Fast growing hand lesion

Article Type
Changed
Mon, 10/17/2022 - 08:18
Display Headline
Fast growing hand lesion

Fast growing hand lesion

A scoop shave biopsy at the lower edge of the lesion revealed that this was a well-differentiated squamous cell carcinoma.

Squamous cell carcinoma is the second most common cancer in the United States and the most common skin cancer in Black people.1 A patient’s age and their accumulated UV radiation from sun exposure or artificial tanning is a major contributing factor. Lesions may manifest as precancers characterized as rough pink or brown papules with a sandpaper-like texture on sun-exposed skin. These lesions may clear spontaneously or develop into invasive disease, as occurred in this case.

Surgical treatment is often curative. Fusiform excision and Mohs micrographic surgery are 2 common options. More advanced squamous cell carcinomas that are large or found to have poorly differentiated cells or large perineural invasion carry a risk of metastasis.

In elderly patients, optimal treatment isn’t always straightforward.1 Nonsurgical options include radiation and intralesional chemotherapy. These nonsurgical choices may seem less aggressive, but total inconvenience, wound care, and discomfort can be equal to or worse than a single session of curative surgery.

This patient’s lesion was excised with a 5-mm margin. The patient tolerated an in-office procedure lasting about 45 minutes but would have struggled with a longer session with Mohs microsurgery. The postoperative period required limiting full use of his left hand for about 2 weeks.

Photos and text for Photo Rounds Friday courtesy of Jonathan Karnes, MD (copyright retained). Dr. Karnes is the medical director of MDFMR Dermatology Services, Augusta, ME.

References

1. Bradford PT. Skin cancer in skin of color. Dermatol Nurs. 2009;21:170-177, 206; quiz 178. 2. Renzi M Jr, Schimmel J, Decker A, et al. Management of skin cancer in the elderly. Dermatol Clin. 2019;37:279-286. doi: 10.1016/j.det.2019.02.003

Issue
The Journal of Family Practice - 71(7)
Publications
Topics
Sections

Fast growing hand lesion

A scoop shave biopsy at the lower edge of the lesion revealed that this was a well-differentiated squamous cell carcinoma.

Squamous cell carcinoma is the second most common cancer in the United States and the most common skin cancer in Black people.1 A patient’s age and their accumulated UV radiation from sun exposure or artificial tanning is a major contributing factor. Lesions may manifest as precancers characterized as rough pink or brown papules with a sandpaper-like texture on sun-exposed skin. These lesions may clear spontaneously or develop into invasive disease, as occurred in this case.

Surgical treatment is often curative. Fusiform excision and Mohs micrographic surgery are 2 common options. More advanced squamous cell carcinomas that are large or found to have poorly differentiated cells or large perineural invasion carry a risk of metastasis.

In elderly patients, optimal treatment isn’t always straightforward.1 Nonsurgical options include radiation and intralesional chemotherapy. These nonsurgical choices may seem less aggressive, but total inconvenience, wound care, and discomfort can be equal to or worse than a single session of curative surgery.

This patient’s lesion was excised with a 5-mm margin. The patient tolerated an in-office procedure lasting about 45 minutes but would have struggled with a longer session with Mohs microsurgery. The postoperative period required limiting full use of his left hand for about 2 weeks.

Photos and text for Photo Rounds Friday courtesy of Jonathan Karnes, MD (copyright retained). Dr. Karnes is the medical director of MDFMR Dermatology Services, Augusta, ME.

Fast growing hand lesion

A scoop shave biopsy at the lower edge of the lesion revealed that this was a well-differentiated squamous cell carcinoma.

Squamous cell carcinoma is the second most common cancer in the United States and the most common skin cancer in Black people.1 A patient’s age and their accumulated UV radiation from sun exposure or artificial tanning is a major contributing factor. Lesions may manifest as precancers characterized as rough pink or brown papules with a sandpaper-like texture on sun-exposed skin. These lesions may clear spontaneously or develop into invasive disease, as occurred in this case.

Surgical treatment is often curative. Fusiform excision and Mohs micrographic surgery are 2 common options. More advanced squamous cell carcinomas that are large or found to have poorly differentiated cells or large perineural invasion carry a risk of metastasis.

