > 200 mg/dL), after 12 hours on a nothing-by-mouth regimen, provide dextrose in the IV fluid to prevent hypoglycemia (eg, the patient received long-acting insulin and the glucose levels are running low) or to prevent starvation ketosis, which may result in ketones in the blood or urine. We recommend 5% dextrose in half-normal (0.45%) saline at 50 to 75 mL/hour as maintenance fluid; the infusion rate should be lower if fluid overload is a concern.

POSTOPERATIVE INSULIN MANAGEMENT

Once patients are discharged and can go back to their previous routine, they can restart their usual insulin regimen the same evening. The prandial insulin will be resumed when the regular diet is reintroduced, and the doses will be adjusted according to food intake.

References
  1. Joshi GP, Chung F, Vann MA, et al; Society for Ambulatory Anesthesia. Society for Ambulatory Anesthesia consensus statement on perioperative blood glucose management in diabetic patients undergoing ambulatory surgery. Anesth Analg 2010; 111:13781387.
  2. DiNardo M, Donihi AC, Forte P, Gieraltowski L, Korytkowski M. Standardized glycemic management and perioperative glycemic outcomes in patients with diabetes mellitus who undergo same-day surgery. Endocr Pract 2011; 17:404411.
  3. Vann MA. Perioperative management of ambulatory surgical patients with diabetes mellitus. Curr Opin Anaesthesiol 2009; 22:718724.
  4. Meneghini LF. Perioperative management of diabetes: translating evidence into practice. Cleve Clin J Med 2009; 76(suppl 4):S53S59.
References
  1. Joshi GP, Chung F, Vann MA, et al; Society for Ambulatory Anesthesia. Society for Ambulatory Anesthesia consensus statement on perioperative blood glucose management in diabetic patients undergoing ambulatory surgery. Anesth Analg 2010; 111:13781387.
  2. DiNardo M, Donihi AC, Forte P, Gieraltowski L, Korytkowski M. Standardized glycemic management and perioperative glycemic outcomes in patients with diabetes mellitus who undergo same-day surgery. Endocr Pract 2011; 17:404411.
  3. Vann MA. Perioperative management of ambulatory surgical patients with diabetes mellitus. Curr Opin Anaesthesiol 2009; 22:718724.
  4. Meneghini LF. Perioperative management of diabetes: translating evidence into practice. Cleve Clin J Med 2009; 76(suppl 4):S53S59.
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Issue
Cleveland Clinic Journal of Medicine - 80(11)
Page Number
702-704
Page Number
702-704
Publications
Publications
Topics
Article Type
Display Headline
How should we manage insulin therapy before surgery?
Display Headline
How should we manage insulin therapy before surgery?
Sections
Disallow All Ads
Alternative CME
Article PDF Media

BioniCare&reg; in the Treatment of Arthritis of the Hand and Wrist

Article Type
Changed
Tue, 05/21/2019 - 12:17
Display Headline
BioniCare® in the Treatment of Arthritis of the Hand and Wrist

This educational supplement to RHEUMATOLOGY NEWS was sponsored by BIONICARE® By VQOrthoCare®.

Peter A. Holt, MD
Associate Professor of Medicine
The Johns Hopkins University School of Medicine
Baltimore, Maryland

Tuna Ozyurekoglu, MD
Christine M. Kleinert Institute for Hand and Microsurgery
Louisville, Kentucky

Shaili Deveshwar, MD
Sports Medicine and Orthopedic Center
Greensboro, North Carolina 

Edmund J. MacLaughlin, MD 
Cambridge, Maryland

Shirley W. Pang, MD
St. Jude Heritage Medical Group
Fullerton, California

Jack S. Tuber, DO 
SunValley Arthritis Center
Peoria, Arizona

Joy Schechtman, DO 
SunValley Arthritis CenterPeoria, Arizona

Thomas M. Zizic, MD
Associate Professor of Medicine
The Johns Hopkins University School of Medicine
Baltimore, Maryland

Article PDF
Publications
Sections
Article PDF
Article PDF

This educational supplement to RHEUMATOLOGY NEWS was sponsored by BIONICARE® By VQOrthoCare®.

Peter A. Holt, MD
Associate Professor of Medicine
The Johns Hopkins University School of Medicine
Baltimore, Maryland

Tuna Ozyurekoglu, MD
Christine M. Kleinert Institute for Hand and Microsurgery
Louisville, Kentucky

Shaili Deveshwar, MD
Sports Medicine and Orthopedic Center
Greensboro, North Carolina 

Edmund J. MacLaughlin, MD 
Cambridge, Maryland

Shirley W. Pang, MD
St. Jude Heritage Medical Group
Fullerton, California

Jack S. Tuber, DO 
SunValley Arthritis Center
Peoria, Arizona

Joy Schechtman, DO 
SunValley Arthritis CenterPeoria, Arizona

Thomas M. Zizic, MD
Associate Professor of Medicine
The Johns Hopkins University School of Medicine
Baltimore, Maryland

This educational supplement to RHEUMATOLOGY NEWS was sponsored by BIONICARE® By VQOrthoCare®.

Peter A. Holt, MD
Associate Professor of Medicine
The Johns Hopkins University School of Medicine
Baltimore, Maryland

Tuna Ozyurekoglu, MD
Christine M. Kleinert Institute for Hand and Microsurgery
Louisville, Kentucky

Shaili Deveshwar, MD
Sports Medicine and Orthopedic Center
Greensboro, North Carolina 

Edmund J. MacLaughlin, MD 
Cambridge, Maryland

Shirley W. Pang, MD
St. Jude Heritage Medical Group
Fullerton, California

Jack S. Tuber, DO 
SunValley Arthritis Center
Peoria, Arizona

Joy Schechtman, DO 
SunValley Arthritis CenterPeoria, Arizona

Thomas M. Zizic, MD
Associate Professor of Medicine
The Johns Hopkins University School of Medicine
Baltimore, Maryland

Publications
Publications
Article Type
Display Headline
BioniCare® in the Treatment of Arthritis of the Hand and Wrist
Display Headline
BioniCare® in the Treatment of Arthritis of the Hand and Wrist
Sections
Disallow All Ads
Article PDF Media

Drug resistance: What FPs can do

Article Type
Changed
Mon, 01/14/2019 - 13:44
Display Headline
Drug resistance: What FPs can do

Author and Disclosure Information

University of Arizona College of Medicine, Phoenix

Issue
The Journal of Family Practice - 62(11)
Publications
Topics
Page Number
audio
Author and Disclosure Information

University of Arizona College of Medicine, Phoenix

Author and Disclosure Information

University of Arizona College of Medicine, Phoenix

Issue
The Journal of Family Practice - 62(11)
Issue
The Journal of Family Practice - 62(11)
Page Number
audio
Page Number
audio
Publications
Publications
Topics
Article Type
Display Headline
Drug resistance: What FPs can do
Display Headline
Drug resistance: What FPs can do
Disallow All Ads
Alternative CME
Use ProPublica

High Users of Hospital Care

Article Type
Changed
Sun, 05/21/2017 - 17:35
Display Headline
What's cost got to do with it? Association between hospital costs and frequency of admissions among “high users” of hospital care

Despite signs of a slowing trend,[1, 2, 3] US healthcare costs continue to rise, and cost containment remains a major area of concern. Hospital costs are the largest single category of national healthcare expenditures,[4] and the burden of cost containment is increasingly being shifted to hospitals.[5] As such, hospitals are increasingly focusing on implementing interventions to reduce rates of hospitalizations and readmissions as a mechanism to reduce overall healthcare costs.[5, 6, 7, 8, 9]

Multiple factors potentially contribute to patients being high cost, including acute care utilization,[10, 11, 12] pharmaceuticals,[13, 14] procedures,[15] catastrophic illness,[16] and high‐risk chronic conditions.[11, 17, 18] However, many hospitals are implementing interventions focused on a single subset of these high cost patientshigh users of inpatient services. Care management interventions have received particular attention, due to their perceived potential to improve quality of care while reducing costs through the mechanism of reducing hospital admissions.[19, 20, 21] Despite their increasing prevalence, there is limited evidence demonstrating the effectiveness of these programs. Among interventions targeting high cost individuals, the Medicare Care Management for High Cost Beneficiaries showed no effect on hospital admissions.[19] Another high‐profile intervention, the Citywide Care Management System led by the Camden Coalition, showed promising preliminary results, but data from a systematic evaluation are lacking.[22] Conversely, interventions targeting individuals with frequent hospitalizations have similarly shown mixed results in reducing costs.[6, 7, 9, 23]

Taken together, these data suggest that the relationship between high costs and frequent hospital use is complicated, and the population of individuals with frequent hospitalizations may not represent the entire population of high cost individuals. Thus, focusing on reduction of hospitalizations alone may be inadequate to reduce costs. For these reasons, we sought to determine how many high cost individuals would be captured by focusing only on frequently hospitalized (high admit) individuals by examining the overlap between these populations. We also sought to describe the characteristics and distinctions between the resulting subgroups of high users to better inform the design of future interventions.

METHODS

We examined the cross‐sectional relationship between high cost and high admit populations among adult patients 18 years of age hospitalized at the University of California, San Francisco (UCSF) Medical Center, a 660‐bed urban academic medical center from July 1, 2010 to June 30, 2011.

This study was conducted as part of a quality improvement project to identify high user primary care patients for complex care management. Individuals were included in the study if: (1) they had an assigned UCSF primary care provider (PCP), and (2) they had at least 1 hospitalization at UCSF during the study period. PCP assignments were ascertained from panel lists maintained by clinics; lists included individuals with at least 1 visit at any of the 8 primary care clinics at UCSF in the 2 years prior to the end of the study period. Because individuals are dropped from PCP panels at death, we were unable to ascertain or include individuals who died during the study period.

From the initial study population, we defined the high cost group as those who were in the top decile of total hospital costs, and the high admit group as those who were in the top decile of total hospitalizations during the study period. We elected to use the top decile as a cutoff given that it is a common operational definition used to identify high users to target for intervention.[24]

To examine the relationship between high cost and high admits we defined 3 further subgroups: high costlow admits, high costhigh admits, and low costhigh admits (Figure 1). To explore the face validity of these descriptors and classification scheme, we subsequently examined the proportion of total hospital costs and total hospitalizations each subgroup accounted for with respect to the study population.

Figure 1
Defining “high user” subgroups. aIndividuals with a primary care provider (PCP) were defined as those with a PCP visit in the prior 2 years. Because individuals are dropped from PCP assignments at death, we were unable to ascertain or include individuals who died during the study period.

Data Sources

Hospital costs, demographic data, and encounter diagnoses were obtained from the hospital's Transition Systems Incorporated system (TSI, also known as Eclipsys or Allscripts), a commercially available automated cost accounting system that integrates multiple data sources to calculate total hospital costs on a per patient basis. Several studies have previously used the TSI system to estimate the costs of healthcare services at individual hospitals, and this approach is generally considered the most accurate method to estimate cost.[25, 26] Hospital costs included the sum of actual total costs (not billed charges) for all hospital episodes including lab costs, drug costs, surgical supplies, nurse salaries and benefits, utilities, housekeeping, and allocated administrative overhead. This cost total does not capture the cost of physician labor (pro‐fees), preadmission costs (e.g., outpatient care), or postadmission costs (e.g., home health, nursing home, or other postdischarge care). Preadmission lab, diagnostic tests, and imaging were included in hospital costs if these were ordered within 72 hours of hospital admission. Emergency department (ED) costs were included if an individual was admitted to the hospital via the ED. Hospitalizations were defined as inpatient admissions only to UCSF because we were unable to reliably ascertain hospitalizations outside of UCSF. PCP assignments were ascertained from administrative panel lists maintained by clinics.

Study Variables

We analyzed factors previously shown[13, 27, 28, 29] to be associated with high healthcare cost and utilization. We examined demographic characteristics and hospitalization characteristics, including admission source, length of stay (LOS), cost per hospitalization, whether the episode was a 30‐day readmission, days in the intensive care unit (ICU), and encounter diagnoses.

To ascertain whether a hospitalization was for a medical versus surgical condition, we used discharge diagnosis codes and designations as per the Medicare Severity Diagnosis‐Related Groups (MS‐DRG) versions 27 and 28 definitions manuals. We subsequently grouped medical and surgical conditions by Major Diagnostic Categories as per the MS‐DRG definitions manuals.

Using MS‐DRG codes, we also classified whether hospitalizations were for pneumonia, acute myocardial infarction (AMI), and congestive heart failure (CHF), as these 3 conditions have specific payment penalties under the Centers for Medicare & Medicaid Services (CMS) reimbursement policies. For these CMS core conditions, we included hospitalizations with MS‐DRG codes 193195, 280282, and 291293 (codes 283285 were not included for AMI because individuals who died during the study period were excluded.)

Analysis

We used descriptive statistics to compare patient and hospitalization characteristics between subgroups. Non‐normally distributed variables including LOS and cost per hospitalization were log transformed. Because a single individual could account for multiple hospitalizations, we performed a companion analysis of hospitalization characteristics using generalized estimating equations with an independent correlation structure to account for clustering of hospitalizations within individuals. Our findings were robust using either approach. For ease of interpretation, P values from the former analysis are presented.

To determine whether the overall distribution and characteristics we observed for high user subgroups were a single‐year anomaly or representative of temporally stable trends, we compared non‐high users and high user subgroup characteristics over the 3 years preceding the study period using linear regression for trend.

The institutional review board at UCSF approved this study protocol.

RESULTS

Of the 2566 unique individuals included in the analysis (Figure 1), 256 individuals were identified as high cost (top decile, $65,000). This group accounted for 45% of all costs and 22% of all hospitalizations (Figure 2). Two hundred fifty individuals were identified as high admits (top decile, 3 hospitalizations). This group accounted for 32% of all costs and 28% of all hospitalizations.

Figure 2
“High users” account for a disproportionate share of costs and admissions. The top 10% of individuals by costs = high cost–low admit   high cost–high admit subgroups; the top 10% of individuals by admissions = high cost–high admit   low cost–high admit subgroups. aTotal costs = sum of all hospital costs for study population. bTotal admissions = sum of all hospital admissions for study population.

Only 48% of high cost individuals were also high admit ($65,000 and 3 hospitalizations; n=125, Figure 1). Among high users, we subsequently defined 3 subgroups based on the relationship between cost and hospitalizations (high costlow admits, high costhigh admits, and low costhigh admits). Each subgroup accounted for approximately 5% of the overall study population (Figure 2). The high costlow admits subgroup incurred a proportionate share of hospitalizations (6%) but a disproportionate share of costs (20%). The high costhigh admits subgroup had a disproportionate share of both costs (25%) and hospitalizations (16%). The low costhigh admits subgroup had a proportionate share of costs (7%) but a disproportionate share of hospitalizations (12%).

Patient and Hospitalization Characteristics

Compared to non‐high users, all high user subgroups were more likely to have public insurance (Medicare or Medicaid) or have dualeligible status, and the two high cost subgroups were more likely to be male and African American (P<0.05 for all). Compared to each other, subgroups were similar with respect to race/ethnicity, payer, and age (Table 1).

Patient Characteristics Among High User Subgroups
 Non‐High Users,n=2145High User Subgroups 
123P Value for Comparison
High CostLow Admit, n=131High CostHigh Admit, n=125Low CostHigh Admit, n=1251 vs 22 vs 31 vs 3
  • NOTE: Abbreviations: SD, standard deviation. *Dual eligible=Medicare and Medicaid as primary and secondary payers, respectively.

Male, %345742380.020.520.003
Race/ethnicity, %    0.150.760.18
White45463638   
Black14212619   
Hispanic951010   
Asian22182226   
Other101066   
Primary payer, %    0.230.440.51
Commercial42242017   
Medicare42585561   
Medicaid15172521   
Other<12 2   
Dual eligible, %*182726310.850.380.48
Age, mean yearsSD57206017581763200.400.040.19
No. of hospitalizations per individual, median (interquartile range)1 (11)2 (12)4 (36)3 (34)<0.001<0.001<0.001
Hospital costs per individual, median $1000 (interquartile range)12 (722)93 (75122)113 (85174)37 (3051)<0.001<0.001<0.001

Regarding hospitalization characteristics, each high user subgroup was distinct and significantly different from each of the other subgroups with respect to admission source, proportion of 30‐day readmissions, LOS, and cost per hospitalization (Table 2, P<0.001 for all). The low costhigh admit subgroup had the highest proportion of admissions from the ED (73%), a moderate proportion of 30‐day readmissions (32%), the shortest LOS (median, 3 days; interquartile range [IQR], 24 days) and the lowest cost per hospitalization (median, $12,000; IQR, $8,000$15,000). In contrast, the high costlow admit subgroup had the highest proportion of admissions from clinic or physician referrals (45%), lowest proportion of 30‐day readmissions (17%), the longest LOS (median, 10; IQR, 417), the most ICU days per hospitalization (median, 1; range, 049) and the highest cost per hospitalization (median, $68,000; IQR, $43,000$95,000). High costhigh admit individuals had the highest proportion of 30‐day readmissions (47%) and a moderate cost per hospitalization (median, $28,000; IQR, $23,000$38,000), but the highest median cost per individual over 1 year ($113,000; IQR, $85,000$174,000, Table 1). Hospitalizations classified as 30‐day readmissions accounted for 42% of costs incurred by this subgroup; 30‐day readmissions specifically associated with CMS core conditions accounted for <1% of costs.

Hospitalization Characteristics and Encounter Diagnoses
 Non‐High UsersHigh User Subgroups 
123P Value for Comparison
High CostLow AdmitHigh CostHigh AdmitLow CostHigh Admit1 vs 22 vs 31 vs 3
  • NOTE: Abbreviations: LOS, length of stay; IQR, interquartile range; MS‐DRG, Medicare Severity Diagnosis‐Related Group; MDC, Major Diagnostic Category; CMS, Centers for Medicare & Medicaid Services. *Interquartile ranges for non‐high users and each high user subgroup were 00, 04, 00, and 00, respectively. Comparisons were done for proportion of hospitalizations for surgical versus medical MS‐DRGs (not for MDCs). CMS core conditions defined using MS‐DRG codes for pneumonia, acute myocardial infarction, and congestive heart failure.

No. of admissions2500206605431   
Admission source, %    <0.001<0.001<0.001
Emergency department53506573   
Clinic or physician referral44453020   
Transfer from outside facility2544   
Self‐referral1<113   
Other<1      
30‐day readmission, %5174732<0.001<0.001<0.001
LOS, median days (IQR)3 (24)10 (417)5 (310)3 (24)<0.001<0.001<0.001
ICU days, median (range)*0 (08)1 (049)0 (021)0 (03)<0.001<0.001<0.001
Cost per hospitalization, median $1,000 (IQR)11 (719)68 (4395)28 (2338)12 (815)<0.001<0.001<0.001
Encounter diagnoses    <0.0010.002<0.001
Surgical MS‐DRGs, %30582213   
Most common MDCs       
Cardiovascular41586   
Orthopedic101364   
Transplant<1711   
Medical MS‐DRGs, %70427887   
Most common MDCs       
Pregnancy related17222   
Cardiovascular1010713   
Respiratory941417   
Gastrointestinal731014   
Hematologic1296   
Myeloproliferative<1496   
CMS core condition736120.1740.010.004

Encounter diagnoses associated with hospitalizations were also significantly different between each of the high user subgroups (Table 2, P<0.001 for all). The high costlow admit subgroup was predominantly hospitalized for surgical conditions (58% vs 42% for medical MS‐DRGs) and had the lowest proportion of hospitalizations for CMS core conditions (3%). The most common types of surgical hospitalizations in this subgroup were for cardiovascular procedures (15%; including coronary artery bypass grafting and cardiac valve replacement) and orthopedic procedures (13%; including hip, knee, and other joint replacements). Most surgical hospitalizations were from referrals (67%) rather than admissions through the ED. In contrast, the low costhigh admit group was predominantly hospitalized for medical conditions (87% vs 13% for surgical MS‐DRGs) and had the highest proportion of hospitalizations for CMS core conditions (12%). The most common types of medical hospitalizations in this subgroup were for respiratory conditions (17%; including chronic obstructive pulmonary disease and pneumonia), gastrointestinal conditions (14%), and cardiovascular conditions (13%; including CHF, AMI, arrhythmia, and chest pain). High costhigh admit individuals were also hospitalized primarily for medical rather than surgical conditions (78% vs 22% medical vs surgical MS‐DRGs). Only 6% of hospitalizations in this subgroup were for CMS core conditions, and only 2% of hospitalizations were 30‐day readmissions for CMS core conditions.

The overlap between the high cost and high admit groups was persistently 48% or less for the 3 years prior to the study period (Table 3). Although the extent of overlap was similar across years, the absolute dollar value for the cutoff to define the top decile by hospital costs gradually increased over time from $47,000 in 2008 to $65,000 in 2011 (P<0.001 for trend). Among the high costlow admit subgroup, there was a trend toward a decrease in the proportion of surgical hospitalizations from 67% in 2008 to 58% in 2011 (P=0.09).