In elderly patients, optimal treatment isn’t always straightforward.1 Nonsurgical options include radiation and intralesional chemotherapy. These nonsurgical choices may seem less aggressive, but total inconvenience, wound care, and discomfort can be equal to or worse than a single session of curative surgery.

This patient’s lesion was excised with a 5-mm margin. The patient tolerated an in-office procedure lasting about 45 minutes but would have struggled with a longer session with Mohs microsurgery. The postoperative period required limiting full use of his left hand for about 2 weeks.

Photos and text for Photo Rounds Friday courtesy of Jonathan Karnes, MD (copyright retained). Dr. Karnes is the medical director of MDFMR Dermatology Services, Augusta, ME.

References

1. Bradford PT. Skin cancer in skin of color. Dermatol Nurs. 2009;21:170-177, 206; quiz 178. 2. Renzi M Jr, Schimmel J, Decker A, et al. Management of skin cancer in the elderly. Dermatol Clin. 2019;37:279-286. doi: 10.1016/j.det.2019.02.003

References

1. Bradford PT. Skin cancer in skin of color. Dermatol Nurs. 2009;21:170-177, 206; quiz 178. 2. Renzi M Jr, Schimmel J, Decker A, et al. Management of skin cancer in the elderly. Dermatol Clin. 2019;37:279-286. doi: 10.1016/j.det.2019.02.003

Issue
The Journal of Family Practice - 71(7)
Issue
The Journal of Family Practice - 71(7)
Publications
Publications
Topics
Article Type
Display Headline
Fast growing hand lesion
Display Headline
Fast growing hand lesion
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 09/28/2022 - 13:00
Un-Gate On Date
Wed, 09/28/2022 - 13:00
Use ProPublica
CFC Schedule Remove Status
Wed, 09/28/2022 - 13:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

How well are we doing with adolescent vaccination?

Article Type
Changed
Fri, 10/14/2022 - 07:55
Display Headline
How well are we doing with adolescent vaccination?

Every year, the Centers for Disease Control and Prevention (CDC) conducts a national survey to provide an estimate of vaccination rates among adolescents ages 13 to 17 years. The results for 2021, published recently, illustrate the progress that we’ve made and the areas in which improvement is still needed; notably, human papillomavirus (HPV) vaccine is an example of both.1

First, what’s recommended? The CDC recommends the following vaccines at age 11 to 12 years: tetanus, diphtheria, and acellular pertussis vaccine (Tdap); HPV vaccine series (2 doses if the first dose is received prior to age 15 years; 3 doses if the first dose is received at age 15 years or older); and quadrivalent meningococcal conjugate vaccine (MenACWY). A second (booster) dose of MenACWY is recommended at age 16 years. Adolescents should also receive an annual influenza vaccine and a COVID-19 vaccine series.2

For adolescents not fully vaccinated in childhood, catch-up vaccination is recommended for hepatitis A (HepA); hepatitis B (HepB); measles, mumps, and rubella (MMR); and varicella (VAR).2

How are we doing? In 2021, 89.6% of adolescents had received ≥ 1 Tdap dose and 89.0% had received ≥ 1 MenACWY dose; both these rates remained stable from the year before. For HPV vaccine, 76.9% had received ≥ 1 dose (an increase of 1.8 percentage points from 2020); 61.7% were HPV vaccine “up to date” (an increase of 3.1 percentage points). The teen HPV vaccination rate has increased slowly but progressively since the first recommendation for routine HPV vaccination was made for females in 2006 and for males in 2011.1

Among those age 17 years, coverage with ≥ 2 MenACWY doses was 60.0% (an increase of 5.6 percentage points from 2020). Coverage was 85% for ≥ 2 HepA doses (an increase of 2.9 percentage points from 2020) and remained stable at > 90% for each of the following: ≥ 2 doses of MMR, ≥ 3 doses of HepB, and both VAR doses.1

Keeping the momentum. As a country, we continue to make progress at increasing vaccination rates among US adolescents—but there is still plenty of room for improvement. Family physicians should check vaccine status at each clinical encounter and encourage parents and caregivers to schedule future wellness and vaccine visits for these young patients. This may be especially important among adolescents who were due for and missed a vaccination during the COVID-19 pandemic.

References

1. Pingali C, Yankey D, Elam-Evans LD, et al. National vaccination coverage among adolescents aged 13-17 years—National Immunization Survey-Teen, United States, 2021. MMWR Morb Mortal Wkly Rep. 2022;71:1101-1108.

2. Wodi AP, Murthy N, Bernstein H, et al. Advisory Committee on Immunization Practices recommended immunization schedule for children and adolescents aged 18 years or younger—United States, 2022. MMWR Morb Mortal Wkly Rep. 2022;71:234-237.

Author and Disclosure Information

Doug Campos-Outcalt, MD, MPA, is a clinical professor at the University of Arizona College of Medicine and a senior lecturer with the University of Arizona College of Public Health. He’s also an assistant editor at The Journal of Family Practice.

The author is a paid consultant to the CDC’s Advisory Committee on Immunization Practices.

Issue
The Journal of Family Practice - 71(8)
Publications
Topics
Sections
Author and Disclosure Information

Doug Campos-Outcalt, MD, MPA, is a clinical professor at the University of Arizona College of Medicine and a senior lecturer with the University of Arizona College of Public Health. He’s also an assistant editor at The Journal of Family Practice.

The author is a paid consultant to the CDC’s Advisory Committee on Immunization Practices.

Author and Disclosure Information

Doug Campos-Outcalt, MD, MPA, is a clinical professor at the University of Arizona College of Medicine and a senior lecturer with the University of Arizona College of Public Health. He’s also an assistant editor at The Journal of Family Practice.

The author is a paid consultant to the CDC’s Advisory Committee on Immunization Practices.

Every year, the Centers for Disease Control and Prevention (CDC) conducts a national survey to provide an estimate of vaccination rates among adolescents ages 13 to 17 years. The results for 2021, published recently, illustrate the progress that we’ve made and the areas in which improvement is still needed; notably, human papillomavirus (HPV) vaccine is an example of both.1

First, what’s recommended? The CDC recommends the following vaccines at age 11 to 12 years: tetanus, diphtheria, and acellular pertussis vaccine (Tdap); HPV vaccine series (2 doses if the first dose is received prior to age 15 years; 3 doses if the first dose is received at age 15 years or older); and quadrivalent meningococcal conjugate vaccine (MenACWY). A second (booster) dose of MenACWY is recommended at age 16 years. Adolescents should also receive an annual influenza vaccine and a COVID-19 vaccine series.2

For adolescents not fully vaccinated in childhood, catch-up vaccination is recommended for hepatitis A (HepA); hepatitis B (HepB); measles, mumps, and rubella (MMR); and varicella (VAR).2

How are we doing? In 2021, 89.6% of adolescents had received ≥ 1 Tdap dose and 89.0% had received ≥ 1 MenACWY dose; both these rates remained stable from the year before. For HPV vaccine, 76.9% had received ≥ 1 dose (an increase of 1.8 percentage points from 2020); 61.7% were HPV vaccine “up to date” (an increase of 3.1 percentage points). The teen HPV vaccination rate has increased slowly but progressively since the first recommendation for routine HPV vaccination was made for females in 2006 and for males in 2011.1

Among those age 17 years, coverage with ≥ 2 MenACWY doses was 60.0% (an increase of 5.6 percentage points from 2020). Coverage was 85% for ≥ 2 HepA doses (an increase of 2.9 percentage points from 2020) and remained stable at > 90% for each of the following: ≥ 2 doses of MMR, ≥ 3 doses of HepB, and both VAR doses.1

Keeping the momentum. As a country, we continue to make progress at increasing vaccination rates among US adolescents—but there is still plenty of room for improvement. Family physicians should check vaccine status at each clinical encounter and encourage parents and caregivers to schedule future wellness and vaccine visits for these young patients. This may be especially important among adolescents who were due for and missed a vaccination during the COVID-19 pandemic.

Every year, the Centers for Disease Control and Prevention (CDC) conducts a national survey to provide an estimate of vaccination rates among adolescents ages 13 to 17 years. The results for 2021, published recently, illustrate the progress that we’ve made and the areas in which improvement is still needed; notably, human papillomavirus (HPV) vaccine is an example of both.1

First, what’s recommended? The CDC recommends the following vaccines at age 11 to 12 years: tetanus, diphtheria, and acellular pertussis vaccine (Tdap); HPV vaccine series (2 doses if the first dose is received prior to age 15 years; 3 doses if the first dose is received at age 15 years or older); and quadrivalent meningococcal conjugate vaccine (MenACWY). A second (booster) dose of MenACWY is recommended at age 16 years. Adolescents should also receive an annual influenza vaccine and a COVID-19 vaccine series.2