Temporal Stability in High User Subgroup Distribution and Discharge Diagnoses
 2008200920102011P Value (For Linear Trend)
  • NOTE: Abbreviations: MS‐DRG, Medicare Severity Diagnosis‐Related Group.*Values are given as percentage and (number) of admissions for each subgroup.

Study population2408251826472566 
Characteristics, n     
Cutoff for high cost (top decile), nearest $1000>47>51>54>65<0.001
Proportion of total hospital costs incurred by high cost group, %46474748 
Cutoff for high admit (top decile), no. of admissions3333 
High cost who are also high admit, %42484848 
Discharge diagnoses by subgroup*     
Non‐high user population     
Surgical MS‐DRG32 (751)33 (842)36 (932)30 (751)0.51
Medical MS‐DRG68 (1598)67 (1676)64 (1673)70 (1749) 
High costlow admit     
Surgical MS‐DRG67 (138)68 (132)61 (120)58 (119)0.09
Medical MS‐DRG33(67)32 (63)39 (78)42 (87) 
High costhigh admit     
Surgical MS‐DRG23 (104)25 (133)24 (150)22 (134)0.60
Medical MS‐DRG77 (341)75 (392)76 (464)78 (471) 
Low costhigh admit     
Surgical MS‐DRG11 (35)17 (44)13 (40)13 (54)0.90
Medical MS‐DRG89 (277)83 (219)87 (269)87 (377) 

DISCUSSION

In this study, we found that only half of high cost individuals were also high admit. Further categorizing high users into high costlow admit versus high costhigh admit versus low costhigh admit identified distinct patterns between each group. High costhigh admit individuals were more likely to be hospitalized for medical conditions, whereas high costlow admit individuals were more likely to be hospitalized for surgical conditions. CMS core conditions accounted for a low proportion of overall hospitalizations across all groups.

Our findings suggest several distinct types of high users with different clinical characteristics, utilization, and cost patterns. From a hospital perspective, one implication is that a multifaceted approach to cost containment, rather than the one‐size‐fits‐all strategy of reducing hospitalizations, may be more effective in reducing costs. For example, our findings show that high costlow admit individuals have a disproportionate number of hospitalizations for surgical conditions, longer LOS, and more ICU days. Costs incurred by this subgroup may be more responsive to in‐hospital interventions aimed at reducing procedural costs, LOS, unnecessary use of the ICU, and minimizing postoperative infections and complications rather than to a care management approach.

In contrast, care management strategies such as improving postdischarge care and chronic disease management, which aim to achieve cost savings through reducing hospitalizations, may be more effective in reducing costs among high costhigh admit individuals, who have a high proportion of hospitalizations for medical conditions and the highest proportion of 30‐day readmissions. Such strategies may also be important in optimizing the quality of care for low costhigh admit individuals, who have the highest proportion of medical hospitalizations among all high users, though the potential for cost savings may be more limited in this subgroup.

Our results suggest that current hospital‐based approachesdriven by readmissions penalties for CMS core conditionsmay have less than the expected impact on costs. For example, although high costhigh admit individuals had the highest proportion of 30‐day readmissions, readmissions specifically for CMS core conditions accounted for <1% of costs in this subgroup. Thus, the potential return on an expensive investment in a care management intervention is unclear, given the small number of readmissions for these select conditions. From a broader perspective, the focus on readmissions for CMS core conditions, which overall contribute relatively little to high hospital costs, may not be a comprehensive enough strategy for cost containment. To date, there have been limited policies targeting factors contributing to high hospital costs outside of frequent medical hospitalizations. Medicare's nonpayment policy for treatment of preventable hospital conditions, including surgical site infections, translates prevention of these conditions into cost savings for hospitals.[30] However, this rule has been criticized for not going far enough to drive substantial savings.[31] A new CMS rule authorizes states to identify other provider‐preventable conditions for which Medicaid payment will be prohibited.[32] Future policy efforts should further emphasize a comprehensive, multipronged approach beyond readmissions penalties for select conditions if healthcare cost containment remains a policy priority.

Our results should be interpreted in light of several limitations. First, this was a single‐site study at an academic medical center; the generalizability of our results to other settings is unclear. Our cost data likely reflect local market factors, including the highest wage rates for skilled healthcare labor in the United States.[33] Although the explicit distribution of high user subgroups may be institution‐specific due to variations in our cost structure, we anticipate that the general classification paradigm will be similar in other health systems. Second, we captured utilization and costs only at a single hospital. However, our study population includes only individuals with PCPs at UCSF, and internal data from both Medicare and UCSF's largest private payer show that over 85% of hospitalizations among this population are to UCSF Medical Center. Third, we were able to capture only hospital costs rather than overall healthcare costs. Given that hospital costs account for the single largest category of total national health costs,[4] we expect that future studies examining total health costs will show similar findings. Fourth, our data did not include measures of health status, socioeconomic status, housing, or mental health comorbidities to permit an analysis of these factors, which have been previously related to frequent hospitalizations and high costs.[34, 35, 36, 37, 38, 39] Fifth, due to resource constraints, we were unable to conduct a longitudinal analysis to examine the extent to which individuals are consistently high users over time. Previous studies have described that 20% to 30% of individuals are consistently high users; the remainder have discrete periods of high utilization.[34, 40] This may be an important consideration in the design of future interventions.

Finally, our analysis was limited to individuals with a PCP to allow identification of an accessible cohort for care management. Thus, we did not capture individuals without a PCP and individuals who died during the study period, because these individuals no longer had an assigned PCP following death. Although this approach is consistent with that of many care management programs,[19] these populations are likely to incur higher than average utilization and healthcare costs, and represent important areas for future investigation.

In summary, our study identifies three types of high‐user populations that differ in the proportion of costs attributable to frequent hospitalizations, clinical conditions associated with hospital use, and frequency of 30‐day readmissions. Stratifying high users by both costs and hospitalizations may help identify tailored strategies to more effectively reduce costs and utilization.

Acknowledgments

The authors acknowledge Diana Patterson, Leanna Zaporozhets, and Andre Devito for their assistance in data collection.

Disclosures: Dr. Nguyen had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Dr. Nguyen's work on this project was completed as a primary care research fellow at the University of California, San Francisco funded by a federal training grant from the National Research Service Award (NRSA T32HP19025‐07‐00).

Files
References
  1. Martin A, Lassman D, Whittle L, Catlin A; National Health Expenditure Accounts Team. Recession contributes to slowest annual rate of increase in health spending in five decades. Health Aff (Millwood). 2011;30(1):1122.
  2. Martin AB, Lassman D, Washington B, Catlin A. Growth in US health spending remained slow in 2010; health share of gross domestic product was unchanged from 2009. Health Aff (Millwood). 2012;31(1):208219.
  3. Hartman M, Martin AB, Benson J, Catlin A; the National Health Expenditure Accounts Team. National health spending in 2011: overall growth remains low, but some payers and services show signs of acceleration. Health Aff (Millwood). 2013;32(1):8799.
  4. Centers for Medicare and Medicaid Services; Office of the Actuary; National Health Statistics Group. National healthcare expenditures data. 2012. Available at: https://www.cms.gov/Research‐Statistics‐Data‐and‐Systems/Statistics‐Trends‐and‐Reports/NationalHealthExpendData/downloads/tables.pdf.
  5. Medicare Payment Advisory Commission. Report to the Congress: Promoting Greater Efficiency in Medicare. Washington, DC: Medicare Payment Advisory Commission; 2007.
  6. Coleman EA, Parry C, Chalmers S, Min SJ. The care transitions intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):18221828.
  7. Jack BW, Chetty VK, Anthony D, et al. A reengineered hospital discharge program to decrease rehospitalization: a randomized trial. Ann Intern Med. 2009;150(3):178187.
  8. Schall M, Coleman E, Rutherford P, Taylor J. How‐to Guide: Improving Transitions from the Hospital to the Clinical Office Practice to Reduce Avoidable Rehospitalizations. Cambridge, MA: Institute for Healthcare Improvement; 2012.
  9. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow‐up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613620.
  10. Zook CJ, Moore FD. High‐cost users of medical care. N Engl J Med. 1980;302(18):9961002.
  11. Schroeder SA, Showstack JA, Roberts HE. Frequency and clinical description of high‐cost patients in 17 acute‐care hospitals. N Engl J Med. 1979;300(23):13061309.
  12. Althaus F, Paroz S, Hugli O, et al. Effectiveness of interventions targeting frequent users of emergency departments: a systematic review. Ann Emerg Med. 2011;58(1):4152.e42.
  13. Forrest CB, Lemke KW, Bodycombe DP, Weiner JP. Medication, diagnostic, and cost information as predictors of high‐risk patients in need of care management. Am J Manag Care. 2009;15(1):4148.
  14. Strunk BC, Ginsburg PB. Tracking health care costs: trends stabilize but remain high in 2002. Health Aff (Millwood). 2003;Suppl Web Exclusives:W3266–274.
  15. Newhouse JP. An iconoclastic view of health cost containment. Health Aff (Millwood). 1993;12(suppl):152171.
  16. Nan‐Ping Y, Yi‐Hui L, Chi‐Yu C, et al. Comparisons of medical utilizations and categorical diagnoses of emergency visits between the elderly with catastrophic illness certificates and those without. BMC Health Serv Res. 2013;13:152.
  17. Ash AS, Zhao Y, Ellis RP, Schlein Kramer M. Finding future high‐cost cases: comparing prior cost versus diagnosis‐based methods. Health Serv Res. 2001;36(6 pt 2):194206.
  18. Cohen JW, Krauss NA. Spending and service use among people with the fifteen most costly medical conditions, 1997. Health Aff (Millwood). 2003;22(2):129138.
  19. Nelson L. Lessons from Medicare's Demonstration Projects on Disease Management and Care Coordination. Washington, D.C.: Congressional Budget Office; 2012.
  20. Gawande A. The hot spotters. The New Yorker. January 24, 2011.
  21. Freshman B, Rubino LG, Reid Chassiakos Y. Collaboration Across the Disciplines in Health Care. Burlington, MA: Jones and Bartlett; 2010.
  22. Green SR, Singh V, O'Byrne W. Hope for New Jersey's city hospitals: the Camden Initiative. Perspect Health Inf Manag. 2010:7:1d.
  23. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital‐initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433440.
  24. Agency for Healthcare Research and Quality. The concentration and persistence in the level of health expenditures over time: estimates for the U.S. population, 2008–2009. 2012. Available at: http://meps.ahrq.gov/mepsweb/data_files/publications/st354/stat354.shtml.
  25. Azoulay A, Doris NM, Filion KB, Caron J, Pilote L, Eisenberg MJ. The use of the transition cost accounting system in health services research. Cost Eff Resour Alloc. 2007;5:11.
  26. Pronovost P, Angus DC. Cost reduction and quality improvement: it takes two to tango. Crit Care Med. 2000;28(2):581583.
  27. Billings J, Dixon J, Mijanovich T, Wennberg D. Case finding for patients at risk of readmission to hospital: development of algorithm to identify high risk patients. BMJ. 2006;333(7563):327.
  28. Kansagara D, Englander H, Salanitro A, et al. Risk prediction models for hospital readmission: a systematic review. JAMA. 2011;306(15):16881698.
  29. Putnam KG, Buist DS, Fishman P, et al. Chronic disease score as a predictor of hospitalization. Epidemiology. 2002;13(3):340346.
  30. Centers for Medicare and Medicaid Services. Hospital‐acquired conditions (HAC) in acute inpatient prospective payment system (IPPS) hospitals fact sheet. 2011. Available at: https://www.cms.gov/Medicare/Medicare‐Fee‐for‐Service‐Payment/HospitalAcqCond/downloads/HACFactsheet.pdf. Accessed August 31, 2013.
  31. McNair PD, Luft HS, Bindman AB. Medicare's policy not to pay for treating hospital‐acquired conditions: the impact. Health Aff (Millwood). 2009;28(5):14851493.
  32. Department of Health and Human Services, Centers for Medicare 2013.
  33. Freeborn DK, Pope CR, Mullooly JP, McFarland BH. Consistently high users of medical care among the elderly. Med Care. 1990;28(6):527540.
  34. McFarland BH, Freeborn DK, Mullooly JP, Pope CR. Utilization patterns among long‐term enrollees in a prepaid group practice health maintenance organization. Med Care. 1985;23(11):12211233.
  35. Waxman HM, Carner EA, Blum A. Depressive symptoms and health service utilization among the community elderly. J Am Geriatr Soc. 1983;31(7):417420.
  36. Kuo RN, Lai MS. The influence of socio‐economic status and multimorbidity patterns on healthcare costs: a six‐year follow‐up under a universal healthcare system. Int J Equity Health. 2013;12(1):69.
  37. Reid KW, Vittinghoff E, Kushel MB. Association between the level of housing instability, economic standing and health care access: a meta‐regression. J Health Care Poor Underserved. 2008;19(4):12121228.
  38. Lemstra M, Mackenbach J, Neudorf C, Nannapaneni U. High health care utilization and costs associated with lower socio‐economic status: results from a linked dataset. Can J Public Health. 2009;100(3):180183.
  39. McCall N, Wai HS. An analysis of the use of Medicare services by the continuously enrolled aged. Med Care. 1983;21(6):567585.
Article PDF
Issue
Journal of Hospital Medicine - 8(12)
Page Number
665-671
Sections
Files
Files
Article PDF
Article PDF

Despite signs of a slowing trend,[1, 2, 3] US healthcare costs continue to rise, and cost containment remains a major area of concern. Hospital costs are the largest single category of national healthcare expenditures,[4] and the burden of cost containment is increasingly being shifted to hospitals.[5] As such, hospitals are increasingly focusing on implementing interventions to reduce rates of hospitalizations and readmissions as a mechanism to reduce overall healthcare costs.[5, 6, 7, 8, 9]

Multiple factors potentially contribute to patients being high cost, including acute care utilization,[10, 11, 12] pharmaceuticals,[13, 14] procedures,[15] catastrophic illness,[16] and high‐risk chronic conditions.[11, 17, 18] However, many hospitals are implementing interventions focused on a single subset of these high cost patientshigh users of inpatient services. Care management interventions have received particular attention, due to their perceived potential to improve quality of care while reducing costs through the mechanism of reducing hospital admissions.[19, 20, 21] Despite their increasing prevalence, there is limited evidence demonstrating the effectiveness of these programs. Among interventions targeting high cost individuals, the Medicare Care Management for High Cost Beneficiaries showed no effect on hospital admissions.[19] Another high‐profile intervention, the Citywide Care Management System led by the Camden Coalition, showed promising preliminary results, but data from a systematic evaluation are lacking.[22] Conversely, interventions targeting individuals with frequent hospitalizations have similarly shown mixed results in reducing costs.[6, 7, 9, 23]

Taken together, these data suggest that the relationship between high costs and frequent hospital use is complicated, and the population of individuals with frequent hospitalizations may not represent the entire population of high cost individuals. Thus, focusing on reduction of hospitalizations alone may be inadequate to reduce costs. For these reasons, we sought to determine how many high cost individuals would be captured by focusing only on frequently hospitalized (high admit) individuals by examining the overlap between these populations. We also sought to describe the characteristics and distinctions between the resulting subgroups of high users to better inform the design of future interventions.

METHODS

We examined the cross‐sectional relationship between high cost and high admit populations among adult patients 18 years of age hospitalized at the University of California, San Francisco (UCSF) Medical Center, a 660‐bed urban academic medical center from July 1, 2010 to June 30, 2011.

This study was conducted as part of a quality improvement project to identify high user primary care patients for complex care management. Individuals were included in the study if: (1) they had an assigned UCSF primary care provider (PCP), and (2) they had at least 1 hospitalization at UCSF during the study period. PCP assignments were ascertained from panel lists maintained by clinics; lists included individuals with at least 1 visit at any of the 8 primary care clinics at UCSF in the 2 years prior to the end of the study period. Because individuals are dropped from PCP panels at death, we were unable to ascertain or include individuals who died during the study period.

From the initial study population, we defined the high cost group as those who were in the top decile of total hospital costs, and the high admit group as those who were in the top decile of total hospitalizations during the study period. We elected to use the top decile as a cutoff given that it is a common operational definition used to identify high users to target for intervention.[24]

To examine the relationship between high cost and high admits we defined 3 further subgroups: high costlow admits, high costhigh admits, and low costhigh admits (Figure 1). To explore the face validity of these descriptors and classification scheme, we subsequently examined the proportion of total hospital costs and total hospitalizations each subgroup accounted for with respect to the study population.

Figure 1
Defining “high user” subgroups. aIndividuals with a primary care provider (PCP) were defined as those with a PCP visit in the prior 2 years. Because individuals are dropped from PCP assignments at death, we were unable to ascertain or include individuals who died during the study period.

Data Sources

Hospital costs, demographic data, and encounter diagnoses were obtained from the hospital's Transition Systems Incorporated system (TSI, also known as Eclipsys or Allscripts), a commercially available automated cost accounting system that integrates multiple data sources to calculate total hospital costs on a per patient basis. Several studies have previously used the TSI system to estimate the costs of healthcare services at individual hospitals, and this approach is generally considered the most accurate method to estimate cost.[25, 26] Hospital costs included the sum of actual total costs (not billed charges) for all hospital episodes including lab costs, drug costs, surgical supplies, nurse salaries and benefits, utilities, housekeeping, and allocated administrative overhead. This cost total does not capture the cost of physician labor (pro‐fees), preadmission costs (e.g., outpatient care), or postadmission costs (e.g., home health, nursing home, or other postdischarge care). Preadmission lab, diagnostic tests, and imaging were included in hospital costs if these were ordered within 72 hours of hospital admission. Emergency department (ED) costs were included if an individual was admitted to the hospital via the ED. Hospitalizations were defined as inpatient admissions only to UCSF because we were unable to reliably ascertain hospitalizations outside of UCSF. PCP assignments were ascertained from administrative panel lists maintained by clinics.

Study Variables

We analyzed factors previously shown[13, 27, 28, 29] to be associated with high healthcare cost and utilization. We examined demographic characteristics and hospitalization characteristics, including admission source, length of stay (LOS), cost per hospitalization, whether the episode was a 30‐day readmission, days in the intensive care unit (ICU), and encounter diagnoses.

To ascertain whether a hospitalization was for a medical versus surgical condition, we used discharge diagnosis codes and designations as per the Medicare Severity Diagnosis‐Related Groups (MS‐DRG) versions 27 and 28 definitions manuals. We subsequently grouped medical and surgical conditions by Major Diagnostic Categories as per the MS‐DRG definitions manuals.

Using MS‐DRG codes, we also classified whether hospitalizations were for pneumonia, acute myocardial infarction (AMI), and congestive heart failure (CHF), as these 3 conditions have specific payment penalties under the Centers for Medicare & Medicaid Services (CMS) reimbursement policies. For these CMS core conditions, we included hospitalizations with MS‐DRG codes 193195, 280282, and 291293 (codes 283285 were not included for AMI because individuals who died during the study period were excluded.)

Analysis

We used descriptive statistics to compare patient and hospitalization characteristics between subgroups. Non‐normally distributed variables including LOS and cost per hospitalization were log transformed. Because a single individual could account for multiple hospitalizations, we performed a companion analysis of hospitalization characteristics using generalized estimating equations with an independent correlation structure to account for clustering of hospitalizations within individuals. Our findings were robust using either approach. For ease of interpretation, P values from the former analysis are presented.

To determine whether the overall distribution and characteristics we observed for high user subgroups were a single‐year anomaly or representative of temporally stable trends, we compared non‐high users and high user subgroup characteristics over the 3 years preceding the study period using linear regression for trend.

The institutional review board at UCSF approved this study protocol.

RESULTS

Of the 2566 unique individuals included in the analysis (Figure 1), 256 individuals were identified as high cost (top decile, $65,000). This group accounted for 45% of all costs and 22% of all hospitalizations (Figure 2). Two hundred fifty individuals were identified as high admits (top decile, 3 hospitalizations). This group accounted for 32% of all costs and 28% of all hospitalizations.

Figure 2
“High users” account for a disproportionate share of costs and admissions. The top 10% of individuals by costs = high cost–low admit   high cost–high admit subgroups; the top 10% of individuals by admissions = high cost–high admit   low cost–high admit subgroups. aTotal costs = sum of all hospital costs for study population. bTotal admissions = sum of all hospital admissions for study population.

Only 48% of high cost individuals were also high admit ($65,000 and 3 hospitalizations; n=125, Figure 1). Among high users, we subsequently defined 3 subgroups based on the relationship between cost and hospitalizations (high costlow admits, high costhigh admits, and low costhigh admits). Each subgroup accounted for approximately 5% of the overall study population (Figure 2). The high costlow admits subgroup incurred a proportionate share of hospitalizations (6%) but a disproportionate share of costs (20%). The high costhigh admits subgroup had a disproportionate share of both costs (25%) and hospitalizations (16%). The low costhigh admits subgroup had a proportionate share of costs (7%) but a disproportionate share of hospitalizations (12%).

Patient and Hospitalization Characteristics

Compared to non‐high users, all high user subgroups were more likely to have public insurance (Medicare or Medicaid) or have dualeligible status, and the two high cost subgroups were more likely to be male and African American (P<0.05 for all). Compared to each other, subgroups were similar with respect to race/ethnicity, payer, and age (Table 1).

Patient Characteristics Among High User Subgroups
 Non‐High Users,n=2145High User Subgroups 
123P Value for Comparison
High CostLow Admit, n=131High CostHigh Admit, n=125Low CostHigh Admit, n=1251 vs 22 vs 31 vs 3
  • NOTE: Abbreviations: SD, standard deviation. *Dual eligible=Medicare and Medicaid as primary and secondary payers, respectively.

Male, %345742380.020.520.003
Race/ethnicity, %    0.150.760.18
White45463638   
Black14212619   
Hispanic951010   
Asian22182226   
Other101066   
Primary payer, %    0.230.440.51
Commercial42242017   
Medicare42585561   
Medicaid15172521   
Other<12 2   
Dual eligible, %*182726310.850.380.48
Age, mean yearsSD57206017581763200.400.040.19
No. of hospitalizations per individual, median (interquartile range)1 (11)2 (12)4 (36)3 (34)<0.001<0.001<0.001
Hospital costs per individual, median $1000 (interquartile range)12 (722)93 (75122)113 (85174)37 (3051)<0.001<0.001<0.001

Regarding hospitalization characteristics, each high user subgroup was distinct and significantly different from each of the other subgroups with respect to admission source, proportion of 30‐day readmissions, LOS, and cost per hospitalization (Table 2, P<0.001 for all). The low costhigh admit subgroup had the highest proportion of admissions from the ED (73%), a moderate proportion of 30‐day readmissions (32%), the shortest LOS (median, 3 days; interquartile range [IQR], 24 days) and the lowest cost per hospitalization (median, $12,000; IQR, $8,000$15,000). In contrast, the high costlow admit subgroup had the highest proportion of admissions from clinic or physician referrals (45%), lowest proportion of 30‐day readmissions (17%), the longest LOS (median, 10; IQR, 417), the most ICU days per hospitalization (median, 1; range, 049) and the highest cost per hospitalization (median, $68,000; IQR, $43,000$95,000). High costhigh admit individuals had the highest proportion of 30‐day readmissions (47%) and a moderate cost per hospitalization (median, $28,000; IQR, $23,000$38,000), but the highest median cost per individual over 1 year ($113,000; IQR, $85,000$174,000, Table 1). Hospitalizations classified as 30‐day readmissions accounted for 42% of costs incurred by this subgroup; 30‐day readmissions specifically associated with CMS core conditions accounted for <1% of costs.

Hospitalization Characteristics and Encounter Diagnoses
 Non‐High UsersHigh User Subgroups 
123P Value for Comparison
High CostLow AdmitHigh CostHigh AdmitLow CostHigh Admit1 vs 22 vs 31 vs 3
  • NOTE: Abbreviations: LOS, length of stay; IQR, interquartile range; MS‐DRG, Medicare Severity Diagnosis‐Related Group; MDC, Major Diagnostic Category; CMS, Centers for Medicare & Medicaid Services. *Interquartile ranges for non‐high users and each high user subgroup were 00, 04, 00, and 00, respectively. Comparisons were done for proportion of hospitalizations for surgical versus medical MS‐DRGs (not for MDCs). CMS core conditions defined using MS‐DRG codes for pneumonia, acute myocardial infarction, and congestive heart failure.

No. of admissions2500206605431   
Admission source, %    <0.001<0.001<0.001
Emergency department53506573   
Clinic or physician referral44453020   
Transfer from outside facility2544   
Self‐referral1<113   
Other<1      
30‐day readmission, %5174732<0.001<0.001<0.001
LOS, median days (IQR)3 (24)10 (417)5 (310)3 (24)<0.001<0.001<0.001
ICU days, median (range)*0 (08)1 (049)0 (021)0 (03)<0.001<0.001<0.001
Cost per hospitalization, median $1,000 (IQR)11 (719)68 (4395)28 (2338)12 (815)<0.001<0.001<0.001
Encounter diagnoses    <0.0010.002<0.001
Surgical MS‐DRGs, %30582213   
Most common MDCs       
Cardiovascular41586   
Orthopedic101364   
Transplant<1711   
Medical MS‐DRGs, %70427887   
Most common MDCs       
Pregnancy related17222   
Cardiovascular1010713   
Respiratory941417   
Gastrointestinal731014   
Hematologic1296   
Myeloproliferative<1496   
CMS core condition736120.1740.010.004

Encounter diagnoses associated with hospitalizations were also significantly different between each of the high user subgroups (Table 2, P<0.001 for all). The high costlow admit subgroup was predominantly hospitalized for surgical conditions (58% vs 42% for medical MS‐DRGs) and had the lowest proportion of hospitalizations for CMS core conditions (3%). The most common types of surgical hospitalizations in this subgroup were for cardiovascular procedures (15%; including coronary artery bypass grafting and cardiac valve replacement) and orthopedic procedures (13%; including hip, knee, and other joint replacements). Most surgical hospitalizations were from referrals (67%) rather than admissions through the ED. In contrast, the low costhigh admit group was predominantly hospitalized for medical conditions (87% vs 13% for surgical MS‐DRGs) and had the highest proportion of hospitalizations for CMS core conditions (12%). The most common types of medical hospitalizations in this subgroup were for respiratory conditions (17%; including chronic obstructive pulmonary disease and pneumonia), gastrointestinal conditions (14%), and cardiovascular conditions (13%; including CHF, AMI, arrhythmia, and chest pain). High costhigh admit individuals were also hospitalized primarily for medical rather than surgical conditions (78% vs 22% medical vs surgical MS‐DRGs). Only 6% of hospitalizations in this subgroup were for CMS core conditions, and only 2% of hospitalizations were 30‐day readmissions for CMS core conditions.

The overlap between the high cost and high admit groups was persistently 48% or less for the 3 years prior to the study period (Table 3). Although the extent of overlap was similar across years, the absolute dollar value for the cutoff to define the top decile by hospital costs gradually increased over time from $47,000 in 2008 to $65,000 in 2011 (P<0.001 for trend). Among the high costlow admit subgroup, there was a trend toward a decrease in the proportion of surgical hospitalizations from 67% in 2008 to 58% in 2011 (P=0.09).

Temporal Stability in High User Subgroup Distribution and Discharge Diagnoses
 2008200920102011P Value (For Linear Trend)
  • NOTE: Abbreviations: MS‐DRG, Medicare Severity Diagnosis‐Related Group.*Values are given as percentage and (number) of admissions for each subgroup.

Study population2408251826472566 
Characteristics, n     
Cutoff for high cost (top decile), nearest $1000>47>51>54>65<0.001
Proportion of total hospital costs incurred by high cost group, %46474748 
Cutoff for high admit (top decile), no. of admissions3333 
High cost who are also high admit, %42484848 
Discharge diagnoses by subgroup*     
Non‐high user population     
Surgical MS‐DRG32 (751)33 (842)36 (932)30 (751)0.51
Medical MS‐DRG68 (1598)67 (1676)64 (1673)70 (1749) 
High costlow admit     
Surgical MS‐DRG67 (138)68 (132)61 (120)58 (119)0.09
Medical MS‐DRG33(67)32 (63)39 (78)42 (87) 
High costhigh admit     
Surgical MS‐DRG23 (104)25 (133)24 (150)22 (134)0.60
Medical MS‐DRG77 (341)75 (392)76 (464)78 (471) 
Low costhigh admit     
Surgical MS‐DRG11 (35)17 (44)13 (40)13 (54)0.90
Medical MS‐DRG89 (277)83 (219)87 (269)87 (377) 

DISCUSSION

In this study, we found that only half of high cost individuals were also high admit. Further categorizing high users into high costlow admit versus high costhigh admit versus low costhigh admit identified distinct patterns between each group. High costhigh admit individuals were more likely to be hospitalized for medical conditions, whereas high costlow admit individuals were more likely to be hospitalized for surgical conditions. CMS core conditions accounted for a low proportion of overall hospitalizations across all groups.

Our findings suggest several distinct types of high users with different clinical characteristics, utilization, and cost patterns. From a hospital perspective, one implication is that a multifaceted approach to cost containment, rather than the one‐size‐fits‐all strategy of reducing hospitalizations, may be more effective in reducing costs. For example, our findings show that high costlow admit individuals have a disproportionate number of hospitalizations for surgical conditions, longer LOS, and more ICU days. Costs incurred by this subgroup may be more responsive to in‐hospital interventions aimed at reducing procedural costs, LOS, unnecessary use of the ICU, and minimizing postoperative infections and complications rather than to a care management approach.

In contrast, care management strategies such as improving postdischarge care and chronic disease management, which aim to achieve cost savings through reducing hospitalizations, may be more effective in reducing costs among high costhigh admit individuals, who have a high proportion of hospitalizations for medical conditions and the highest proportion of 30‐day readmissions. Such strategies may also be important in optimizing the quality of care for low costhigh admit individuals, who have the highest proportion of medical hospitalizations among all high users, though the potential for cost savings may be more limited in this subgroup.

Our results suggest that current hospital‐based approachesdriven by readmissions penalties for CMS core conditionsmay have less than the expected impact on costs. For example, although high costhigh admit individuals had the highest proportion of 30‐day readmissions, readmissions specifically for CMS core conditions accounted for <1% of costs in this subgroup. Thus, the potential return on an expensive investment in a care management intervention is unclear, given the small number of readmissions for these select conditions. From a broader perspective, the focus on readmissions for CMS core conditions, which overall contribute relatively little to high hospital costs, may not be a comprehensive enough strategy for cost containment. To date, there have been limited policies targeting factors contributing to high hospital costs outside of frequent medical hospitalizations. Medicare's nonpayment policy for treatment of preventable hospital conditions, including surgical site infections, translates prevention of these conditions into cost savings for hospitals.[30] However, this rule has been criticized for not going far enough to drive substantial savings.[31] A new CMS rule authorizes states to identify other provider‐preventable conditions for which Medicaid payment will be prohibited.[32] Future policy efforts should further emphasize a comprehensive, multipronged approach beyond readmissions penalties for select conditions if healthcare cost containment remains a policy priority.

Our results should be interpreted in light of several limitations. First, this was a single‐site study at an academic medical center; the generalizability of our results to other settings is unclear. Our cost data likely reflect local market factors, including the highest wage rates for skilled healthcare labor in the United States.[33] Although the explicit distribution of high user subgroups may be institution‐specific due to variations in our cost structure, we anticipate that the general classification paradigm will be similar in other health systems. Second, we captured utilization and costs only at a single hospital. However, our study population includes only individuals with PCPs at UCSF, and internal data from both Medicare and UCSF's largest private payer show that over 85% of hospitalizations among this population are to UCSF Medical Center. Third, we were able to capture only hospital costs rather than overall healthcare costs. Given that hospital costs account for the single largest category of total national health costs,[4] we expect that future studies examining total health costs will show similar findings. Fourth, our data did not include measures of health status, socioeconomic status, housing, or mental health comorbidities to permit an analysis of these factors, which have been previously related to frequent hospitalizations and high costs.[34, 35, 36, 37, 38, 39] Fifth, due to resource constraints, we were unable to conduct a longitudinal analysis to examine the extent to which individuals are consistently high users over time. Previous studies have described that 20% to 30% of individuals are consistently high users; the remainder have discrete periods of high utilization.[34, 40] This may be an important consideration in the design of future interventions.

Finally, our analysis was limited to individuals with a PCP to allow identification of an accessible cohort for care management. Thus, we did not capture individuals without a PCP and individuals who died during the study period, because these individuals no longer had an assigned PCP following death. Although this approach is consistent with that of many care management programs,[19] these populations are likely to incur higher than average utilization and healthcare costs, and represent important areas for future investigation.

In summary, our study identifies three types of high‐user populations that differ in the proportion of costs attributable to frequent hospitalizations, clinical conditions associated with hospital use, and frequency of 30‐day readmissions. Stratifying high users by both costs and hospitalizations may help identify tailored strategies to more effectively reduce costs and utilization.

Acknowledgments

The authors acknowledge Diana Patterson, Leanna Zaporozhets, and Andre Devito for their assistance in data collection.

Disclosures: Dr. Nguyen had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Dr. Nguyen's work on this project was completed as a primary care research fellow at the University of California, San Francisco funded by a federal training grant from the National Research Service Award (NRSA T32HP19025‐07‐00).

Despite signs of a slowing trend,[1, 2, 3] US healthcare costs continue to rise, and cost containment remains a major area of concern. Hospital costs are the largest single category of national healthcare expenditures,[4] and the burden of cost containment is increasingly being shifted to hospitals.[5] As such, hospitals are increasingly focusing on implementing interventions to reduce rates of hospitalizations and readmissions as a mechanism to reduce overall healthcare costs.[5, 6, 7, 8, 9]

Multiple factors potentially contribute to patients being high cost, including acute care utilization,[10, 11, 12] pharmaceuticals,[13, 14] procedures,[15] catastrophic illness,[16] and high‐risk chronic conditions.[11, 17, 18] However, many hospitals are implementing interventions focused on a single subset of these high cost patientshigh users of inpatient services. Care management interventions have received particular attention, due to their perceived potential to improve quality of care while reducing costs through the mechanism of reducing hospital admissions.[19, 20, 21] Despite their increasing prevalence, there is limited evidence demonstrating the effectiveness of these programs. Among interventions targeting high cost individuals, the Medicare Care Management for High Cost Beneficiaries showed no effect on hospital admissions.[19] Another high‐profile intervention, the Citywide Care Management System led by the Camden Coalition, showed promising preliminary results, but data from a systematic evaluation are lacking.[22] Conversely, interventions targeting individuals with frequent hospitalizations have similarly shown mixed results in reducing costs.[6, 7, 9, 23]

Taken together, these data suggest that the relationship between high costs and frequent hospital use is complicated, and the population of individuals with frequent hospitalizations may not represent the entire population of high cost individuals. Thus, focusing on reduction of hospitalizations alone may be inadequate to reduce costs. For these reasons, we sought to determine how many high cost individuals would be captured by focusing only on frequently hospitalized (high admit) individuals by examining the overlap between these populations. We also sought to describe the characteristics and distinctions between the resulting subgroups of high users to better inform the design of future interventions.

METHODS

We examined the cross‐sectional relationship between high cost and high admit populations among adult patients 18 years of age hospitalized at the University of California, San Francisco (UCSF) Medical Center, a 660‐bed urban academic medical center from July 1, 2010 to June 30, 2011.

This study was conducted as part of a quality improvement project to identify high user primary care patients for complex care management. Individuals were included in the study if: (1) they had an assigned UCSF primary care provider (PCP), and (2) they had at least 1 hospitalization at UCSF during the study period. PCP assignments were ascertained from panel lists maintained by clinics; lists included individuals with at least 1 visit at any of the 8 primary care clinics at UCSF in the 2 years prior to the end of the study period. Because individuals are dropped from PCP panels at death, we were unable to ascertain or include individuals who died during the study period.

From the initial study population, we defined the high cost group as those who were in the top decile of total hospital costs, and the high admit group as those who were in the top decile of total hospitalizations during the study period. We elected to use the top decile as a cutoff given that it is a common operational definition used to identify high users to target for intervention.[24]

To examine the relationship between high cost and high admits we defined 3 further subgroups: high costlow admits, high costhigh admits, and low costhigh admits (Figure 1). To explore the face validity of these descriptors and classification scheme, we subsequently examined the proportion of total hospital costs and total hospitalizations each subgroup accounted for with respect to the study population.

Figure 1
Defining “high user” subgroups. aIndividuals with a primary care provider (PCP) were defined as those with a PCP visit in the prior 2 years. Because individuals are dropped from PCP assignments at death, we were unable to ascertain or include individuals who died during the study period.

Data Sources

Hospital costs, demographic data, and encounter diagnoses were obtained from the hospital's Transition Systems Incorporated system (TSI, also known as Eclipsys or Allscripts), a commercially available automated cost accounting system that integrates multiple data sources to calculate total hospital costs on a per patient basis. Several studies have previously used the TSI system to estimate the costs of healthcare services at individual hospitals, and this approach is generally considered the most accurate method to estimate cost.[25, 26] Hospital costs included the sum of actual total costs (not billed charges) for all hospital episodes including lab costs, drug costs, surgical supplies, nurse salaries and benefits, utilities, housekeeping, and allocated administrative overhead. This cost total does not capture the cost of physician labor (pro‐fees), preadmission costs (e.g., outpatient care), or postadmission costs (e.g., home health, nursing home, or other postdischarge care). Preadmission lab, diagnostic tests, and imaging were included in hospital costs if these were ordered within 72 hours of hospital admission. Emergency department (ED) costs were included if an individual was admitted to the hospital via the ED. Hospitalizations were defined as inpatient admissions only to UCSF because we were unable to reliably ascertain hospitalizations outside of UCSF. PCP assignments were ascertained from administrative panel lists maintained by clinics.

Study Variables

We analyzed factors previously shown[13, 27, 28, 29] to be associated with high healthcare cost and utilization. We examined demographic characteristics and hospitalization characteristics, including admission source, length of stay (LOS), cost per hospitalization, whether the episode was a 30‐day readmission, days in the intensive care unit (ICU), and encounter diagnoses.

To ascertain whether a hospitalization was for a medical versus surgical condition, we used discharge diagnosis codes and designations as per the Medicare Severity Diagnosis‐Related Groups (MS‐DRG) versions 27 and 28 definitions manuals. We subsequently grouped medical and surgical conditions by Major Diagnostic Categories as per the MS‐DRG definitions manuals.

Using MS‐DRG codes, we also classified whether hospitalizations were for pneumonia, acute myocardial infarction (AMI), and congestive heart failure (CHF), as these 3 conditions have specific payment penalties under the Centers for Medicare & Medicaid Services (CMS) reimbursement policies. For these CMS core conditions, we included hospitalizations with MS‐DRG codes 193195, 280282, and 291293 (codes 283285 were not included for AMI because individuals who died during the study period were excluded.)

Analysis

We used descriptive statistics to compare patient and hospitalization characteristics between subgroups. Non‐normally distributed variables including LOS and cost per hospitalization were log transformed. Because a single individual could account for multiple hospitalizations, we performed a companion analysis of hospitalization characteristics using generalized estimating equations with an independent correlation structure to account for clustering of hospitalizations within individuals. Our findings were robust using either approach. For ease of interpretation, P values from the former analysis are presented.

To determine whether the overall distribution and characteristics we observed for high user subgroups were a single‐year anomaly or representative of temporally stable trends, we compared non‐high users and high user subgroup characteristics over the 3 years preceding the study period using linear regression for trend.

The institutional review board at UCSF approved this study protocol.

RESULTS

Of the 2566 unique individuals included in the analysis (Figure 1), 256 individuals were identified as high cost (top decile, $65,000). This group accounted for 45% of all costs and 22% of all hospitalizations (Figure 2). Two hundred fifty individuals were identified as high admits (top decile, 3 hospitalizations). This group accounted for 32% of all costs and 28% of all hospitalizations.

Figure 2
“High users” account for a disproportionate share of costs and admissions. The top 10% of individuals by costs = high cost–low admit   high cost–high admit subgroups; the top 10% of individuals by admissions = high cost–high admit   low cost–high admit subgroups. aTotal costs = sum of all hospital costs for study population. bTotal admissions = sum of all hospital admissions for study population.

Only 48% of high cost individuals were also high admit ($65,000 and 3 hospitalizations; n=125, Figure 1). Among high users, we subsequently defined 3 subgroups based on the relationship between cost and hospitalizations (high costlow admits, high costhigh admits, and low costhigh admits). Each subgroup accounted for approximately 5% of the overall study population (Figure 2). The high costlow admits subgroup incurred a proportionate share of hospitalizations (6%) but a disproportionate share of costs (20%). The high costhigh admits subgroup had a disproportionate share of both costs (25%) and hospitalizations (16%). The low costhigh admits subgroup had a proportionate share of costs (7%) but a disproportionate share of hospitalizations (12%).

Patient and Hospitalization Characteristics

Compared to non‐high users, all high user subgroups were more likely to have public insurance (Medicare or Medicaid) or have dualeligible status, and the two high cost subgroups were more likely to be male and African American (P<0.05 for all). Compared to each other, subgroups were similar with respect to race/ethnicity, payer, and age (Table 1).

Patient Characteristics Among High User Subgroups
 Non‐High Users,n=2145High User Subgroups 
123P Value for Comparison
High CostLow Admit, n=131High CostHigh Admit, n=125Low CostHigh Admit, n=1251 vs 22 vs 31 vs 3
  • NOTE: Abbreviations: SD, standard deviation. *Dual eligible=Medicare and Medicaid as primary and secondary payers, respectively.