For adolescents not fully vaccinated in childhood, catch-up vaccination is recommended for hepatitis A (HepA); hepatitis B (HepB); measles, mumps, and rubella (MMR); and varicella (VAR).2

How are we doing? In 2021, 89.6% of adolescents had received ≥ 1 Tdap dose and 89.0% had received ≥ 1 MenACWY dose; both these rates remained stable from the year before. For HPV vaccine, 76.9% had received ≥ 1 dose (an increase of 1.8 percentage points from 2020); 61.7% were HPV vaccine “up to date” (an increase of 3.1 percentage points). The teen HPV vaccination rate has increased slowly but progressively since the first recommendation for routine HPV vaccination was made for females in 2006 and for males in 2011.1

Among those age 17 years, coverage with ≥ 2 MenACWY doses was 60.0% (an increase of 5.6 percentage points from 2020). Coverage was 85% for ≥ 2 HepA doses (an increase of 2.9 percentage points from 2020) and remained stable at > 90% for each of the following: ≥ 2 doses of MMR, ≥ 3 doses of HepB, and both VAR doses.1

Keeping the momentum. As a country, we continue to make progress at increasing vaccination rates among US adolescents—but there is still plenty of room for improvement. Family physicians should check vaccine status at each clinical encounter and encourage parents and caregivers to schedule future wellness and vaccine visits for these young patients. This may be especially important among adolescents who were due for and missed a vaccination during the COVID-19 pandemic.

References

1. Pingali C, Yankey D, Elam-Evans LD, et al. National vaccination coverage among adolescents aged 13-17 years—National Immunization Survey-Teen, United States, 2021. MMWR Morb Mortal Wkly Rep. 2022;71:1101-1108.

2. Wodi AP, Murthy N, Bernstein H, et al. Advisory Committee on Immunization Practices recommended immunization schedule for children and adolescents aged 18 years or younger—United States, 2022. MMWR Morb Mortal Wkly Rep. 2022;71:234-237.

References

1. Pingali C, Yankey D, Elam-Evans LD, et al. National vaccination coverage among adolescents aged 13-17 years—National Immunization Survey-Teen, United States, 2021. MMWR Morb Mortal Wkly Rep. 2022;71:1101-1108.

2. Wodi AP, Murthy N, Bernstein H, et al. Advisory Committee on Immunization Practices recommended immunization schedule for children and adolescents aged 18 years or younger—United States, 2022. MMWR Morb Mortal Wkly Rep. 2022;71:234-237.

Issue
The Journal of Family Practice - 71(8)
Issue
The Journal of Family Practice - 71(8)
Publications
Publications
Topics
Article Type
Display Headline
How well are we doing with adolescent vaccination?
Display Headline
How well are we doing with adolescent vaccination?
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 09/28/2022 - 13:00
Un-Gate On Date
Wed, 09/28/2022 - 13:00
Use ProPublica
CFC Schedule Remove Status
Wed, 09/28/2022 - 13:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Tender Nonhealing Lesion on the Leg

Article Type
Changed
Wed, 01/04/2023 - 10:25
Display Headline
Tender Nonhealing Lesion on the Leg

The Diagnosis: Calciphylaxis

Calciphylaxis is a rare life-threatening condition that most often is seen in patients with end-stage renal disease at a rate of 35 per 10,000 chronic dialysis patients.1 It less commonly has been described in nonuremic patients. The exact incidence of nonuremic calciphylaxis is unknown, but multiple risk factors have been identified, such as alcoholic liver disease, primary hyperparathyroidism, connective tissue diseases, and underlying malignancies. Other less common risk factors include type 2 diabetes mellitus, hypercoagulable disorders, obesity, hypoalbuminemia, and warfarin/ corticosteroid use.2 However, most often no obvious triggers are identified.1

Regardless of the etiology, calciphylaxis is characterized by the calcification of blood vessels and connective tissues, leading to vessel injury, intimal fibrosis, and thrombosis, followed by ischemic necrosis of the skin and soft tissue. It is postulated that microvascular calcification occurs as an active cell-mediated process that depends on the balance between the promoters and inhibitors of calcification.1 In our patient, liver disease likely predisposed formation of calcification through the creation of an environment susceptible to vascular injury via decreased synthesis of proteins C and S.3 Synthesis of fetuin-A, a protein that acts as a circulating inhibitor of vascular ossification/calcification, also is decreased in calcification. Another inhibitor of calcification, matrix Gla protein, is unable to undergo activation through vitamin K–dependent carboxylation secondary to liver disease–induced vitamin K deficiency.3 Microvascular calcification without calciphylaxis may occur in other conditions such as type 2 diabetes mellitus. Therefore, clinicopathologic correlation is important in determining the diagnosis.