Male, %345742380.020.520.003
Race/ethnicity, %    0.150.760.18
White45463638   
Black14212619   
Hispanic951010   
Asian22182226   
Other101066   
Primary payer, %    0.230.440.51
Commercial42242017   
Medicare42585561   
Medicaid15172521   
Other<12 2   
Dual eligible, %*182726310.850.380.48
Age, mean yearsSD57206017581763200.400.040.19
No. of hospitalizations per individual, median (interquartile range)1 (11)2 (12)4 (36)3 (34)<0.001<0.001<0.001
Hospital costs per individual, median $1000 (interquartile range)12 (722)93 (75122)113 (85174)37 (3051)<0.001<0.001<0.001

Regarding hospitalization characteristics, each high user subgroup was distinct and significantly different from each of the other subgroups with respect to admission source, proportion of 30‐day readmissions, LOS, and cost per hospitalization (Table 2, P<0.001 for all). The low costhigh admit subgroup had the highest proportion of admissions from the ED (73%), a moderate proportion of 30‐day readmissions (32%), the shortest LOS (median, 3 days; interquartile range [IQR], 24 days) and the lowest cost per hospitalization (median, $12,000; IQR, $8,000$15,000). In contrast, the high costlow admit subgroup had the highest proportion of admissions from clinic or physician referrals (45%), lowest proportion of 30‐day readmissions (17%), the longest LOS (median, 10; IQR, 417), the most ICU days per hospitalization (median, 1; range, 049) and the highest cost per hospitalization (median, $68,000; IQR, $43,000$95,000). High costhigh admit individuals had the highest proportion of 30‐day readmissions (47%) and a moderate cost per hospitalization (median, $28,000; IQR, $23,000$38,000), but the highest median cost per individual over 1 year ($113,000; IQR, $85,000$174,000, Table 1). Hospitalizations classified as 30‐day readmissions accounted for 42% of costs incurred by this subgroup; 30‐day readmissions specifically associated with CMS core conditions accounted for <1% of costs.

Hospitalization Characteristics and Encounter Diagnoses
 Non‐High UsersHigh User Subgroups 
123P Value for Comparison
High CostLow AdmitHigh CostHigh AdmitLow CostHigh Admit1 vs 22 vs 31 vs 3
  • NOTE: Abbreviations: LOS, length of stay; IQR, interquartile range; MS‐DRG, Medicare Severity Diagnosis‐Related Group; MDC, Major Diagnostic Category; CMS, Centers for Medicare & Medicaid Services. *Interquartile ranges for non‐high users and each high user subgroup were 00, 04, 00, and 00, respectively. Comparisons were done for proportion of hospitalizations for surgical versus medical MS‐DRGs (not for MDCs). CMS core conditions defined using MS‐DRG codes for pneumonia, acute myocardial infarction, and congestive heart failure.

No. of admissions2500206605431   
Admission source, %    <0.001<0.001<0.001
Emergency department53506573   
Clinic or physician referral44453020   
Transfer from outside facility2544   
Self‐referral1<113   
Other<1      
30‐day readmission, %5174732<0.001<0.001<0.001
LOS, median days (IQR)3 (24)10 (417)5 (310)3 (24)<0.001<0.001<0.001
ICU days, median (range)*0 (08)1 (049)0 (021)0 (03)<0.001<0.001<0.001
Cost per hospitalization, median $1,000 (IQR)11 (719)68 (4395)28 (2338)12 (815)<0.001<0.001<0.001
Encounter diagnoses    <0.0010.002<0.001
Surgical MS‐DRGs, %30582213   
Most common MDCs       
Cardiovascular41586   
Orthopedic101364   
Transplant<1711   
Medical MS‐DRGs, %70427887   
Most common MDCs       
Pregnancy related17222   
Cardiovascular1010713   
Respiratory941417   
Gastrointestinal731014   
Hematologic1296   
Myeloproliferative<1496   
CMS core condition736120.1740.010.004

Encounter diagnoses associated with hospitalizations were also significantly different between each of the high user subgroups (Table 2, P<0.001 for all). The high costlow admit subgroup was predominantly hospitalized for surgical conditions (58% vs 42% for medical MS‐DRGs) and had the lowest proportion of hospitalizations for CMS core conditions (3%). The most common types of surgical hospitalizations in this subgroup were for cardiovascular procedures (15%; including coronary artery bypass grafting and cardiac valve replacement) and orthopedic procedures (13%; including hip, knee, and other joint replacements). Most surgical hospitalizations were from referrals (67%) rather than admissions through the ED. In contrast, the low costhigh admit group was predominantly hospitalized for medical conditions (87% vs 13% for surgical MS‐DRGs) and had the highest proportion of hospitalizations for CMS core conditions (12%). The most common types of medical hospitalizations in this subgroup were for respiratory conditions (17%; including chronic obstructive pulmonary disease and pneumonia), gastrointestinal conditions (14%), and cardiovascular conditions (13%; including CHF, AMI, arrhythmia, and chest pain). High costhigh admit individuals were also hospitalized primarily for medical rather than surgical conditions (78% vs 22% medical vs surgical MS‐DRGs). Only 6% of hospitalizations in this subgroup were for CMS core conditions, and only 2% of hospitalizations were 30‐day readmissions for CMS core conditions.

The overlap between the high cost and high admit groups was persistently 48% or less for the 3 years prior to the study period (Table 3). Although the extent of overlap was similar across years, the absolute dollar value for the cutoff to define the top decile by hospital costs gradually increased over time from $47,000 in 2008 to $65,000 in 2011 (P<0.001 for trend). Among the high costlow admit subgroup, there was a trend toward a decrease in the proportion of surgical hospitalizations from 67% in 2008 to 58% in 2011 (P=0.09).

Temporal Stability in High User Subgroup Distribution and Discharge Diagnoses
 2008200920102011P Value (For Linear Trend)
  • NOTE: Abbreviations: MS‐DRG, Medicare Severity Diagnosis‐Related Group.*Values are given as percentage and (number) of admissions for each subgroup.

Study population2408251826472566 
Characteristics, n     
Cutoff for high cost (top decile), nearest $1000>47>51>54>65<0.001
Proportion of total hospital costs incurred by high cost group, %46474748 
Cutoff for high admit (top decile), no. of admissions3333 
High cost who are also high admit, %42484848 
Discharge diagnoses by subgroup*     
Non‐high user population     
Surgical MS‐DRG32 (751)33 (842)36 (932)30 (751)0.51
Medical MS‐DRG68 (1598)67 (1676)64 (1673)70 (1749) 
High costlow admit     
Surgical MS‐DRG67 (138)68 (132)61 (120)58 (119)0.09
Medical MS‐DRG33(67)32 (63)39 (78)42 (87) 
High costhigh admit     
Surgical MS‐DRG23 (104)25 (133)24 (150)22 (134)0.60
Medical MS‐DRG77 (341)75 (392)76 (464)78 (471) 
Low costhigh admit     
Surgical MS‐DRG11 (35)17 (44)13 (40)13 (54)0.90
Medical MS‐DRG89 (277)83 (219)87 (269)87 (377) 

DISCUSSION

In this study, we found that only half of high cost individuals were also high admit. Further categorizing high users into high costlow admit versus high costhigh admit versus low costhigh admit identified distinct patterns between each group. High costhigh admit individuals were more likely to be hospitalized for medical conditions, whereas high costlow admit individuals were more likely to be hospitalized for surgical conditions. CMS core conditions accounted for a low proportion of overall hospitalizations across all groups.

Our findings suggest several distinct types of high users with different clinical characteristics, utilization, and cost patterns. From a hospital perspective, one implication is that a multifaceted approach to cost containment, rather than the one‐size‐fits‐all strategy of reducing hospitalizations, may be more effective in reducing costs. For example, our findings show that high costlow admit individuals have a disproportionate number of hospitalizations for surgical conditions, longer LOS, and more ICU days. Costs incurred by this subgroup may be more responsive to in‐hospital interventions aimed at reducing procedural costs, LOS, unnecessary use of the ICU, and minimizing postoperative infections and complications rather than to a care management approach.

In contrast, care management strategies such as improving postdischarge care and chronic disease management, which aim to achieve cost savings through reducing hospitalizations, may be more effective in reducing costs among high costhigh admit individuals, who have a high proportion of hospitalizations for medical conditions and the highest proportion of 30‐day readmissions. Such strategies may also be important in optimizing the quality of care for low costhigh admit individuals, who have the highest proportion of medical hospitalizations among all high users, though the potential for cost savings may be more limited in this subgroup.

Our results suggest that current hospital‐based approachesdriven by readmissions penalties for CMS core conditionsmay have less than the expected impact on costs. For example, although high costhigh admit individuals had the highest proportion of 30‐day readmissions, readmissions specifically for CMS core conditions accounted for <1% of costs in this subgroup. Thus, the potential return on an expensive investment in a care management intervention is unclear, given the small number of readmissions for these select conditions. From a broader perspective, the focus on readmissions for CMS core conditions, which overall contribute relatively little to high hospital costs, may not be a comprehensive enough strategy for cost containment. To date, there have been limited policies targeting factors contributing to high hospital costs outside of frequent medical hospitalizations. Medicare's nonpayment policy for treatment of preventable hospital conditions, including surgical site infections, translates prevention of these conditions into cost savings for hospitals.[30] However, this rule has been criticized for not going far enough to drive substantial savings.[31] A new CMS rule authorizes states to identify other provider‐preventable conditions for which Medicaid payment will be prohibited.[32] Future policy efforts should further emphasize a comprehensive, multipronged approach beyond readmissions penalties for select conditions if healthcare cost containment remains a policy priority.

Our results should be interpreted in light of several limitations. First, this was a single‐site study at an academic medical center; the generalizability of our results to other settings is unclear. Our cost data likely reflect local market factors, including the highest wage rates for skilled healthcare labor in the United States.[33] Although the explicit distribution of high user subgroups may be institution‐specific due to variations in our cost structure, we anticipate that the general classification paradigm will be similar in other health systems. Second, we captured utilization and costs only at a single hospital. However, our study population includes only individuals with PCPs at UCSF, and internal data from both Medicare and UCSF's largest private payer show that over 85% of hospitalizations among this population are to UCSF Medical Center. Third, we were able to capture only hospital costs rather than overall healthcare costs. Given that hospital costs account for the single largest category of total national health costs,[4] we expect that future studies examining total health costs will show similar findings. Fourth, our data did not include measures of health status, socioeconomic status, housing, or mental health comorbidities to permit an analysis of these factors, which have been previously related to frequent hospitalizations and high costs.[34, 35, 36, 37, 38, 39] Fifth, due to resource constraints, we were unable to conduct a longitudinal analysis to examine the extent to which individuals are consistently high users over time. Previous studies have described that 20% to 30% of individuals are consistently high users; the remainder have discrete periods of high utilization.[34, 40] This may be an important consideration in the design of future interventions.

Finally, our analysis was limited to individuals with a PCP to allow identification of an accessible cohort for care management. Thus, we did not capture individuals without a PCP and individuals who died during the study period, because these individuals no longer had an assigned PCP following death. Although this approach is consistent with that of many care management programs,[19] these populations are likely to incur higher than average utilization and healthcare costs, and represent important areas for future investigation.

In summary, our study identifies three types of high‐user populations that differ in the proportion of costs attributable to frequent hospitalizations, clinical conditions associated with hospital use, and frequency of 30‐day readmissions. Stratifying high users by both costs and hospitalizations may help identify tailored strategies to more effectively reduce costs and utilization.

Acknowledgments

The authors acknowledge Diana Patterson, Leanna Zaporozhets, and Andre Devito for their assistance in data collection.

Disclosures: Dr. Nguyen had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Dr. Nguyen's work on this project was completed as a primary care research fellow at the University of California, San Francisco funded by a federal training grant from the National Research Service Award (NRSA T32HP19025‐07‐00).

References
  1. Martin A, Lassman D, Whittle L, Catlin A; National Health Expenditure Accounts Team. Recession contributes to slowest annual rate of increase in health spending in five decades. Health Aff (Millwood). 2011;30(1):1122.
  2. Martin AB, Lassman D, Washington B, Catlin A. Growth in US health spending remained slow in 2010; health share of gross domestic product was unchanged from 2009. Health Aff (Millwood). 2012;31(1):208219.
  3. Hartman M, Martin AB, Benson J, Catlin A; the National Health Expenditure Accounts Team. National health spending in 2011: overall growth remains low, but some payers and services show signs of acceleration. Health Aff (Millwood). 2013;32(1):8799.
  4. Centers for Medicare and Medicaid Services; Office of the Actuary; National Health Statistics Group. National healthcare expenditures data. 2012. Available at: https://www.cms.gov/Research‐Statistics‐Data‐and‐Systems/Statistics‐Trends‐and‐Reports/NationalHealthExpendData/downloads/tables.pdf.
  5. Medicare Payment Advisory Commission. Report to the Congress: Promoting Greater Efficiency in Medicare. Washington, DC: Medicare Payment Advisory Commission; 2007.
  6. Coleman EA, Parry C, Chalmers S, Min SJ. The care transitions intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):18221828.
  7. Jack BW, Chetty VK, Anthony D, et al. A reengineered hospital discharge program to decrease rehospitalization: a randomized trial. Ann Intern Med. 2009;150(3):178187.
  8. Schall M, Coleman E, Rutherford P, Taylor J. How‐to Guide: Improving Transitions from the Hospital to the Clinical Office Practice to Reduce Avoidable Rehospitalizations. Cambridge, MA: Institute for Healthcare Improvement; 2012.
  9. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow‐up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613620.
  10. Zook CJ, Moore FD. High‐cost users of medical care. N Engl J Med. 1980;302(18):9961002.
  11. Schroeder SA, Showstack JA, Roberts HE. Frequency and clinical description of high‐cost patients in 17 acute‐care hospitals. N Engl J Med. 1979;300(23):13061309.
  12. Althaus F, Paroz S, Hugli O, et al. Effectiveness of interventions targeting frequent users of emergency departments: a systematic review. Ann Emerg Med. 2011;58(1):4152.e42.
  13. Forrest CB, Lemke KW, Bodycombe DP, Weiner JP. Medication, diagnostic, and cost information as predictors of high‐risk patients in need of care management. Am J Manag Care. 2009;15(1):4148.
  14. Strunk BC, Ginsburg PB. Tracking health care costs: trends stabilize but remain high in 2002. Health Aff (Millwood). 2003;Suppl Web Exclusives:W3266–274.
  15. Newhouse JP. An iconoclastic view of health cost containment. Health Aff (Millwood). 1993;12(suppl):152171.
  16. Nan‐Ping Y, Yi‐Hui L, Chi‐Yu C, et al. Comparisons of medical utilizations and categorical diagnoses of emergency visits between the elderly with catastrophic illness certificates and those without. BMC Health Serv Res. 2013;13:152.
  17. Ash AS, Zhao Y, Ellis RP, Schlein Kramer M. Finding future high‐cost cases: comparing prior cost versus diagnosis‐based methods. Health Serv Res. 2001;36(6 pt 2):194206.
  18. Cohen JW, Krauss NA. Spending and service use among people with the fifteen most costly medical conditions, 1997. Health Aff (Millwood). 2003;22(2):129138.
  19. Nelson L. Lessons from Medicare's Demonstration Projects on Disease Management and Care Coordination. Washington, D.C.: Congressional Budget Office; 2012.
  20. Gawande A. The hot spotters. The New Yorker. January 24, 2011.
  21. Freshman B, Rubino LG, Reid Chassiakos Y. Collaboration Across the Disciplines in Health Care. Burlington, MA: Jones and Bartlett; 2010.
  22. Green SR, Singh V, O'Byrne W. Hope for New Jersey's city hospitals: the Camden Initiative. Perspect Health Inf Manag. 2010:7:1d.
  23. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital‐initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433440.
  24. Agency for Healthcare Research and Quality. The concentration and persistence in the level of health expenditures over time: estimates for the U.S. population, 2008–2009. 2012. Available at: http://meps.ahrq.gov/mepsweb/data_files/publications/st354/stat354.shtml.
  25. Azoulay A, Doris NM, Filion KB, Caron J, Pilote L, Eisenberg MJ. The use of the transition cost accounting system in health services research. Cost Eff Resour Alloc. 2007;5:11.
  26. Pronovost P, Angus DC. Cost reduction and quality improvement: it takes two to tango. Crit Care Med. 2000;28(2):581583.
  27. Billings J, Dixon J, Mijanovich T, Wennberg D. Case finding for patients at risk of readmission to hospital: development of algorithm to identify high risk patients. BMJ. 2006;333(7563):327.
  28. Kansagara D, Englander H, Salanitro A, et al. Risk prediction models for hospital readmission: a systematic review. JAMA. 2011;306(15):16881698.
  29. Putnam KG, Buist DS, Fishman P, et al. Chronic disease score as a predictor of hospitalization. Epidemiology. 2002;13(3):340346.
  30. Centers for Medicare and Medicaid Services. Hospital‐acquired conditions (HAC) in acute inpatient prospective payment system (IPPS) hospitals fact sheet. 2011. Available at: https://www.cms.gov/Medicare/Medicare‐Fee‐for‐Service‐Payment/HospitalAcqCond/downloads/HACFactsheet.pdf. Accessed August 31, 2013.
  31. McNair PD, Luft HS, Bindman AB. Medicare's policy not to pay for treating hospital‐acquired conditions: the impact. Health Aff (Millwood). 2009;28(5):14851493.
  32. Department of Health and Human Services, Centers for Medicare 2013.
  33. Freeborn DK, Pope CR, Mullooly JP, McFarland BH. Consistently high users of medical care among the elderly. Med Care. 1990;28(6):527540.
  34. McFarland BH, Freeborn DK, Mullooly JP, Pope CR. Utilization patterns among long‐term enrollees in a prepaid group practice health maintenance organization. Med Care. 1985;23(11):12211233.
  35. Waxman HM, Carner EA, Blum A. Depressive symptoms and health service utilization among the community elderly. J Am Geriatr Soc. 1983;31(7):417420.
  36. Kuo RN, Lai MS. The influence of socio‐economic status and multimorbidity patterns on healthcare costs: a six‐year follow‐up under a universal healthcare system. Int J Equity Health. 2013;12(1):69.
  37. Reid KW, Vittinghoff E, Kushel MB. Association between the level of housing instability, economic standing and health care access: a meta‐regression. J Health Care Poor Underserved. 2008;19(4):12121228.
  38. Lemstra M, Mackenbach J, Neudorf C, Nannapaneni U. High health care utilization and costs associated with lower socio‐economic status: results from a linked dataset. Can J Public Health. 2009;100(3):180183.
  39. McCall N, Wai HS. An analysis of the use of Medicare services by the continuously enrolled aged. Med Care. 1983;21(6):567585.
References
  1. Martin A, Lassman D, Whittle L, Catlin A; National Health Expenditure Accounts Team. Recession contributes to slowest annual rate of increase in health spending in five decades. Health Aff (Millwood). 2011;30(1):1122.
  2. Martin AB, Lassman D, Washington B, Catlin A. Growth in US health spending remained slow in 2010; health share of gross domestic product was unchanged from 2009. Health Aff (Millwood). 2012;31(1):208219.
  3. Hartman M, Martin AB, Benson J, Catlin A; the National Health Expenditure Accounts Team. National health spending in 2011: overall growth remains low, but some payers and services show signs of acceleration. Health Aff (Millwood). 2013;32(1):8799.
  4. Centers for Medicare and Medicaid Services; Office of the Actuary; National Health Statistics Group. National healthcare expenditures data. 2012. Available at: https://www.cms.gov/Research‐Statistics‐Data‐and‐Systems/Statistics‐Trends‐and‐Reports/NationalHealthExpendData/downloads/tables.pdf.
  5. Medicare Payment Advisory Commission. Report to the Congress: Promoting Greater Efficiency in Medicare. Washington, DC: Medicare Payment Advisory Commission; 2007.
  6. Coleman EA, Parry C, Chalmers S, Min SJ. The care transitions intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):18221828.
  7. Jack BW, Chetty VK, Anthony D, et al. A reengineered hospital discharge program to decrease rehospitalization: a randomized trial. Ann Intern Med. 2009;150(3):178187.
  8. Schall M, Coleman E, Rutherford P, Taylor J. How‐to Guide: Improving Transitions from the Hospital to the Clinical Office Practice to Reduce Avoidable Rehospitalizations. Cambridge, MA: Institute for Healthcare Improvement; 2012.
  9. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow‐up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613620.
  10. Zook CJ, Moore FD. High‐cost users of medical care. N Engl J Med. 1980;302(18):9961002.
  11. Schroeder SA, Showstack JA, Roberts HE. Frequency and clinical description of high‐cost patients in 17 acute‐care hospitals. N Engl J Med. 1979;300(23):13061309.
  12. Althaus F, Paroz S, Hugli O, et al. Effectiveness of interventions targeting frequent users of emergency departments: a systematic review. Ann Emerg Med. 2011;58(1):4152.e42.
  13. Forrest CB, Lemke KW, Bodycombe DP, Weiner JP. Medication, diagnostic, and cost information as predictors of high‐risk patients in need of care management. Am J Manag Care. 2009;15(1):4148.
  14. Strunk BC, Ginsburg PB. Tracking health care costs: trends stabilize but remain high in 2002. Health Aff (Millwood). 2003;Suppl Web Exclusives:W3266–274.
  15. Newhouse JP. An iconoclastic view of health cost containment. Health Aff (Millwood). 1993;12(suppl):152171.
  16. Nan‐Ping Y, Yi‐Hui L, Chi‐Yu C, et al. Comparisons of medical utilizations and categorical diagnoses of emergency visits between the elderly with catastrophic illness certificates and those without. BMC Health Serv Res. 2013;13:152.
  17. Ash AS, Zhao Y, Ellis RP, Schlein Kramer M. Finding future high‐cost cases: comparing prior cost versus diagnosis‐based methods. Health Serv Res. 2001;36(6 pt 2):194206.
  18. Cohen JW, Krauss NA. Spending and service use among people with the fifteen most costly medical conditions, 1997. Health Aff (Millwood). 2003;22(2):129138.
  19. Nelson L. Lessons from Medicare's Demonstration Projects on Disease Management and Care Coordination. Washington, D.C.: Congressional Budget Office; 2012.
  20. Gawande A. The hot spotters. The New Yorker. January 24, 2011.
  21. Freshman B, Rubino LG, Reid Chassiakos Y. Collaboration Across the Disciplines in Health Care. Burlington, MA: Jones and Bartlett; 2010.
  22. Green SR, Singh V, O'Byrne W. Hope for New Jersey's city hospitals: the Camden Initiative. Perspect Health Inf Manag. 2010:7:1d.
  23. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital‐initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433440.
  24. Agency for Healthcare Research and Quality. The concentration and persistence in the level of health expenditures over time: estimates for the U.S. population, 2008–2009. 2012. Available at: http://meps.ahrq.gov/mepsweb/data_files/publications/st354/stat354.shtml.
  25. Azoulay A, Doris NM, Filion KB, Caron J, Pilote L, Eisenberg MJ. The use of the transition cost accounting system in health services research. Cost Eff Resour Alloc. 2007;5:11.
  26. Pronovost P, Angus DC. Cost reduction and quality improvement: it takes two to tango. Crit Care Med. 2000;28(2):581583.
  27. Billings J, Dixon J, Mijanovich T, Wennberg D. Case finding for patients at risk of readmission to hospital: development of algorithm to identify high risk patients. BMJ. 2006;333(7563):327.
  28. Kansagara D, Englander H, Salanitro A, et al. Risk prediction models for hospital readmission: a systematic review. JAMA. 2011;306(15):16881698.
  29. Putnam KG, Buist DS, Fishman P, et al. Chronic disease score as a predictor of hospitalization. Epidemiology. 2002;13(3):340346.
  30. Centers for Medicare and Medicaid Services. Hospital‐acquired conditions (HAC) in acute inpatient prospective payment system (IPPS) hospitals fact sheet. 2011. Available at: https://www.cms.gov/Medicare/Medicare‐Fee‐for‐Service‐Payment/HospitalAcqCond/downloads/HACFactsheet.pdf. Accessed August 31, 2013.
  31. McNair PD, Luft HS, Bindman AB. Medicare's policy not to pay for treating hospital‐acquired conditions: the impact. Health Aff (Millwood). 2009;28(5):14851493.
  32. Department of Health and Human Services, Centers for Medicare 2013.
  33. Freeborn DK, Pope CR, Mullooly JP, McFarland BH. Consistently high users of medical care among the elderly. Med Care. 1990;28(6):527540.
  34. McFarland BH, Freeborn DK, Mullooly JP, Pope CR. Utilization patterns among long‐term enrollees in a prepaid group practice health maintenance organization. Med Care. 1985;23(11):12211233.
  35. Waxman HM, Carner EA, Blum A. Depressive symptoms and health service utilization among the community elderly. J Am Geriatr Soc. 1983;31(7):417420.
  36. Kuo RN, Lai MS. The influence of socio‐economic status and multimorbidity patterns on healthcare costs: a six‐year follow‐up under a universal healthcare system. Int J Equity Health. 2013;12(1):69.
  37. Reid KW, Vittinghoff E, Kushel MB. Association between the level of housing instability, economic standing and health care access: a meta‐regression. J Health Care Poor Underserved. 2008;19(4):12121228.
  38. Lemstra M, Mackenbach J, Neudorf C, Nannapaneni U. High health care utilization and costs associated with lower socio‐economic status: results from a linked dataset. Can J Public Health. 2009;100(3):180183.
  39. McCall N, Wai HS. An analysis of the use of Medicare services by the continuously enrolled aged. Med Care. 1983;21(6):567585.
Issue
Journal of Hospital Medicine - 8(12)
Issue
Journal of Hospital Medicine - 8(12)
Page Number
665-671
Page Number
665-671
Article Type
Display Headline
What's cost got to do with it? Association between hospital costs and frequency of admissions among “high users” of hospital care
Display Headline
What's cost got to do with it? Association between hospital costs and frequency of admissions among “high users” of hospital care
Sections
Article Source