Calciphylaxis has a variety of clinical presentations depending on the stage of disease. It begins as a fixed, indurated, livedo reticularis–like plaque. The lesions become increasingly violaceous with intermixed areas of light blanched skin secondary to ischemia and then develop retiform pupura.4 Eventually, affected sites can become bullous and ulcerate or form a necrotic eschar. Severe pain is a cardinal feature throughout all stages.4 Lesions in nonuremic calciphylaxis most commonly are located in the central and/or proximal areas of the body.2

Clinical suspicion is essential for diagnosis. Skin biopsy is the standard method for confirmation in unclear cases. The classic histologic features include intravascular and extravascular calcification, microthrombosis, and fibrointimal hyperplasia of the small dermal and subcutaneous arteries and arterioles, leading to ischemia and intense septal panniculitis.1 Von Kossa immunostaining is used to increase the detection of calcium deposits (Figure 1).1 In addition to the classic changes, our case demonstrated a rare histologic variant with pseudoxanthoma elasticum (PXE)–like changes (Figure 2), which are thought to occur secondary to pathologic elastin fibrogenesis or increased proteolytic activity resulting in abnormal remodeling of the extracellular matrix in the setting of increased calcification of elastin fibers.5 Detection of PXE-like changes may be a helpful clue when specimens lack other characteristic signs.

Von Kossa immunostaining showed calcifications in the subcutis that appeared black (original magnification ×200).
FIGURE 1. Von Kossa immunostaining showed calcifications in the subcutis that appeared black (original magnification ×200).

Wound care, pain control, and addressing underlying causes are mainstays of therapy. Sodium thiosulfate, an antioxidant with vasodilatory properties that also inhibits adipocyte calcification and blocks the ability of adipocytes to induce calcification of vascular smooth-muscle cells, also is useful. Antibiotic prophylaxis is not indicated.1

Scattered purple calcifications were interspersed in both dermal collagen and subcutaneous adipose tissue with pseudoxanthoma elasticum–like changes (H&E, original magnification ×100).
FIGURE 2. Scattered purple calcifications were interspersed in both dermal collagen and subcutaneous adipose tissue with pseudoxanthoma elasticum–like changes (H&E, original magnification ×100).

Even with treatment, both uremic and nonuremic calciphylaxis have a dismal prognosis; 1-year mortality is approximately 50% to 60% and rises to 80% at 2 years.4 Lesion location affects prognosis, and more proximal lesions portend worse outcomes. In patients with both proximal and distal lesions, there is a 90% mortality rate within 1 year. Ulceration also portends worse outcomes, as the wounds often are resistant to healing and act as nidi for infection.4 Septicemia is the most common cause of death.1

Ecthyma gangrenosum is a cutaneous manifestation secondary to an infection most commonly associated with Pseudomonas aeruginosa.6 It often presents in immunocompromised patients with an underlying gramnegative septicemia.7 The clinical presentation initially begins with painless macules that rapidly progress into necrotic ulcers, usually accompanied by associated systemic symptoms such as fever, chills, and hypotension. Histopathology reveals numerous gram-negative rods around necrotic vessels.7

Idiopathic purpura fulminans is the rarest form of purpura fulminans. It is caused by autoantibody formation against protein S, resulting in protein S depletion and subsequent hypercoagulability.8 It usually occurs 7 to 10 days after the onset of a precipitating infection. Lesions begin as erythematous macules that progress within hours to painful, sharply defined areas of purpura and hemorrhagic cutaneous necrosis that may extend to deeper tissues.8 Secondary infection of gangrenous tissue may occur. Distribution usually is diffuse and signs of septic shock and disseminated intravascular coagulation usually are present.