© 2013 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Oanh Kieu Nguyen, MD, Division of General Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Mail Code 9169, Dallas, TX 75390‐9169; Telephone: 214–648‐3135; Fax: 214–648‐3232; E‐mail: [email protected]
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Media Files

PSA screening: The USPSTF got it right

Article Type
Changed
Mon, 01/14/2019 - 13:44
Display Headline
PSA screening: The USPSTF got it right

In his book, How We Do Harm: A Doctor Breaks Ranks About Being Sick in America, Otis Brawley1 writes, “I believe that a man should know what we know, what we don’t know, and what we believe about prostate cancer. I have been concerned that many patients and physicians have confused what is believed with what is known.” I agree.

Common sense is what we believe. Does common sense trump science? Did the US Preventive Services Task Force (USPSTF) get it wrong? I don’t think so.

The USPSTF bases its recommendations on an explicit assessment of the science that informs us of the benefits and harms of a preventive service, and a judgment about the magnitude of net benefit.

So what do we know about the benefits of prostate cancer screening? When attempting to answer the question of whether an intervention is beneficial, there is a hierarchy of evidence, from most likely to be wrong to most likely to be right. Relying on our personal stories is the former; relying on well-conducted randomized trials is the latter.

We do an enormous disservice to our patients if we pretend that this is just a blood test. Men will get biopsies and there will be complications.In the multicenter Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial2 conducted in the United States, there was a nonsignificant increase in prostate-cancer mortality in the screening group, while the European Randomized Study of Screening for Prostate Cancer (ERSPC)3 trial showed a statistically significant absolute reduction of 0.10 prostate-cancer deaths per 1000 person-years after a median follow-up of 11 years. In the ERSPC trial, all-cause mortality was 19.1% in the screened group and 19.3% in the control group, a difference that was not significant. What we know is that after 10 years, even with aggressive treatment of 80% to 90% of screen-detected cancers, very few, if any, men will have lived longer because they were screened.

What don’t we know about the benefits? We don’t know whether following screened and nonscreened men for 15 or 20 years or longer will demonstrate a larger difference in mortality. Competing causes of mortality make it progressively less likely that men who are screened will actually live longer. The average age of death from prostate cancer is 80 years, and 70% of all deaths occur after age 75.4 Contrast those statistics to breast cancer, for which the average age of death is 68 years and 63% of all deaths occur before age 75.5

What do we believe about the benefits? Some certainly believe the trials must be wrong; common sense tells us that early detection and treatment must provide more benefit than what the evidence has shown. Common sense tells us that the decline in prostate cancer mortality over the past 2 decades must be due to screening, although the ERSPC results clearly show that neither the magnitude nor the timing of the decline can be attributed to screening.

What do we know—and not know—about the harms?

We know that much of the suffering from prostate cancer is a consequence of the diagnosis and management of the disease, rather than the disease itself. Complications of both diagnosis and treatment of prostate cancer are frequent and serious.6 We also know that many screen-detected cancers would never become apparent in a man’s lifetime without screening.

We don’t know the precise magnitude of overdiagnosis, although all estimates suggest it is substantial. In the ERSPC trial, 9.6% of the screened group received a prostate cancer diagnosis, vs 6.0% of the control group—a 60% increase in the rate of diagnosis. The recently published long-term results from the Prostate Cancer Prevention Trial7 are enlightening. Finasteride reduced the incidence of screen-detected cancers by 30%, with no impact on all-cause mortality at 18 years. If those screen-detected cancers had been a significant threat to health, then after 18 years we would have expected some mortality benefit from finasteride.

What do we believe about the harms of screening?
We believe that by being more conservative about who gets treated, we shift the balance of benefits and harms of screening. There is no question that reducing the burden of overdiagnosis and overtreatment would provide a welcome reduction in the harms. But can we do it?8

In the United States 90% of men found to have prostate cancer are treated (including about 75% of men with low-risk cancers).6 And although we hope to be able to reduce harms without changing benefits, we do not know what impact more conservative management of screen-detected cancers would have on the already small effect of screening on prostate cancer mortality.

 

 

So what is the balance of benefit and harms? Should we make that judgment on what we know, or on what we believe?

Science trumps common sense. For every 1000 men screened, at most, one will avoid a prostate cancer death at 10 years. But 30 to 40 will have erectile dysfunction, urinary incontinence, or both due to treatment, 2 men will experience a serious cardiovascular event, one will have a venous thromboembolic event, and one in 3000 screened will die from complications of surgical treatment.6

The USPSTF concluded that the benefits of PSA screening do not outweigh the harms, but acknowledged that shared decision making is still appropriate when a physician feels obliged to offer the test or a patient requests it.

What does shared decision making look like? Just offering screening and answering any questions is not good enough. We do an enormous disservice to our patients if we pretend that this is just a blood test and that we can decide later what to do with the information. Men will get biopsies and there will be complications. Cancer will be detected, and men will be treated, many unnecessarily.

Routine screening for prostate cancer in the absence of a truly formed decision is unacceptable.We need to tell our patients that the likelihood of avoiding a prostate cancer death over 10 years as a result of regular PSA screening is at most very small, and that many more men will suffer the harms of unnecessary treatment than will benefit. A few will die prematurely as a result of the complications of treating a screen-detected cancer.

If, with this knowledge, a patient places a higher value on the possibility of avoiding a prostate cancer death than he does on the known harms of diagnosis and treatment, he can still decide to be screened. He has made an informed decision. However, routine screening for prostate cancer in the absence of a truly informed decision is unacceptable.

References

1. Brawley OW, Goldberg P. How We Do Harm: A Doctor Breaks Ranks About Being Sick in America. NY: St. Martin's Press; 2011.

2. Andriole GL, Crawford ED, Grub III RL, et al. Prostate cancer screening in the randomized prostate, lung, colorectal, and ovarian cancer screening trial. J Natl Cancer Inst. 2012;104:125-132.

3. Schröder FH, et al; ERSPC investigators. Prostate cancer mortality at 11 years of follow-up. N Engl J Med. 2012;366:981-990.

4. SEER Stat Fact Sheets: Prostate. Surveillance, Epidemiology and End Results (SEER) Program Web site. 2012. Available at: http://seer.cancer.gov/statfacts/html/prost.html. Accessed August 28, 2013.

5. SEER Stat Fact Sheets: Breast. Surveillance, Epidemiology and End Results (SEER) Program Web site. 2012. Available at: http://seer.cancer.gov/statfacts/html/breast.html. Accessed August 28, 2013.

6. Chou R, Crosswell, JM, Dana T, et al. Screening for prostate cancer: A review of the evidence for the US Preventive Services Task Force. Ann Intern Med. 2011;155:762-771.

7. Thompson IM Jr, Goodman PJ, Tangen CM, et al. Long-term survival of participants in the Prostate Cancer Prevention Trial. N Engl J Med. 2013;369:603-610.

8. Sartor AO. Surveillance for prostate cancer: are the proceduralists running amok? Oncology (Williston Park). 2013;27:523, 589.

Article PDF
Author and Disclosure Information

Michael LeFevre, MD, MSPH
University of Missouri School of Medicine, Columbia

The author reported no potential conflict of interest relevant to this article.

Issue
The Journal of Family Practice - 62(11)
Publications
Topics
Page Number
616, 618
Legacy Keywords
Michael LeFevre; MD; MSPH; USPSTF; US Preventive Services Task Force; biopsies; PSA screening; prostate specific antigen
Sections
Author and Disclosure Information

Michael LeFevre, MD, MSPH
University of Missouri School of Medicine, Columbia

The author reported no potential conflict of interest relevant to this article.

Author and Disclosure Information

Michael LeFevre, MD, MSPH
University of Missouri School of Medicine, Columbia

The author reported no potential conflict of interest relevant to this article.

Article PDF
Article PDF

In his book, How We Do Harm: A Doctor Breaks Ranks About Being Sick in America, Otis Brawley1 writes, “I believe that a man should know what we know, what we don’t know, and what we believe about prostate cancer. I have been concerned that many patients and physicians have confused what is believed with what is known.” I agree.

Common sense is what we believe. Does common sense trump science? Did the US Preventive Services Task Force (USPSTF) get it wrong? I don’t think so.

The USPSTF bases its recommendations on an explicit assessment of the science that informs us of the benefits and harms of a preventive service, and a judgment about the magnitude of net benefit.

So what do we know about the benefits of prostate cancer screening? When attempting to answer the question of whether an intervention is beneficial, there is a hierarchy of evidence, from most likely to be wrong to most likely to be right. Relying on our personal stories is the former; relying on well-conducted randomized trials is the latter.

We do an enormous disservice to our patients if we pretend that this is just a blood test. Men will get biopsies and there will be complications.In the multicenter Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial2 conducted in the United States, there was a nonsignificant increase in prostate-cancer mortality in the screening group, while the European Randomized Study of Screening for Prostate Cancer (ERSPC)3 trial showed a statistically significant absolute reduction of 0.10 prostate-cancer deaths per 1000 person-years after a median follow-up of 11 years. In the ERSPC trial, all-cause mortality was 19.1% in the screened group and 19.3% in the control group, a difference that was not significant. What we know is that after 10 years, even with aggressive treatment of 80% to 90% of screen-detected cancers, very few, if any, men will have lived longer because they were screened.

What don’t we know about the benefits? We don’t know whether following screened and nonscreened men for 15 or 20 years or longer will demonstrate a larger difference in mortality. Competing causes of mortality make it progressively less likely that men who are screened will actually live longer. The average age of death from prostate cancer is 80 years, and 70% of all deaths occur after age 75.4 Contrast those statistics to breast cancer, for which the average age of death is 68 years and 63% of all deaths occur before age 75.5

What do we believe about the benefits? Some certainly believe the trials must be wrong; common sense tells us that early detection and treatment must provide more benefit than what the evidence has shown. Common sense tells us that the decline in prostate cancer mortality over the past 2 decades must be due to screening, although the ERSPC results clearly show that neither the magnitude nor the timing of the decline can be attributed to screening.

What do we know—and not know—about the harms?

We know that much of the suffering from prostate cancer is a consequence of the diagnosis and management of the disease, rather than the disease itself. Complications of both diagnosis and treatment of prostate cancer are frequent and serious.6 We also know that many screen-detected cancers would never become apparent in a man’s lifetime without screening.

We don’t know the precise magnitude of overdiagnosis, although all estimates suggest it is substantial. In the ERSPC trial, 9.6% of the screened group received a prostate cancer diagnosis, vs 6.0% of the control group—a 60% increase in the rate of diagnosis. The recently published long-term results from the Prostate Cancer Prevention Trial7 are enlightening. Finasteride reduced the incidence of screen-detected cancers by 30%, with no impact on all-cause mortality at 18 years. If those screen-detected cancers had been a significant threat to health, then after 18 years we would have expected some mortality benefit from finasteride.

What do we believe about the harms of screening?
We believe that by being more conservative about who gets treated, we shift the balance of benefits and harms of screening. There is no question that reducing the burden of overdiagnosis and overtreatment would provide a welcome reduction in the harms. But can we do it?8

In the United States 90% of men found to have prostate cancer are treated (including about 75% of men with low-risk cancers).6 And although we hope to be able to reduce harms without changing benefits, we do not know what impact more conservative management of screen-detected cancers would have on the already small effect of screening on prostate cancer mortality.

 

 

So what is the balance of benefit and harms? Should we make that judgment on what we know, or on what we believe?

Science trumps common sense. For every 1000 men screened, at most, one will avoid a prostate cancer death at 10 years. But 30 to 40 will have erectile dysfunction, urinary incontinence, or both due to treatment, 2 men will experience a serious cardiovascular event, one will have a venous thromboembolic event, and one in 3000 screened will die from complications of surgical treatment.6

The USPSTF concluded that the benefits of PSA screening do not outweigh the harms, but acknowledged that shared decision making is still appropriate when a physician feels obliged to offer the test or a patient requests it.

What does shared decision making look like? Just offering screening and answering any questions is not good enough. We do an enormous disservice to our patients if we pretend that this is just a blood test and that we can decide later what to do with the information. Men will get biopsies and there will be complications. Cancer will be detected, and men will be treated, many unnecessarily.

Routine screening for prostate cancer in the absence of a truly formed decision is unacceptable.We need to tell our patients that the likelihood of avoiding a prostate cancer death over 10 years as a result of regular PSA screening is at most very small, and that many more men will suffer the harms of unnecessary treatment than will benefit. A few will die prematurely as a result of the complications of treating a screen-detected cancer.

If, with this knowledge, a patient places a higher value on the possibility of avoiding a prostate cancer death than he does on the known harms of diagnosis and treatment, he can still decide to be screened. He has made an informed decision. However, routine screening for prostate cancer in the absence of a truly informed decision is unacceptable.

In his book, How We Do Harm: A Doctor Breaks Ranks About Being Sick in America, Otis Brawley1 writes, “I believe that a man should know what we know, what we don’t know, and what we believe about prostate cancer. I have been concerned that many patients and physicians have confused what is believed with what is known.” I agree.

Common sense is what we believe. Does common sense trump science? Did the US Preventive Services Task Force (USPSTF) get it wrong? I don’t think so.

The USPSTF bases its recommendations on an explicit assessment of the science that informs us of the benefits and harms of a preventive service, and a judgment about the magnitude of net benefit.

So what do we know about the benefits of prostate cancer screening? When attempting to answer the question of whether an intervention is beneficial, there is a hierarchy of evidence, from most likely to be wrong to most likely to be right. Relying on our personal stories is the former; relying on well-conducted randomized trials is the latter.

We do an enormous disservice to our patients if we pretend that this is just a blood test. Men will get biopsies and there will be complications.In the multicenter Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial2 conducted in the United States, there was a nonsignificant increase in prostate-cancer mortality in the screening group, while the European Randomized Study of Screening for Prostate Cancer (ERSPC)3 trial showed a statistically significant absolute reduction of 0.10 prostate-cancer deaths per 1000 person-years after a median follow-up of 11 years. In the ERSPC trial, all-cause mortality was 19.1% in the screened group and 19.3% in the control group, a difference that was not significant. What we know is that after 10 years, even with aggressive treatment of 80% to 90% of screen-detected cancers, very few, if any, men will have lived longer because they were screened.

What don’t we know about the benefits? We don’t know whether following screened and nonscreened men for 15 or 20 years or longer will demonstrate a larger difference in mortality. Competing causes of mortality make it progressively less likely that men who are screened will actually live longer. The average age of death from prostate cancer is 80 years, and 70% of all deaths occur after age 75.4 Contrast those statistics to breast cancer, for which the average age of death is 68 years and 63% of all deaths occur before age 75.5

What do we believe about the benefits? Some certainly believe the trials must be wrong; common sense tells us that early detection and treatment must provide more benefit than what the evidence has shown. Common sense tells us that the decline in prostate cancer mortality over the past 2 decades must be due to screening, although the ERSPC results clearly show that neither the magnitude nor the timing of the decline can be attributed to screening.

What do we know—and not know—about the harms?

We know that much of the suffering from prostate cancer is a consequence of the diagnosis and management of the disease, rather than the disease itself. Complications of both diagnosis and treatment of prostate cancer are frequent and serious.6 We also know that many screen-detected cancers would never become apparent in a man’s lifetime without screening.

We don’t know the precise magnitude of overdiagnosis, although all estimates suggest it is substantial. In the ERSPC trial, 9.6% of the screened group received a prostate cancer diagnosis, vs 6.0% of the control group—a 60% increase in the rate of diagnosis. The recently published long-term results from the Prostate Cancer Prevention Trial7 are enlightening. Finasteride reduced the incidence of screen-detected cancers by 30%, with no impact on all-cause mortality at 18 years. If those screen-detected cancers had been a significant threat to health, then after 18 years we would have expected some mortality benefit from finasteride.

What do we believe about the harms of screening?
We believe that by being more conservative about who gets treated, we shift the balance of benefits and harms of screening. There is no question that reducing the burden of overdiagnosis and overtreatment would provide a welcome reduction in the harms. But can we do it?8

In the United States 90% of men found to have prostate cancer are treated (including about 75% of men with low-risk cancers).6 And although we hope to be able to reduce harms without changing benefits, we do not know what impact more conservative management of screen-detected cancers would have on the already small effect of screening on prostate cancer mortality.

 

 

So what is the balance of benefit and harms? Should we make that judgment on what we know, or on what we believe?

Science trumps common sense. For every 1000 men screened, at most, one will avoid a prostate cancer death at 10 years. But 30 to 40 will have erectile dysfunction, urinary incontinence, or both due to treatment, 2 men will experience a serious cardiovascular event, one will have a venous thromboembolic event, and one in 3000 screened will die from complications of surgical treatment.6

The USPSTF concluded that the benefits of PSA screening do not outweigh the harms, but acknowledged that shared decision making is still appropriate when a physician feels obliged to offer the test or a patient requests it.

What does shared decision making look like? Just offering screening and answering any questions is not good enough. We do an enormous disservice to our patients if we pretend that this is just a blood test and that we can decide later what to do with the information. Men will get biopsies and there will be complications. Cancer will be detected, and men will be treated, many unnecessarily.

Routine screening for prostate cancer in the absence of a truly formed decision is unacceptable.We need to tell our patients that the likelihood of avoiding a prostate cancer death over 10 years as a result of regular PSA screening is at most very small, and that many more men will suffer the harms of unnecessary treatment than will benefit. A few will die prematurely as a result of the complications of treating a screen-detected cancer.

If, with this knowledge, a patient places a higher value on the possibility of avoiding a prostate cancer death than he does on the known harms of diagnosis and treatment, he can still decide to be screened. He has made an informed decision. However, routine screening for prostate cancer in the absence of a truly informed decision is unacceptable.

References

1. Brawley OW, Goldberg P. How We Do Harm: A Doctor Breaks Ranks About Being Sick in America. NY: St. Martin's Press; 2011.

2. Andriole GL, Crawford ED, Grub III RL, et al. Prostate cancer screening in the randomized prostate, lung, colorectal, and ovarian cancer screening trial. J Natl Cancer Inst. 2012;104:125-132.

3. Schröder FH, et al; ERSPC investigators. Prostate cancer mortality at 11 years of follow-up. N Engl J Med. 2012;366:981-990.

4. SEER Stat Fact Sheets: Prostate. Surveillance, Epidemiology and End Results (SEER) Program Web site. 2012. Available at: http://seer.cancer.gov/statfacts/html/prost.html. Accessed August 28, 2013.

5. SEER Stat Fact Sheets: Breast. Surveillance, Epidemiology and End Results (SEER) Program Web site. 2012. Available at: http://seer.cancer.gov/statfacts/html/breast.html. Accessed August 28, 2013.

6. Chou R, Crosswell, JM, Dana T, et al. Screening for prostate cancer: A review of the evidence for the US Preventive Services Task Force. Ann Intern Med. 2011;155:762-771.

7. Thompson IM Jr, Goodman PJ, Tangen CM, et al. Long-term survival of participants in the Prostate Cancer Prevention Trial. N Engl J Med. 2013;369:603-610.

8. Sartor AO. Surveillance for prostate cancer: are the proceduralists running amok? Oncology (Williston Park). 2013;27:523, 589.

References

1. Brawley OW, Goldberg P. How We Do Harm: A Doctor Breaks Ranks About Being Sick in America. NY: St. Martin's Press; 2011.

2. Andriole GL, Crawford ED, Grub III RL, et al. Prostate cancer screening in the randomized prostate, lung, colorectal, and ovarian cancer screening trial. J Natl Cancer Inst. 2012;104:125-132.

3. Schröder FH, et al; ERSPC investigators. Prostate cancer mortality at 11 years of follow-up. N Engl J Med. 2012;366:981-990.

4. SEER Stat Fact Sheets: Prostate. Surveillance, Epidemiology and End Results (SEER) Program Web site. 2012. Available at: http://seer.cancer.gov/statfacts/html/prost.html. Accessed August 28, 2013.

5. SEER Stat Fact Sheets: Breast. Surveillance, Epidemiology and End Results (SEER) Program Web site. 2012. Available at: http://seer.cancer.gov/statfacts/html/breast.html. Accessed August 28, 2013.

6. Chou R, Crosswell, JM, Dana T, et al. Screening for prostate cancer: A review of the evidence for the US Preventive Services Task Force. Ann Intern Med. 2011;155:762-771.

7. Thompson IM Jr, Goodman PJ, Tangen CM, et al. Long-term survival of participants in the Prostate Cancer Prevention Trial. N Engl J Med. 2013;369:603-610.

8. Sartor AO. Surveillance for prostate cancer: are the proceduralists running amok? Oncology (Williston Park). 2013;27:523, 589.

Issue
The Journal of Family Practice - 62(11)
Issue
The Journal of Family Practice - 62(11)
Page Number
616, 618
Page Number
616, 618
Publications
Publications
Topics
Article Type
Display Headline
PSA screening: The USPSTF got it right
Display Headline
PSA screening: The USPSTF got it right
Legacy Keywords
Michael LeFevre; MD; MSPH; USPSTF; US Preventive Services Task Force; biopsies; PSA screening; prostate specific antigen
Legacy Keywords
Michael LeFevre; MD; MSPH; USPSTF; US Preventive Services Task Force; biopsies; PSA screening; prostate specific antigen
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media

Update on pelvic floor dysfunction: Focus on urinary incontinence

Article Type
Changed
Tue, 08/28/2018 - 11:03
Display Headline
Update on pelvic floor dysfunction: Focus on urinary incontinence

Urinary incontinence (UI) affects almost half of all women in the United States.1,2 Estimates suggest that the prevalence of UI gradually rises during young adult life, comes to a broad plateau in middle age, and then steadily increases from that plateau after age 65. Therefore, over the next 40 years, as the elderly population expands in size, the number of women affected by UI will significantly grow.3

For patients with UI, a multitude of therapeutic options are available. Which option is the best for your patient? In this article, we aim to answer that question by interpreting the results of four randomized trials, each of which directly compare two available treatment options. The first study examines patients with stress urinary incontinence (SUI), comparing the patients’ subjective improvement in urinary leakage and bladder function at 12 months after randomization to treatment with physiotherapy or midurethral sling surgery.

The three other trials examine patients with overactive bladder (OAB) and urgency urinary incontinence (UUI). Each trial directly compares the use of anticholinergic medications to an alternate treatment modality. Currently, anticholinergic medications and behavioral therapy are the recommended first-line therapies for OAB. Unfortunately, anticholinergic medications have poor patient compliance and significant systemic side effects.4 Caution should be used when considering anticholinergic medications in patients with impaired gastric emptying or a history of urinary retention. They also should be used with caution in elderly patients who are extremely frail. Additionally, clearance from an ophthalmologist must be obtained prior to starting anticholinergic medication in patients with narrow-angle glaucoma.5 Due to poor adherence and potential side effects, there is a growing effort to discover alternative treatment modalities that are safe and effective. Therefore, we chose to examine trials comparing: mirabegron versus tolterodine, percutaneous tibial nerve stimulation versus tolterodine, and onabotulinumtoxinA versus anticholingeric medications.

UI defined
Before discussing treatment options, we want to clarify the main types of UI (FIGURE). UI is defined as the complaint of involuntary loss of urine.  UI can be subdivided into SUI, OAB/UUI, or mixed urinary incontinence.6 While there are other less common genitourinary etiologies that can lead to UI, nongenitourinary etiologies are prevalent and can aggravate existing SUI or OAB (TABLE).

SUI is the complaint of involuntary loss of urine on effort or physical exertion (such as during sporting activities) or on sneezing or coughing. Often, SUI can be diagnosed by patient report alone and surgery can be considered in symptomatic patients who demonstrate cough leakage on physical examination and normal postvoid residual volumes.

UUI is the involuntary loss of urine associated with urgency; it often occurs in the setting of OAB, which is defined as the syndrome of urinary urgency, usually accompanied by frequency and nocturia, with or without UUI, in the absence of urinary tract infection or other obvious pathology (such as neurologic dysfunction, infection, or urologic neoplasm). OAB-dry is present when patients do not have leakage with urgency, but are bothered by urgency, frequency, and/or nocturia. OAB-wet occurs when a patient has urgencyincontinence.

The presence of both SUI and OAB/UUI is known as mixed urinary incontinence. Stress and urgency urinary symptoms often present together. In fact, 10% to 30% of women with stress symptoms are found to have bladder overactivity on subsequent evaluation.2,7 Therefore, it is important to take a good history and consider urodynamic evaluation to confirm the diagnosis of SUI prior to surgery in women with mixed stress and urge symptoms, a history of a previous surgery for incontinence, or when there is a poor correlation of physical examination findings to reported symptoms.

 

Is surgery a first-line option for patients with SUI?

Labrie J, Berghmans BL, Fischer K, et al. Surgery versus physiotherapy for stress urinary incontinence. NEJM. 2013;369(12):1124−1133.

Physiotherapy, including pelvic floor muscle training (“Kegel exercises”), is utilized as a first-line treatment option for women with SUI that carries minimal risk for the patient. Midurethral sling surgery is often recommended if an initial trial of conservative treatment fails.7 Up to 50% of women treated with pelvic floor physiotherapy will ultimately undergo surgery to treat their SUI.8

Related article: Does urodynamic testing before surgery for stress incontinence improve outcomes? G. Willy Davila, MD (Examining the Evidence, December 2012)

Details of the study
This was a randomized, multicenter trial of women aged 35 to 80 years with moderate to severe SUI. After excluding women with previous incontinence surgery and stage 2 or higher pelvic organ prolapse, 460 participants were randomly assigned to undergo either a midurethral sling surgery or physiotherapy (pelvic floor muscle training). The primary outcome was subjective improvement in urinary leakage and bladder function at 12 months, as measured by the Patient Global Impression of Improvement (PGI-I), a 7-point Likert scale ranging from “very much worse” to “very much better.”

 

 

In an intention-to-treat analysis, subjective improvement at 12 months was significantly higher in women randomized to midurethral sling surgery than in women randomized to physiotherapy (91% vs 64%, respectively).

Ten percent of patients had adverse events (AEs); all were related to surgery. The most common AEs were hematoma, vaginal epithelial perforation, and bladder perforation.

Notably, women had the option to cross over to the other treatment modality if they desired. In the physiotherapy group, 49% of women elected to cross over to surgery, while 11% of those who underwent midurethral sling surgery elected to cross over to physiotherapy during the 12-month follow-up period. When analyzing results by treatment received, the investigators found that the proportion of women who reported improvement was significantly lower among women who underwent physiotherapy only (32%), versus sling only (94%), or sling after physiotherapy (91%).

This randomized trial was well-designed and included a variety of treatment centers (university and general hospitals) with interventions performed by experienced surgeons (all of whom had performed at least 20 sling surgeries) and physiotherapists educated specifically in pelvic floor physiotherapy. The study population was limited to patients with moderate to severe SUI as defined by the Sandvik severity index.9 Therefore, these results may not be applicable to patients with milder symptoms, for whom physiotherapy has been recommended as first-line therapy with consideration of surgery if physiotherapy fails to sufficiently improve symptoms.7

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Women with moderate to severe SUI without significant prolapse or a history of prior incontinence surgery have significantly better outcomes at 12 months after undergoing midurethral sling surgery versus physiotherapy. Physiotherapy carries little to no risk of adverse effects. Women with moderate to severe SUI should be counseled regarding the risks and benefits of both physiotherapy and midurethral sling surgery as initial treatment options.
Because stress and urgency urinary symptoms often present together, it is important to consider urodynamic evaluation to confirm SUI prior to surgery in women with:
• mixed stress and urge symptoms
• a history of a previous surgery for incontinence, or
• poor correlation of physical examination findings to reported symptoms.

Safety and tolerability of mirabegron versus tolterodine for OAB

Chapple CR, Kaplan SA, Mitcheson D, et al. Randomized double-blind, active-controlled phase 3 study to assess 12-month safety and efficacy of mirabegron, a beta(3)-adrenoceptor agonist, in overactive bladder. Eur Urol. 2013;63(2):296−305.

In the bladder, beta3-receptors located within the detrusor smooth muscle facilitate urine storage by relaxing the detrusor, enabling the bladder to fill.10 The activation of beta3-receptors is thought to increase the bladder’s ability to store urine, with the goal of decreasing urgency, frequency, nocturia, and urgency incontinence. An alternative to anticholinergic medications, mirabegron is a beta3-agonist approved by the US Food and Drug Administration (FDA) in 2012 for clinical use in the treatment of OAB.

Details of the study
Chapple and colleagues aimed to assess the 12-month efficacy and safety of mirabegron in a randomized, double-blind active controlled trial. The primary outcome was incidence and severity of treatment-emergent adverse effects (TEAEs); the secondary outcome was the change in OAB symptoms from baseline to up to 12 months. Patients experiencing OAB symptoms for more than 3 months were eligible and were subsequently enrolled if they averaged 8 or more voids per day and 3 or more episodes of urgency with or without incontinence on a 3-day bladder diary. A total of 2,444 patients were randomly assigned in a 1:1:1 fashion to mirabegron 50 mg daily, mirabegron 100 mg daily, or tolterodine extended release (ER) 4 mg daily.

There was a similar incidence (60% to 63%) of TEAEs across all three groups. The most common TEAEs were hypertension (defined as average systolic blood pressure [BP] >140 mm Hg or average diastolic BP >90 mm Hg at two consecutive visits), UTI, headache, nasopharyngitis, and constipation. The adjusted mean changes in BP from baseline to final visit were less than 1 mm Hg for both systolic and diastolic BP for patients taking both doses of mirabegron, as well as for patients taking tolterodine. The incidence of dry mouth was higher in the tolterodine group than the mirabegron groups. Mirabegron 50 mg daily and 100 mg daily improved incontinence symptoms within 1 month of starting therapy; the degree of improvement was similar to that seen in the patients taking tolterodine ER 4 mg daily.

Related article: New overactive bladder treatment approved by the FDA (August 2012)

Some caveats
This study was well-designed to assess the safety and tolerability of mirabegron versus tolterodine. The doses utilized in the study were at or above the FDA-approved dosage of 25 mg to 50 mg daily for OAB treatment. Although investigators found mirabegron to be a safe alternative to anticholinergic medication, the study was not designed or powered to examine the efficacy of mirabegron versus tolterodine. No formal comparison of efficacy was made between mirabegron or tolterodine, or between the 50-mg and 100-mg doses of mirabegron. Moreover, 81% of participants had been treated with mirabegron in earlier Phase 3 studies, so most were not treatment naïve, limiting the applicability of results.

 

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Mirabegron should be considered as a potential treatment option for patients who demonstrate poor tolerance of or response to anticholinergic medications; however, caution should be used in patients with severe uncontrolled high BP, end-stage kidney disease, or severe liver impairment.

Consider percutaneous tibial nerve stimulation over tolterodine for OAB in select patients

Peters KM, Macdiarmid SA, Wooldridge LS, et al. Randomized trial of percutaneous tibial nerve stimulation versus extended-release tolterodine: Results from the overactive bladder innovative therapy trial. J Urol. 2009;182(3):1055−1061.

Neuromodulation utilizes electrical stimulation to improve bladder function and decrease OAB symptoms. First developed in the early 1980s by McGuire and colleagues, percutaneous tibial nerve stimulation (PTNS) was approved by the FDA in 2000 as Urgent PC and provides an outpatient, nonimplantable neuromodulation alternative to medication therapy for patients with OAB.11,12 By directly stimulating the posterior tibial nerve, PTNS works via the S3 sacral nerve plexus to alter the micturition reflex and improve bladder function.

Details of the study
Patients were eligible for the study if they demonstrated 8 or more voids per day on a 3-day bladder diary (whether or not they had a history of previous anticholinergic drug use). A total of 100 ambulatory adults with OAB symptoms were enrolled and randomly assigned to PTNS 30-minutes per week or tolterodine ER 4 mg daily.

At 12 weeks, both groups demonstrated a significant improvement in OAB measures as well as validated symptom severity and quality-of-life questionnaire scores. Subjective assessment of improvement in OAB symptoms was significantly greater in the PTNS group than in the tolterodine group (79.5% vs 54.8%, respectively; P = .01). However, mean reduction of voids for 24 hours was not significantly different between the two groups.

Both treatments were well tolerated, with only 15% to 16% of patients in both groups reporting mild to moderate side effects. The tolterodine group did have a significantly higher risk of dry mouth; however, the risk of constipation was not significantly different between the groups.

Study limitations
The authors performed an important multicenter, nonblinded, randomized, controlled trial, which was one of the first trials to directly compare two OAB therapies. The generalizability of the findings were limited, as the cohort included mostly patients with dry OAB who had no objective measures on UUI episodes. In addition, this trial had a limited observation period of only 12 weeks. Information regarding the effect of treatment after cessation of weekly PTNS therapy was not examined. Therefore, we are not able to determine whether repeat sessions provide adequate maintenance in the long term.

WHAT THIS EVIDENCE MEANS FOR PRACTICE
PTNS 30 minutes daily is as effective as tolterodine ER 4 mg daily for 12 weeks in reducing OAB symptoms. PTNS is a safe alternative that should be considered in patients with OAB who poorly tolerate or have contraindications to medication therapy.

OnabotulinumtoxinA is an effective therapy for OAB

Visco AG, Brubaker L, Richter HE, et al. Anticholinergic therapy vs onabotulinumtoxinA for urgency urinary incontinence. NEJM. 2012;367(19):1803−1813.

The newest therapy for OAB is onabotulinumtoxinA, or Botox, which was FDA approved this year for the treatment of OAB in adults who cannot use or do not tolerate anticholinergic medications. Recommended doses are 100 U onabotulinumtoxinA in patients with idiopathic refractory OAB and 200 U onabotulinumtoxinA for patients with neurogenic OAB.

OnabotulinumtoxinA is a neurotoxin that blocks synaptic transmission at the neuromuscular junction to cause muscle paralysis and atrophy.13 Injecting onabotulinumtoxinA into the detrusor smooth muscle should relax the bladder and decrease sensations of urgency and frequency to achieve a longer duration of time for bladder filling and reduce the risk of urgency incontinence.

Effects of onabotulinumtoxinA appear to wear off over time, and patients may require repeat injections. Side effects of onabotulinumtoxinA therapy include an increased risk of UTI and the potential for urinary retention requiring intermittent self-catheterization.

Related article: Update on Pelvic Floor Dysfunction Autumn L. Edenfield, MD, and Cindy L. Amundsen, MD (October 2012)

Details of the study
The Anticholinergic Versus Botulinum Toxin Comparison (ABC) study was a multicenter, randomized, double-blind, double-placebo–controlled trial conducted in women without known neurologic disease with moderate to severe UUI (defined as >5 UUI episodes on a 3-day bladder diary). Women were randomly assigned to a single intradetrusor injection of 100 U onabotulinumtoxinA plus oral placebo or to a single intradetrusor injection of saline plus solifenacin 5 mg daily (with the option of dose escalation and then switching to trospium XR if no improvement was seen).

Of the 241 women included in the final analysis, approximately 70% in each group reported adequate control of symptoms at 6 months. Adequate control was defined as a response of “agree strongly” or “agree” to the statement: “This treatment has given me adequate control of my urinary leakage.” Women in the onabotulinumtoxinA group were significantly more likely than women in the anticholinergic medication group to report complete resolution of UUI at 6 months (27% vs 13%, P = .003). However, the mean reduction in episodes of UUI per day and the improvements in quality-of-life questionnaire scores were found to be similar. Interestingly, worse baseline UUI was associated with greater reduction in episodes of UUI for both therapies.

 

 

This was a rigorous and well-executed double-blind, double-placebo−controlled randomized trial. By utilizing broad inclusion criteria and enrolling patients both with and without previous exposure to anticholinergic medications, the generalizability of study findings are greatly improved. Because this study did not examine the effect or efficacy of repeat injections, these findings have limited applicability to patients undergoing multiple onabotulinumtoxinA injections.

When considering use in your patient population, keep the possible side effects in mind.There were important differences in the side effects experienced with each therapy. Specifically, while the anticholinergic group had a higher frequency of dry mouth (46% anticholinergic vs 31% onabotulinumtoxinA, P = .02), the onabotulinumtoxinA group demonstrated higher rates of incomplete bladder emptying requiring catheterization (peak of 5% at 2 months) and greater risk of UTI (33% onabotulinumtoxinA vs 13% anticholinergic, P <.001).

WHAT THIS EVIDENCE MEANS FOR PRACTICE
This study showed that, among women with UUI, anticholinergic medication and onabotulinumtoxinA are equally effective in reducing UUI episodes and improving quality of life. It is important to consider the side effect profile, determine the patient’s preferences, and weigh the risks and benefits of each therapy when deciding what is the best treatment for your individual patient.

We want to hear from you! Tell us what you think.