Hughes syndrome, also known as antiphospholipid syndrome, is an acquired autoimmune disorder that manifests clinically as recurrent arterial or venous thrombosis.9 Cutaneous manifestations consist of livedo reticularis, arterial and venous ulcers, and superficial thrombophlebitis.10 Laboratory testing for antiphospholipid antibodies and obtaining a detailed history of the patient’s cardiovascular health are crucial for diagnosis.9

Necrotizing fasciitis typically begins as an inconspicuous superficial cutaneous infection that rapidly is transmitted to the fascia. Infection can spread along fascial planes for several days without affecting the overlying skin, leading to delayed diagnosis.11 The first signs to appear are disproportionate pain and a change in skin color to reddish-purple or bluish-gray. Next, the skin will become indurated, swollen, shiny, and more painful.11 Skin breakdown will begin in 3 to 5 days and is accompanied by bullae and cutaneous gangrene. The involved area becomes painless due to thrombosis of the small vessels that supply the superficial nerves.12 Septic shock ultimately will develop if untreated.

We present a rare case of nonuremic calciphylaxis. We encourage dermatologists to include calciphylaxis in the differential when evaluating any patient with a painful retiform rash or ulcerated eschar, even in the absence of renal disease.

References
  1. Nigwekar SU, Thadhani R, Brandenburg VM. Calciphylaxis. N Engl J Med. 2018;378:1704-1714.
  2. Nigwekar SU, Wolf M, Sterns RH, et al. Calciphylaxis from nonuremic causes: a systematic review. Clin J Am Soc Nephrol. 2008;3:1139-1143.
  3. Sammour YM, Saleh HM, Gad MM, et al. Non-uremic calciphylaxis associated with alcoholic hepatitis: a case report. World J Hepatol. 2019;11:127-132.
  4. James WD, Elston DM, Treat J, et al, eds. Cutaneous vascular diseases. Andrews’ Diseases of the Skin: Clinical Dermatology. Elsevier; 2020:813-861.
  5. Nathoo RK, Harb JN, Auerbach J, et al. Pseudoxanthoma elasticum-like changes in nonuremic calciphylaxis: case series and brief review of a helpful diagnostic clue. J Cutan Pathol. 2017;44:1064-1069.
  6. Vaiman M, Lazarovitch T, Heller L, et al. Ecthyma gangrenosum and ecthyma-like lesions: review article. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2015;34:633-639.
  7. Greene SL, Su WP, Muller SA. Ecthyma gangrenosum: report of clinical, histopathologic, and bacteriologic aspects of eight cases. J Am Acad Dermatol. 1984;11(5 pt 1):781-787.
  8. Levin M, Eley BS, Louis J, et al. Postinfectious purpura fulminans caused by an autoantibody directed against protein S. J Pediatr. 1995;127:355-363.
  9. Hughes G. Hughes syndrome: the antiphospholipid syndrome—a clinical overview. Clin Rev Allergy Immunol. 2007;32:3-12.
  10. Chang Y, Dabiri G, Damstetter E, et al. Coagulation disorders and their cutaneous presentations: pathophysiology. J Am Acad Dermatol. 2016;74:783-792; quiz 793-794.
  11. Fais P, Viero A, Viel G, et al. Necrotizing fasciitis: case series and review of the literature on clinical and medico-legal diagnostic challenges. Int J Legal Med. 2018;132:1357-1366.
  12. Brook I. Microbiology and management of soft tissue and muscle infections. Int J Surg Lond Engl. 2008;6:328-338.
Article PDF
Author and Disclosure Information

Drs. Miller and Tyring are from the Center for Clinical Studies, Webster, Texas. Dr. Tyring also is from the Department of Dermatology, University of Texas Health Science Center, Houston. Dr. Shalabi is from Texas A&M College of Medicine, Dallas. Dr. Rosenthal is from Atlanta Dermatopathology, Georgia. Dr. Cohen is from Skin Care Physicians of Georgia, Macon.

The authors report no conflict of interest.

Correspondence: Austinn C. Miller, MD, Center for Clinical Studies, 451 N Texas Ave, Webster, TX 77598 ([email protected]).