References

  1. Anger JT, Saigal CS, Litwin MS. The prevalence of urinary incontinence among community dwelling adult women: Results from the National Health and Nutrition Examination Survey. J Urol. 2006;175(2):601–604.
  2. Dooley Y, Kenton K, Cao G, et al. Urinary incontinence prevalence: Results from the National Health and Nutrition Examination Survey. J Urol. 2008;179(2):656–661.
  3. Wu JM, Hundley AF, Fulton RG, Myers ER. Forecasting the prevalence of pelvic floor disorders in U.S. Women: 2010 to 2050. Obstetr Gynecol. 2009;114(6):1278–1283.
  4. Gormley EA, Lightner DJ, Burgio KL, et al. Diagnosis and treatment of overactive bladder (non-neurogenic) in adults: AUA/SUFU Guideline. Americal Urological Association. http://www.auanet.org/common/pdf/education/clinical-guidance/Overactive-Bladder.pdf. Published 2012. Revised June 11, 2013. Accessed October 21, 2013.
  5. Yu YF, Nichol MB, Yu AP, Ahn J. Persistence and adherence of medications for chronic overactive bladder/urinary incontinence in the California Medicaid program. Value Health. 2005;8(4):495–505.
  6. Haylen BT, de Ridder D, Freeman RM, et al. An International Urogynecological Association (IUGA)/International Continence Society (ICS) joint report on the terminology for female pelvic floor dysfunction. Int Urogynecol J. 2010;21(1):5–26.
  7. ACOG Practice Bulletin No. 63: Urinary incontinence in women. American College of Obstetricians and Gynecologists. Obstetr Gynecol. 2005;105(6):1533–1545.
  8. Bo K, Kvarstein B, Nygaard I. Lower urinary tract symptoms and pelvic floor muscle exercise adherence after 15 years. Obstetr Gynecol. 2005;105(5 Pt 1):999–1005.
  9. Sandvik H, Hunskaar S, Seim A, Hermstad R, Vanvik A, Bratt H. Validation of a severity index in female urinary incontinence and its implementation in an epidemiological survey. J Epidemiol Community Health. 1993;47(6):497–499.
  10. Fowler CJ, Griffiths D, de Groat WC. The neural control of micturition. Nat Rev Neurosci. 2008;9(6):453–466.
  11. Levin PJ, Wu JM, Kawasaki A, Weidner AC, Amundsen CL. The efficacy of posterior tibial nerve stimulation for the treatment of overactive bladder in women: a systematic review. Int Urogynecol J. 2012;23(11):1591–1597.
  12. McGuire EJ, Zhang SC, Horwinski ER, Lytton B. Treatment of motor and sensory detrusor instability by electrical stimulation. J Urol. 1983;129(1):78–79.
  13. Schiavo G, Santucci A, Dasgupta BR, et al. Botulinum neurotoxins serotypes A and E cleave SNAP-25 at distinct COOH-terminal peptide bonds. FEBS Lett. 1993;335(1):99–103.
Article PDF
Author and Disclosure Information

Alexis A. Dieter, MD, is a Fellow in the Division of Female Pelvic and Reconstructive Pelvic Surgery and Clinical Instructor of Obstetrics and Gynecology at Duke University in Durham, NC.

Cindy L. Amundsen, MD, is Professor and Fellowship Director of the Division of Female Pelvic and Reconstructive Pelvic Surgery, Department of Obstetrics and Gynecology, at Duke University in Durham, NC.

Dr. Dieter reports that she received the American Urogynecologic Society (AUGS) Foundation Thomas Benson Award, and participates in the NIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Loan Repayment Program. Dr. Amundsen reports no financial relationships relevant to this article.

Issue
OBG Management - 25(11)
Publications
Topics
Page Number
56-64
Legacy Keywords
pelvic floor dysfunction,urinary incontinence,UI,stress urinary incontinence,SUI,overactive bladder,OAB,urgency urinary incontinence,UUI,mixed urinary incontinence,anticholinergic medications,mirabegron,tolterodine, percutaneous tibial nerve stimulation,tolterodine,onabotulinumtoxinA
Sections
Author and Disclosure Information

Alexis A. Dieter, MD, is a Fellow in the Division of Female Pelvic and Reconstructive Pelvic Surgery and Clinical Instructor of Obstetrics and Gynecology at Duke University in Durham, NC.

Cindy L. Amundsen, MD, is Professor and Fellowship Director of the Division of Female Pelvic and Reconstructive Pelvic Surgery, Department of Obstetrics and Gynecology, at Duke University in Durham, NC.

Dr. Dieter reports that she received the American Urogynecologic Society (AUGS) Foundation Thomas Benson Award, and participates in the NIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Loan Repayment Program. Dr. Amundsen reports no financial relationships relevant to this article.

Author and Disclosure Information

Alexis A. Dieter, MD, is a Fellow in the Division of Female Pelvic and Reconstructive Pelvic Surgery and Clinical Instructor of Obstetrics and Gynecology at Duke University in Durham, NC.

Cindy L. Amundsen, MD, is Professor and Fellowship Director of the Division of Female Pelvic and Reconstructive Pelvic Surgery, Department of Obstetrics and Gynecology, at Duke University in Durham, NC.

Dr. Dieter reports that she received the American Urogynecologic Society (AUGS) Foundation Thomas Benson Award, and participates in the NIH National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) Loan Repayment Program. Dr. Amundsen reports no financial relationships relevant to this article.

Article PDF
Article PDF
Related Articles

Urinary incontinence (UI) affects almost half of all women in the United States.1,2 Estimates suggest that the prevalence of UI gradually rises during young adult life, comes to a broad plateau in middle age, and then steadily increases from that plateau after age 65. Therefore, over the next 40 years, as the elderly population expands in size, the number of women affected by UI will significantly grow.3

For patients with UI, a multitude of therapeutic options are available. Which option is the best for your patient? In this article, we aim to answer that question by interpreting the results of four randomized trials, each of which directly compare two available treatment options. The first study examines patients with stress urinary incontinence (SUI), comparing the patients’ subjective improvement in urinary leakage and bladder function at 12 months after randomization to treatment with physiotherapy or midurethral sling surgery.

The three other trials examine patients with overactive bladder (OAB) and urgency urinary incontinence (UUI). Each trial directly compares the use of anticholinergic medications to an alternate treatment modality. Currently, anticholinergic medications and behavioral therapy are the recommended first-line therapies for OAB. Unfortunately, anticholinergic medications have poor patient compliance and significant systemic side effects.4 Caution should be used when considering anticholinergic medications in patients with impaired gastric emptying or a history of urinary retention. They also should be used with caution in elderly patients who are extremely frail. Additionally, clearance from an ophthalmologist must be obtained prior to starting anticholinergic medication in patients with narrow-angle glaucoma.5 Due to poor adherence and potential side effects, there is a growing effort to discover alternative treatment modalities that are safe and effective. Therefore, we chose to examine trials comparing: mirabegron versus tolterodine, percutaneous tibial nerve stimulation versus tolterodine, and onabotulinumtoxinA versus anticholingeric medications.

UI defined
Before discussing treatment options, we want to clarify the main types of UI (FIGURE). UI is defined as the complaint of involuntary loss of urine.  UI can be subdivided into SUI, OAB/UUI, or mixed urinary incontinence.6 While there are other less common genitourinary etiologies that can lead to UI, nongenitourinary etiologies are prevalent and can aggravate existing SUI or OAB (TABLE).

SUI is the complaint of involuntary loss of urine on effort or physical exertion (such as during sporting activities) or on sneezing or coughing. Often, SUI can be diagnosed by patient report alone and surgery can be considered in symptomatic patients who demonstrate cough leakage on physical examination and normal postvoid residual volumes.

UUI is the involuntary loss of urine associated with urgency; it often occurs in the setting of OAB, which is defined as the syndrome of urinary urgency, usually accompanied by frequency and nocturia, with or without UUI, in the absence of urinary tract infection or other obvious pathology (such as neurologic dysfunction, infection, or urologic neoplasm). OAB-dry is present when patients do not have leakage with urgency, but are bothered by urgency, frequency, and/or nocturia. OAB-wet occurs when a patient has urgencyincontinence.

The presence of both SUI and OAB/UUI is known as mixed urinary incontinence. Stress and urgency urinary symptoms often present together. In fact, 10% to 30% of women with stress symptoms are found to have bladder overactivity on subsequent evaluation.2,7 Therefore, it is important to take a good history and consider urodynamic evaluation to confirm the diagnosis of SUI prior to surgery in women with mixed stress and urge symptoms, a history of a previous surgery for incontinence, or when there is a poor correlation of physical examination findings to reported symptoms.

 

Is surgery a first-line option for patients with SUI?

Labrie J, Berghmans BL, Fischer K, et al. Surgery versus physiotherapy for stress urinary incontinence. NEJM. 2013;369(12):1124−1133.

Physiotherapy, including pelvic floor muscle training (“Kegel exercises”), is utilized as a first-line treatment option for women with SUI that carries minimal risk for the patient. Midurethral sling surgery is often recommended if an initial trial of conservative treatment fails.7 Up to 50% of women treated with pelvic floor physiotherapy will ultimately undergo surgery to treat their SUI.8

Related article: Does urodynamic testing before surgery for stress incontinence improve outcomes? G. Willy Davila, MD (Examining the Evidence, December 2012)

Details of the study
This was a randomized, multicenter trial of women aged 35 to 80 years with moderate to severe SUI. After excluding women with previous incontinence surgery and stage 2 or higher pelvic organ prolapse, 460 participants were randomly assigned to undergo either a midurethral sling surgery or physiotherapy (pelvic floor muscle training). The primary outcome was subjective improvement in urinary leakage and bladder function at 12 months, as measured by the Patient Global Impression of Improvement (PGI-I), a 7-point Likert scale ranging from “very much worse” to “very much better.”

 

 

In an intention-to-treat analysis, subjective improvement at 12 months was significantly higher in women randomized to midurethral sling surgery than in women randomized to physiotherapy (91% vs 64%, respectively).

Ten percent of patients had adverse events (AEs); all were related to surgery. The most common AEs were hematoma, vaginal epithelial perforation, and bladder perforation.

Notably, women had the option to cross over to the other treatment modality if they desired. In the physiotherapy group, 49% of women elected to cross over to surgery, while 11% of those who underwent midurethral sling surgery elected to cross over to physiotherapy during the 12-month follow-up period. When analyzing results by treatment received, the investigators found that the proportion of women who reported improvement was significantly lower among women who underwent physiotherapy only (32%), versus sling only (94%), or sling after physiotherapy (91%).

This randomized trial was well-designed and included a variety of treatment centers (university and general hospitals) with interventions performed by experienced surgeons (all of whom had performed at least 20 sling surgeries) and physiotherapists educated specifically in pelvic floor physiotherapy. The study population was limited to patients with moderate to severe SUI as defined by the Sandvik severity index.9 Therefore, these results may not be applicable to patients with milder symptoms, for whom physiotherapy has been recommended as first-line therapy with consideration of surgery if physiotherapy fails to sufficiently improve symptoms.7

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Women with moderate to severe SUI without significant prolapse or a history of prior incontinence surgery have significantly better outcomes at 12 months after undergoing midurethral sling surgery versus physiotherapy. Physiotherapy carries little to no risk of adverse effects. Women with moderate to severe SUI should be counseled regarding the risks and benefits of both physiotherapy and midurethral sling surgery as initial treatment options.
Because stress and urgency urinary symptoms often present together, it is important to consider urodynamic evaluation to confirm SUI prior to surgery in women with:
• mixed stress and urge symptoms
• a history of a previous surgery for incontinence, or
• poor correlation of physical examination findings to reported symptoms.

Safety and tolerability of mirabegron versus tolterodine for OAB

Chapple CR, Kaplan SA, Mitcheson D, et al. Randomized double-blind, active-controlled phase 3 study to assess 12-month safety and efficacy of mirabegron, a beta(3)-adrenoceptor agonist, in overactive bladder. Eur Urol. 2013;63(2):296−305.

In the bladder, beta3-receptors located within the detrusor smooth muscle facilitate urine storage by relaxing the detrusor, enabling the bladder to fill.10 The activation of beta3-receptors is thought to increase the bladder’s ability to store urine, with the goal of decreasing urgency, frequency, nocturia, and urgency incontinence. An alternative to anticholinergic medications, mirabegron is a beta3-agonist approved by the US Food and Drug Administration (FDA) in 2012 for clinical use in the treatment of OAB.

Details of the study
Chapple and colleagues aimed to assess the 12-month efficacy and safety of mirabegron in a randomized, double-blind active controlled trial. The primary outcome was incidence and severity of treatment-emergent adverse effects (TEAEs); the secondary outcome was the change in OAB symptoms from baseline to up to 12 months. Patients experiencing OAB symptoms for more than 3 months were eligible and were subsequently enrolled if they averaged 8 or more voids per day and 3 or more episodes of urgency with or without incontinence on a 3-day bladder diary. A total of 2,444 patients were randomly assigned in a 1:1:1 fashion to mirabegron 50 mg daily, mirabegron 100 mg daily, or tolterodine extended release (ER) 4 mg daily.

There was a similar incidence (60% to 63%) of TEAEs across all three groups. The most common TEAEs were hypertension (defined as average systolic blood pressure [BP] >140 mm Hg or average diastolic BP >90 mm Hg at two consecutive visits), UTI, headache, nasopharyngitis, and constipation. The adjusted mean changes in BP from baseline to final visit were less than 1 mm Hg for both systolic and diastolic BP for patients taking both doses of mirabegron, as well as for patients taking tolterodine. The incidence of dry mouth was higher in the tolterodine group than the mirabegron groups. Mirabegron 50 mg daily and 100 mg daily improved incontinence symptoms within 1 month of starting therapy; the degree of improvement was similar to that seen in the patients taking tolterodine ER 4 mg daily.

Related article: New overactive bladder treatment approved by the FDA (August 2012)

Some caveats
This study was well-designed to assess the safety and tolerability of mirabegron versus tolterodine. The doses utilized in the study were at or above the FDA-approved dosage of 25 mg to 50 mg daily for OAB treatment. Although investigators found mirabegron to be a safe alternative to anticholinergic medication, the study was not designed or powered to examine the efficacy of mirabegron versus tolterodine. No formal comparison of efficacy was made between mirabegron or tolterodine, or between the 50-mg and 100-mg doses of mirabegron. Moreover, 81% of participants had been treated with mirabegron in earlier Phase 3 studies, so most were not treatment naïve, limiting the applicability of results.

 

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Mirabegron should be considered as a potential treatment option for patients who demonstrate poor tolerance of or response to anticholinergic medications; however, caution should be used in patients with severe uncontrolled high BP, end-stage kidney disease, or severe liver impairment.

Consider percutaneous tibial nerve stimulation over tolterodine for OAB in select patients

Peters KM, Macdiarmid SA, Wooldridge LS, et al. Randomized trial of percutaneous tibial nerve stimulation versus extended-release tolterodine: Results from the overactive bladder innovative therapy trial. J Urol. 2009;182(3):1055−1061.

Neuromodulation utilizes electrical stimulation to improve bladder function and decrease OAB symptoms. First developed in the early 1980s by McGuire and colleagues, percutaneous tibial nerve stimulation (PTNS) was approved by the FDA in 2000 as Urgent PC and provides an outpatient, nonimplantable neuromodulation alternative to medication therapy for patients with OAB.11,12 By directly stimulating the posterior tibial nerve, PTNS works via the S3 sacral nerve plexus to alter the micturition reflex and improve bladder function.

Details of the study
Patients were eligible for the study if they demonstrated 8 or more voids per day on a 3-day bladder diary (whether or not they had a history of previous anticholinergic drug use). A total of 100 ambulatory adults with OAB symptoms were enrolled and randomly assigned to PTNS 30-minutes per week or tolterodine ER 4 mg daily.

At 12 weeks, both groups demonstrated a significant improvement in OAB measures as well as validated symptom severity and quality-of-life questionnaire scores. Subjective assessment of improvement in OAB symptoms was significantly greater in the PTNS group than in the tolterodine group (79.5% vs 54.8%, respectively; P = .01). However, mean reduction of voids for 24 hours was not significantly different between the two groups.

Both treatments were well tolerated, with only 15% to 16% of patients in both groups reporting mild to moderate side effects. The tolterodine group did have a significantly higher risk of dry mouth; however, the risk of constipation was not significantly different between the groups.

Study limitations
The authors performed an important multicenter, nonblinded, randomized, controlled trial, which was one of the first trials to directly compare two OAB therapies. The generalizability of the findings were limited, as the cohort included mostly patients with dry OAB who had no objective measures on UUI episodes. In addition, this trial had a limited observation period of only 12 weeks. Information regarding the effect of treatment after cessation of weekly PTNS therapy was not examined. Therefore, we are not able to determine whether repeat sessions provide adequate maintenance in the long term.

WHAT THIS EVIDENCE MEANS FOR PRACTICE
PTNS 30 minutes daily is as effective as tolterodine ER 4 mg daily for 12 weeks in reducing OAB symptoms. PTNS is a safe alternative that should be considered in patients with OAB who poorly tolerate or have contraindications to medication therapy.

OnabotulinumtoxinA is an effective therapy for OAB

Visco AG, Brubaker L, Richter HE, et al. Anticholinergic therapy vs onabotulinumtoxinA for urgency urinary incontinence. NEJM. 2012;367(19):1803−1813.

The newest therapy for OAB is onabotulinumtoxinA, or Botox, which was FDA approved this year for the treatment of OAB in adults who cannot use or do not tolerate anticholinergic medications. Recommended doses are 100 U onabotulinumtoxinA in patients with idiopathic refractory OAB and 200 U onabotulinumtoxinA for patients with neurogenic OAB.

OnabotulinumtoxinA is a neurotoxin that blocks synaptic transmission at the neuromuscular junction to cause muscle paralysis and atrophy.13 Injecting onabotulinumtoxinA into the detrusor smooth muscle should relax the bladder and decrease sensations of urgency and frequency to achieve a longer duration of time for bladder filling and reduce the risk of urgency incontinence.

Effects of onabotulinumtoxinA appear to wear off over time, and patients may require repeat injections. Side effects of onabotulinumtoxinA therapy include an increased risk of UTI and the potential for urinary retention requiring intermittent self-catheterization.

Related article: Update on Pelvic Floor Dysfunction Autumn L. Edenfield, MD, and Cindy L. Amundsen, MD (October 2012)

Details of the study
The Anticholinergic Versus Botulinum Toxin Comparison (ABC) study was a multicenter, randomized, double-blind, double-placebo–controlled trial conducted in women without known neurologic disease with moderate to severe UUI (defined as >5 UUI episodes on a 3-day bladder diary). Women were randomly assigned to a single intradetrusor injection of 100 U onabotulinumtoxinA plus oral placebo or to a single intradetrusor injection of saline plus solifenacin 5 mg daily (with the option of dose escalation and then switching to trospium XR if no improvement was seen).

Of the 241 women included in the final analysis, approximately 70% in each group reported adequate control of symptoms at 6 months. Adequate control was defined as a response of “agree strongly” or “agree” to the statement: “This treatment has given me adequate control of my urinary leakage.” Women in the onabotulinumtoxinA group were significantly more likely than women in the anticholinergic medication group to report complete resolution of UUI at 6 months (27% vs 13%, P = .003). However, the mean reduction in episodes of UUI per day and the improvements in quality-of-life questionnaire scores were found to be similar. Interestingly, worse baseline UUI was associated with greater reduction in episodes of UUI for both therapies.

 

 

This was a rigorous and well-executed double-blind, double-placebo−controlled randomized trial. By utilizing broad inclusion criteria and enrolling patients both with and without previous exposure to anticholinergic medications, the generalizability of study findings are greatly improved. Because this study did not examine the effect or efficacy of repeat injections, these findings have limited applicability to patients undergoing multiple onabotulinumtoxinA injections.

When considering use in your patient population, keep the possible side effects in mind.There were important differences in the side effects experienced with each therapy. Specifically, while the anticholinergic group had a higher frequency of dry mouth (46% anticholinergic vs 31% onabotulinumtoxinA, P = .02), the onabotulinumtoxinA group demonstrated higher rates of incomplete bladder emptying requiring catheterization (peak of 5% at 2 months) and greater risk of UTI (33% onabotulinumtoxinA vs 13% anticholinergic, P <.001).

WHAT THIS EVIDENCE MEANS FOR PRACTICE
This study showed that, among women with UUI, anticholinergic medication and onabotulinumtoxinA are equally effective in reducing UUI episodes and improving quality of life. It is important to consider the side effect profile, determine the patient’s preferences, and weigh the risks and benefits of each therapy when deciding what is the best treatment for your individual patient.

We want to hear from you! Tell us what you think.

Urinary incontinence (UI) affects almost half of all women in the United States.1,2 Estimates suggest that the prevalence of UI gradually rises during young adult life, comes to a broad plateau in middle age, and then steadily increases from that plateau after age 65. Therefore, over the next 40 years, as the elderly population expands in size, the number of women affected by UI will significantly grow.3

For patients with UI, a multitude of therapeutic options are available. Which option is the best for your patient? In this article, we aim to answer that question by interpreting the results of four randomized trials, each of which directly compare two available treatment options. The first study examines patients with stress urinary incontinence (SUI), comparing the patients’ subjective improvement in urinary leakage and bladder function at 12 months after randomization to treatment with physiotherapy or midurethral sling surgery.

The three other trials examine patients with overactive bladder (OAB) and urgency urinary incontinence (UUI). Each trial directly compares the use of anticholinergic medications to an alternate treatment modality. Currently, anticholinergic medications and behavioral therapy are the recommended first-line therapies for OAB. Unfortunately, anticholinergic medications have poor patient compliance and significant systemic side effects.4 Caution should be used when considering anticholinergic medications in patients with impaired gastric emptying or a history of urinary retention. They also should be used with caution in elderly patients who are extremely frail. Additionally, clearance from an ophthalmologist must be obtained prior to starting anticholinergic medication in patients with narrow-angle glaucoma.5 Due to poor adherence and potential side effects, there is a growing effort to discover alternative treatment modalities that are safe and effective. Therefore, we chose to examine trials comparing: mirabegron versus tolterodine, percutaneous tibial nerve stimulation versus tolterodine, and onabotulinumtoxinA versus anticholingeric medications.