Issue
Cutis - 110(3)
Publications
Topics
Page Number
E7-E9
Sections
Author and Disclosure Information

Drs. Miller and Tyring are from the Center for Clinical Studies, Webster, Texas. Dr. Tyring also is from the Department of Dermatology, University of Texas Health Science Center, Houston. Dr. Shalabi is from Texas A&M College of Medicine, Dallas. Dr. Rosenthal is from Atlanta Dermatopathology, Georgia. Dr. Cohen is from Skin Care Physicians of Georgia, Macon.

The authors report no conflict of interest.

Correspondence: Austinn C. Miller, MD, Center for Clinical Studies, 451 N Texas Ave, Webster, TX 77598 ([email protected]).

Author and Disclosure Information

Drs. Miller and Tyring are from the Center for Clinical Studies, Webster, Texas. Dr. Tyring also is from the Department of Dermatology, University of Texas Health Science Center, Houston. Dr. Shalabi is from Texas A&M College of Medicine, Dallas. Dr. Rosenthal is from Atlanta Dermatopathology, Georgia. Dr. Cohen is from Skin Care Physicians of Georgia, Macon.

The authors report no conflict of interest.

Correspondence: Austinn C. Miller, MD, Center for Clinical Studies, 451 N Texas Ave, Webster, TX 77598 ([email protected]).

Article PDF
Article PDF
Related Articles

The Diagnosis: Calciphylaxis

Calciphylaxis is a rare life-threatening condition that most often is seen in patients with end-stage renal disease at a rate of 35 per 10,000 chronic dialysis patients.1 It less commonly has been described in nonuremic patients. The exact incidence of nonuremic calciphylaxis is unknown, but multiple risk factors have been identified, such as alcoholic liver disease, primary hyperparathyroidism, connective tissue diseases, and underlying malignancies. Other less common risk factors include type 2 diabetes mellitus, hypercoagulable disorders, obesity, hypoalbuminemia, and warfarin/ corticosteroid use.2 However, most often no obvious triggers are identified.1

Regardless of the etiology, calciphylaxis is characterized by the calcification of blood vessels and connective tissues, leading to vessel injury, intimal fibrosis, and thrombosis, followed by ischemic necrosis of the skin and soft tissue. It is postulated that microvascular calcification occurs as an active cell-mediated process that depends on the balance between the promoters and inhibitors of calcification.1 In our patient, liver disease likely predisposed formation of calcification through the creation of an environment susceptible to vascular injury via decreased synthesis of proteins C and S.3 Synthesis of fetuin-A, a protein that acts as a circulating inhibitor of vascular ossification/calcification, also is decreased in calcification. Another inhibitor of calcification, matrix Gla protein, is unable to undergo activation through vitamin K–dependent carboxylation secondary to liver disease–induced vitamin K deficiency.3 Microvascular calcification without calciphylaxis may occur in other conditions such as type 2 diabetes mellitus. Therefore, clinicopathologic correlation is important in determining the diagnosis.

Calciphylaxis has a variety of clinical presentations depending on the stage of disease. It begins as a fixed, indurated, livedo reticularis–like plaque. The lesions become increasingly violaceous with intermixed areas of light blanched skin secondary to ischemia and then develop retiform pupura.4 Eventually, affected sites can become bullous and ulcerate or form a necrotic eschar. Severe pain is a cardinal feature throughout all stages.4 Lesions in nonuremic calciphylaxis most commonly are located in the central and/or proximal areas of the body.2

Clinical suspicion is essential for diagnosis. Skin biopsy is the standard method for confirmation in unclear cases. The classic histologic features include intravascular and extravascular calcification, microthrombosis, and fibrointimal hyperplasia of the small dermal and subcutaneous arteries and arterioles, leading to ischemia and intense septal panniculitis.1 Von Kossa immunostaining is used to increase the detection of calcium deposits (Figure 1).1 In addition to the classic changes, our case demonstrated a rare histologic variant with pseudoxanthoma elasticum (PXE)–like changes (Figure 2), which are thought to occur secondary to pathologic elastin fibrogenesis or increased proteolytic activity resulting in abnormal remodeling of the extracellular matrix in the setting of increased calcification of elastin fibers.5 Detection of PXE-like changes may be a helpful clue when specimens lack other characteristic signs.

Von Kossa immunostaining showed calcifications in the subcutis that appeared black (original magnification ×200).
FIGURE 1. Von Kossa immunostaining showed calcifications in the subcutis that appeared black (original magnification ×200).