UI defined
Before discussing treatment options, we want to clarify the main types of UI (FIGURE). UI is defined as the complaint of involuntary loss of urine.  UI can be subdivided into SUI, OAB/UUI, or mixed urinary incontinence.6 While there are other less common genitourinary etiologies that can lead to UI, nongenitourinary etiologies are prevalent and can aggravate existing SUI or OAB (TABLE).

SUI is the complaint of involuntary loss of urine on effort or physical exertion (such as during sporting activities) or on sneezing or coughing. Often, SUI can be diagnosed by patient report alone and surgery can be considered in symptomatic patients who demonstrate cough leakage on physical examination and normal postvoid residual volumes.

UUI is the involuntary loss of urine associated with urgency; it often occurs in the setting of OAB, which is defined as the syndrome of urinary urgency, usually accompanied by frequency and nocturia, with or without UUI, in the absence of urinary tract infection or other obvious pathology (such as neurologic dysfunction, infection, or urologic neoplasm). OAB-dry is present when patients do not have leakage with urgency, but are bothered by urgency, frequency, and/or nocturia. OAB-wet occurs when a patient has urgencyincontinence.

The presence of both SUI and OAB/UUI is known as mixed urinary incontinence. Stress and urgency urinary symptoms often present together. In fact, 10% to 30% of women with stress symptoms are found to have bladder overactivity on subsequent evaluation.2,7 Therefore, it is important to take a good history and consider urodynamic evaluation to confirm the diagnosis of SUI prior to surgery in women with mixed stress and urge symptoms, a history of a previous surgery for incontinence, or when there is a poor correlation of physical examination findings to reported symptoms.

 

Is surgery a first-line option for patients with SUI?

Labrie J, Berghmans BL, Fischer K, et al. Surgery versus physiotherapy for stress urinary incontinence. NEJM. 2013;369(12):1124−1133.

Physiotherapy, including pelvic floor muscle training (“Kegel exercises”), is utilized as a first-line treatment option for women with SUI that carries minimal risk for the patient. Midurethral sling surgery is often recommended if an initial trial of conservative treatment fails.7 Up to 50% of women treated with pelvic floor physiotherapy will ultimately undergo surgery to treat their SUI.8

Related article: Does urodynamic testing before surgery for stress incontinence improve outcomes? G. Willy Davila, MD (Examining the Evidence, December 2012)

Details of the study
This was a randomized, multicenter trial of women aged 35 to 80 years with moderate to severe SUI. After excluding women with previous incontinence surgery and stage 2 or higher pelvic organ prolapse, 460 participants were randomly assigned to undergo either a midurethral sling surgery or physiotherapy (pelvic floor muscle training). The primary outcome was subjective improvement in urinary leakage and bladder function at 12 months, as measured by the Patient Global Impression of Improvement (PGI-I), a 7-point Likert scale ranging from “very much worse” to “very much better.”

 

 

In an intention-to-treat analysis, subjective improvement at 12 months was significantly higher in women randomized to midurethral sling surgery than in women randomized to physiotherapy (91% vs 64%, respectively).

Ten percent of patients had adverse events (AEs); all were related to surgery. The most common AEs were hematoma, vaginal epithelial perforation, and bladder perforation.

Notably, women had the option to cross over to the other treatment modality if they desired. In the physiotherapy group, 49% of women elected to cross over to surgery, while 11% of those who underwent midurethral sling surgery elected to cross over to physiotherapy during the 12-month follow-up period. When analyzing results by treatment received, the investigators found that the proportion of women who reported improvement was significantly lower among women who underwent physiotherapy only (32%), versus sling only (94%), or sling after physiotherapy (91%).

This randomized trial was well-designed and included a variety of treatment centers (university and general hospitals) with interventions performed by experienced surgeons (all of whom had performed at least 20 sling surgeries) and physiotherapists educated specifically in pelvic floor physiotherapy. The study population was limited to patients with moderate to severe SUI as defined by the Sandvik severity index.9 Therefore, these results may not be applicable to patients with milder symptoms, for whom physiotherapy has been recommended as first-line therapy with consideration of surgery if physiotherapy fails to sufficiently improve symptoms.7

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Women with moderate to severe SUI without significant prolapse or a history of prior incontinence surgery have significantly better outcomes at 12 months after undergoing midurethral sling surgery versus physiotherapy. Physiotherapy carries little to no risk of adverse effects. Women with moderate to severe SUI should be counseled regarding the risks and benefits of both physiotherapy and midurethral sling surgery as initial treatment options.
Because stress and urgency urinary symptoms often present together, it is important to consider urodynamic evaluation to confirm SUI prior to surgery in women with:
• mixed stress and urge symptoms
• a history of a previous surgery for incontinence, or
• poor correlation of physical examination findings to reported symptoms.

Safety and tolerability of mirabegron versus tolterodine for OAB

Chapple CR, Kaplan SA, Mitcheson D, et al. Randomized double-blind, active-controlled phase 3 study to assess 12-month safety and efficacy of mirabegron, a beta(3)-adrenoceptor agonist, in overactive bladder. Eur Urol. 2013;63(2):296−305.

In the bladder, beta3-receptors located within the detrusor smooth muscle facilitate urine storage by relaxing the detrusor, enabling the bladder to fill.10 The activation of beta3-receptors is thought to increase the bladder’s ability to store urine, with the goal of decreasing urgency, frequency, nocturia, and urgency incontinence. An alternative to anticholinergic medications, mirabegron is a beta3-agonist approved by the US Food and Drug Administration (FDA) in 2012 for clinical use in the treatment of OAB.

Details of the study
Chapple and colleagues aimed to assess the 12-month efficacy and safety of mirabegron in a randomized, double-blind active controlled trial. The primary outcome was incidence and severity of treatment-emergent adverse effects (TEAEs); the secondary outcome was the change in OAB symptoms from baseline to up to 12 months. Patients experiencing OAB symptoms for more than 3 months were eligible and were subsequently enrolled if they averaged 8 or more voids per day and 3 or more episodes of urgency with or without incontinence on a 3-day bladder diary. A total of 2,444 patients were randomly assigned in a 1:1:1 fashion to mirabegron 50 mg daily, mirabegron 100 mg daily, or tolterodine extended release (ER) 4 mg daily.

There was a similar incidence (60% to 63%) of TEAEs across all three groups. The most common TEAEs were hypertension (defined as average systolic blood pressure [BP] >140 mm Hg or average diastolic BP >90 mm Hg at two consecutive visits), UTI, headache, nasopharyngitis, and constipation. The adjusted mean changes in BP from baseline to final visit were less than 1 mm Hg for both systolic and diastolic BP for patients taking both doses of mirabegron, as well as for patients taking tolterodine. The incidence of dry mouth was higher in the tolterodine group than the mirabegron groups. Mirabegron 50 mg daily and 100 mg daily improved incontinence symptoms within 1 month of starting therapy; the degree of improvement was similar to that seen in the patients taking tolterodine ER 4 mg daily.

Related article: New overactive bladder treatment approved by the FDA (August 2012)

Some caveats
This study was well-designed to assess the safety and tolerability of mirabegron versus tolterodine. The doses utilized in the study were at or above the FDA-approved dosage of 25 mg to 50 mg daily for OAB treatment. Although investigators found mirabegron to be a safe alternative to anticholinergic medication, the study was not designed or powered to examine the efficacy of mirabegron versus tolterodine. No formal comparison of efficacy was made between mirabegron or tolterodine, or between the 50-mg and 100-mg doses of mirabegron. Moreover, 81% of participants had been treated with mirabegron in earlier Phase 3 studies, so most were not treatment naïve, limiting the applicability of results.

 

 

WHAT THIS EVIDENCE MEANS FOR PRACTICE
Mirabegron should be considered as a potential treatment option for patients who demonstrate poor tolerance of or response to anticholinergic medications; however, caution should be used in patients with severe uncontrolled high BP, end-stage kidney disease, or severe liver impairment.

Consider percutaneous tibial nerve stimulation over tolterodine for OAB in select patients

Peters KM, Macdiarmid SA, Wooldridge LS, et al. Randomized trial of percutaneous tibial nerve stimulation versus extended-release tolterodine: Results from the overactive bladder innovative therapy trial. J Urol. 2009;182(3):1055−1061.

Neuromodulation utilizes electrical stimulation to improve bladder function and decrease OAB symptoms. First developed in the early 1980s by McGuire and colleagues, percutaneous tibial nerve stimulation (PTNS) was approved by the FDA in 2000 as Urgent PC and provides an outpatient, nonimplantable neuromodulation alternative to medication therapy for patients with OAB.11,12 By directly stimulating the posterior tibial nerve, PTNS works via the S3 sacral nerve plexus to alter the micturition reflex and improve bladder function.

Details of the study
Patients were eligible for the study if they demonstrated 8 or more voids per day on a 3-day bladder diary (whether or not they had a history of previous anticholinergic drug use). A total of 100 ambulatory adults with OAB symptoms were enrolled and randomly assigned to PTNS 30-minutes per week or tolterodine ER 4 mg daily.

At 12 weeks, both groups demonstrated a significant improvement in OAB measures as well as validated symptom severity and quality-of-life questionnaire scores. Subjective assessment of improvement in OAB symptoms was significantly greater in the PTNS group than in the tolterodine group (79.5% vs 54.8%, respectively; P = .01). However, mean reduction of voids for 24 hours was not significantly different between the two groups.

Both treatments were well tolerated, with only 15% to 16% of patients in both groups reporting mild to moderate side effects. The tolterodine group did have a significantly higher risk of dry mouth; however, the risk of constipation was not significantly different between the groups.

Study limitations
The authors performed an important multicenter, nonblinded, randomized, controlled trial, which was one of the first trials to directly compare two OAB therapies. The generalizability of the findings were limited, as the cohort included mostly patients with dry OAB who had no objective measures on UUI episodes. In addition, this trial had a limited observation period of only 12 weeks. Information regarding the effect of treatment after cessation of weekly PTNS therapy was not examined. Therefore, we are not able to determine whether repeat sessions provide adequate maintenance in the long term.

WHAT THIS EVIDENCE MEANS FOR PRACTICE
PTNS 30 minutes daily is as effective as tolterodine ER 4 mg daily for 12 weeks in reducing OAB symptoms. PTNS is a safe alternative that should be considered in patients with OAB who poorly tolerate or have contraindications to medication therapy.

OnabotulinumtoxinA is an effective therapy for OAB

Visco AG, Brubaker L, Richter HE, et al. Anticholinergic therapy vs onabotulinumtoxinA for urgency urinary incontinence. NEJM. 2012;367(19):1803−1813.

The newest therapy for OAB is onabotulinumtoxinA, or Botox, which was FDA approved this year for the treatment of OAB in adults who cannot use or do not tolerate anticholinergic medications. Recommended doses are 100 U onabotulinumtoxinA in patients with idiopathic refractory OAB and 200 U onabotulinumtoxinA for patients with neurogenic OAB.

OnabotulinumtoxinA is a neurotoxin that blocks synaptic transmission at the neuromuscular junction to cause muscle paralysis and atrophy.13 Injecting onabotulinumtoxinA into the detrusor smooth muscle should relax the bladder and decrease sensations of urgency and frequency to achieve a longer duration of time for bladder filling and reduce the risk of urgency incontinence.

Effects of onabotulinumtoxinA appear to wear off over time, and patients may require repeat injections. Side effects of onabotulinumtoxinA therapy include an increased risk of UTI and the potential for urinary retention requiring intermittent self-catheterization.

Related article: Update on Pelvic Floor Dysfunction Autumn L. Edenfield, MD, and Cindy L. Amundsen, MD (October 2012)

Details of the study
The Anticholinergic Versus Botulinum Toxin Comparison (ABC) study was a multicenter, randomized, double-blind, double-placebo–controlled trial conducted in women without known neurologic disease with moderate to severe UUI (defined as >5 UUI episodes on a 3-day bladder diary). Women were randomly assigned to a single intradetrusor injection of 100 U onabotulinumtoxinA plus oral placebo or to a single intradetrusor injection of saline plus solifenacin 5 mg daily (with the option of dose escalation and then switching to trospium XR if no improvement was seen).

Of the 241 women included in the final analysis, approximately 70% in each group reported adequate control of symptoms at 6 months. Adequate control was defined as a response of “agree strongly” or “agree” to the statement: “This treatment has given me adequate control of my urinary leakage.” Women in the onabotulinumtoxinA group were significantly more likely than women in the anticholinergic medication group to report complete resolution of UUI at 6 months (27% vs 13%, P = .003). However, the mean reduction in episodes of UUI per day and the improvements in quality-of-life questionnaire scores were found to be similar. Interestingly, worse baseline UUI was associated with greater reduction in episodes of UUI for both therapies.

 

 

This was a rigorous and well-executed double-blind, double-placebo−controlled randomized trial. By utilizing broad inclusion criteria and enrolling patients both with and without previous exposure to anticholinergic medications, the generalizability of study findings are greatly improved. Because this study did not examine the effect or efficacy of repeat injections, these findings have limited applicability to patients undergoing multiple onabotulinumtoxinA injections.

When considering use in your patient population, keep the possible side effects in mind.There were important differences in the side effects experienced with each therapy. Specifically, while the anticholinergic group had a higher frequency of dry mouth (46% anticholinergic vs 31% onabotulinumtoxinA, P = .02), the onabotulinumtoxinA group demonstrated higher rates of incomplete bladder emptying requiring catheterization (peak of 5% at 2 months) and greater risk of UTI (33% onabotulinumtoxinA vs 13% anticholinergic, P <.001).

WHAT THIS EVIDENCE MEANS FOR PRACTICE
This study showed that, among women with UUI, anticholinergic medication and onabotulinumtoxinA are equally effective in reducing UUI episodes and improving quality of life. It is important to consider the side effect profile, determine the patient’s preferences, and weigh the risks and benefits of each therapy when deciding what is the best treatment for your individual patient.

We want to hear from you! Tell us what you think.

References

  1. Anger JT, Saigal CS, Litwin MS. The prevalence of urinary incontinence among community dwelling adult women: Results from the National Health and Nutrition Examination Survey. J Urol. 2006;175(2):601–604.
  2. Dooley Y, Kenton K, Cao G, et al. Urinary incontinence prevalence: Results from the National Health and Nutrition Examination Survey. J Urol. 2008;179(2):656–661.
  3. Wu JM, Hundley AF, Fulton RG, Myers ER. Forecasting the prevalence of pelvic floor disorders in U.S. Women: 2010 to 2050. Obstetr Gynecol. 2009;114(6):1278–1283.
  4. Gormley EA, Lightner DJ, Burgio KL, et al. Diagnosis and treatment of overactive bladder (non-neurogenic) in adults: AUA/SUFU Guideline. Americal Urological Association. http://www.auanet.org/common/pdf/education/clinical-guidance/Overactive-Bladder.pdf. Published 2012. Revised June 11, 2013. Accessed October 21, 2013.
  5. Yu YF, Nichol MB, Yu AP, Ahn J. Persistence and adherence of medications for chronic overactive bladder/urinary incontinence in the California Medicaid program. Value Health. 2005;8(4):495–505.
  6. Haylen BT, de Ridder D, Freeman RM, et al. An International Urogynecological Association (IUGA)/International Continence Society (ICS) joint report on the terminology for female pelvic floor dysfunction. Int Urogynecol J. 2010;21(1):5–26.
  7. ACOG Practice Bulletin No. 63: Urinary incontinence in women. American College of Obstetricians and Gynecologists. Obstetr Gynecol. 2005;105(6):1533–1545.
  8. Bo K, Kvarstein B, Nygaard I. Lower urinary tract symptoms and pelvic floor muscle exercise adherence after 15 years. Obstetr Gynecol. 2005;105(5 Pt 1):999–1005.
  9. Sandvik H, Hunskaar S, Seim A, Hermstad R, Vanvik A, Bratt H. Validation of a severity index in female urinary incontinence and its implementation in an epidemiological survey. J Epidemiol Community Health. 1993;47(6):497–499.
  10. Fowler CJ, Griffiths D, de Groat WC. The neural control of micturition. Nat Rev Neurosci. 2008;9(6):453–466.
  11. Levin PJ, Wu JM, Kawasaki A, Weidner AC, Amundsen CL. The efficacy of posterior tibial nerve stimulation for the treatment of overactive bladder in women: a systematic review. Int Urogynecol J. 2012;23(11):1591–1597.
  12. McGuire EJ, Zhang SC, Horwinski ER, Lytton B. Treatment of motor and sensory detrusor instability by electrical stimulation. J Urol. 1983;129(1):78–79.
  13. Schiavo G, Santucci A, Dasgupta BR, et al. Botulinum neurotoxins serotypes A and E cleave SNAP-25 at distinct COOH-terminal peptide bonds. FEBS Lett. 1993;335(1):99–103.
References

  1. Anger JT, Saigal CS, Litwin MS. The prevalence of urinary incontinence among community dwelling adult women: Results from the National Health and Nutrition Examination Survey. J Urol. 2006;175(2):601–604.
  2. Dooley Y, Kenton K, Cao G, et al. Urinary incontinence prevalence: Results from the National Health and Nutrition Examination Survey. J Urol. 2008;179(2):656–661.
  3. Wu JM, Hundley AF, Fulton RG, Myers ER. Forecasting the prevalence of pelvic floor disorders in U.S. Women: 2010 to 2050. Obstetr Gynecol. 2009;114(6):1278–1283.
  4. Gormley EA, Lightner DJ, Burgio KL, et al. Diagnosis and treatment of overactive bladder (non-neurogenic) in adults: AUA/SUFU Guideline. Americal Urological Association. http://www.auanet.org/common/pdf/education/clinical-guidance/Overactive-Bladder.pdf. Published 2012. Revised June 11, 2013. Accessed October 21, 2013.
  5. Yu YF, Nichol MB, Yu AP, Ahn J. Persistence and adherence of medications for chronic overactive bladder/urinary incontinence in the California Medicaid program. Value Health. 2005;8(4):495–505.
  6. Haylen BT, de Ridder D, Freeman RM, et al. An International Urogynecological Association (IUGA)/International Continence Society (ICS) joint report on the terminology for female pelvic floor dysfunction. Int Urogynecol J. 2010;21(1):5–26.
  7. ACOG Practice Bulletin No. 63: Urinary incontinence in women. American College of Obstetricians and Gynecologists. Obstetr Gynecol. 2005;105(6):1533–1545.
  8. Bo K, Kvarstein B, Nygaard I. Lower urinary tract symptoms and pelvic floor muscle exercise adherence after 15 years. Obstetr Gynecol. 2005;105(5 Pt 1):999–1005.
  9. Sandvik H, Hunskaar S, Seim A, Hermstad R, Vanvik A, Bratt H. Validation of a severity index in female urinary incontinence and its implementation in an epidemiological survey. J Epidemiol Community Health. 1993;47(6):497–499.
  10. Fowler CJ, Griffiths D, de Groat WC. The neural control of micturition. Nat Rev Neurosci. 2008;9(6):453–466.
  11. Levin PJ, Wu JM, Kawasaki A, Weidner AC, Amundsen CL. The efficacy of posterior tibial nerve stimulation for the treatment of overactive bladder in women: a systematic review. Int Urogynecol J. 2012;23(11):1591–1597.
  12. McGuire EJ, Zhang SC, Horwinski ER, Lytton B. Treatment of motor and sensory detrusor instability by electrical stimulation. J Urol. 1983;129(1):78–79.
  13. Schiavo G, Santucci A, Dasgupta BR, et al. Botulinum neurotoxins serotypes A and E cleave SNAP-25 at distinct COOH-terminal peptide bonds. FEBS Lett. 1993;335(1):99–103.
Issue
OBG Management - 25(11)
Issue
OBG Management - 25(11)
Page Number
56-64
Page Number
56-64
Publications
Publications
Topics
Article Type
Display Headline
Update on pelvic floor dysfunction: Focus on urinary incontinence
Display Headline
Update on pelvic floor dysfunction: Focus on urinary incontinence
Legacy Keywords
pelvic floor dysfunction,urinary incontinence,UI,stress urinary incontinence,SUI,overactive bladder,OAB,urgency urinary incontinence,UUI,mixed urinary incontinence,anticholinergic medications,mirabegron,tolterodine, percutaneous tibial nerve stimulation,tolterodine,onabotulinumtoxinA
Legacy Keywords
pelvic floor dysfunction,urinary incontinence,UI,stress urinary incontinence,SUI,overactive bladder,OAB,urgency urinary incontinence,UUI,mixed urinary incontinence,anticholinergic medications,mirabegron,tolterodine, percutaneous tibial nerve stimulation,tolterodine,onabotulinumtoxinA
Sections
Article Source

PURLs Copyright

Inside the Article

Article PDF Media