Wound care, pain control, and addressing underlying causes are mainstays of therapy. Sodium thiosulfate, an antioxidant with vasodilatory properties that also inhibits adipocyte calcification and blocks the ability of adipocytes to induce calcification of vascular smooth-muscle cells, also is useful. Antibiotic prophylaxis is not indicated.1

Scattered purple calcifications were interspersed in both dermal collagen and subcutaneous adipose tissue with pseudoxanthoma elasticum–like changes (H&E, original magnification ×100).
FIGURE 2. Scattered purple calcifications were interspersed in both dermal collagen and subcutaneous adipose tissue with pseudoxanthoma elasticum–like changes (H&E, original magnification ×100).

Even with treatment, both uremic and nonuremic calciphylaxis have a dismal prognosis; 1-year mortality is approximately 50% to 60% and rises to 80% at 2 years.4 Lesion location affects prognosis, and more proximal lesions portend worse outcomes. In patients with both proximal and distal lesions, there is a 90% mortality rate within 1 year. Ulceration also portends worse outcomes, as the wounds often are resistant to healing and act as nidi for infection.4 Septicemia is the most common cause of death.1

Ecthyma gangrenosum is a cutaneous manifestation secondary to an infection most commonly associated with Pseudomonas aeruginosa.6 It often presents in immunocompromised patients with an underlying gramnegative septicemia.7 The clinical presentation initially begins with painless macules that rapidly progress into necrotic ulcers, usually accompanied by associated systemic symptoms such as fever, chills, and hypotension. Histopathology reveals numerous gram-negative rods around necrotic vessels.7

Idiopathic purpura fulminans is the rarest form of purpura fulminans. It is caused by autoantibody formation against protein S, resulting in protein S depletion and subsequent hypercoagulability.8 It usually occurs 7 to 10 days after the onset of a precipitating infection. Lesions begin as erythematous macules that progress within hours to painful, sharply defined areas of purpura and hemorrhagic cutaneous necrosis that may extend to deeper tissues.8 Secondary infection of gangrenous tissue may occur. Distribution usually is diffuse and signs of septic shock and disseminated intravascular coagulation usually are present.

Hughes syndrome, also known as antiphospholipid syndrome, is an acquired autoimmune disorder that manifests clinically as recurrent arterial or venous thrombosis.9 Cutaneous manifestations consist of livedo reticularis, arterial and venous ulcers, and superficial thrombophlebitis.10 Laboratory testing for antiphospholipid antibodies and obtaining a detailed history of the patient’s cardiovascular health are crucial for diagnosis.9

Necrotizing fasciitis typically begins as an inconspicuous superficial cutaneous infection that rapidly is transmitted to the fascia. Infection can spread along fascial planes for several days without affecting the overlying skin, leading to delayed diagnosis.11 The first signs to appear are disproportionate pain and a change in skin color to reddish-purple or bluish-gray. Next, the skin will become indurated, swollen, shiny, and more painful.11 Skin breakdown will begin in 3 to 5 days and is accompanied by bullae and cutaneous gangrene. The involved area becomes painless due to thrombosis of the small vessels that supply the superficial nerves.12 Septic shock ultimately will develop if untreated.

We present a rare case of nonuremic calciphylaxis. We encourage dermatologists to include calciphylaxis in the differential when evaluating any patient with a painful retiform rash or ulcerated eschar, even in the absence of renal disease.

The Diagnosis: Calciphylaxis

Calciphylaxis is a rare life-threatening condition that most often is seen in patients with end-stage renal disease at a rate of 35 per 10,000 chronic dialysis patients.1 It less commonly has been described in nonuremic patients. The exact incidence of nonuremic calciphylaxis is unknown, but multiple risk factors have been identified, such as alcoholic liver disease, primary hyperparathyroidism, connective tissue diseases, and underlying malignancies. Other less common risk factors include type 2 diabetes mellitus, hypercoagulable disorders, obesity, hypoalbuminemia, and warfarin/ corticosteroid use.2 However, most often no obvious triggers are identified.1

Regardless of the etiology, calciphylaxis is characterized by the calcification of blood vessels and connective tissues, leading to vessel injury, intimal fibrosis, and thrombosis, followed by ischemic necrosis of the skin and soft tissue. It is postulated that microvascular calcification occurs as an active cell-mediated process that depends on the balance between the promoters and inhibitors of calcification.1 In our patient, liver disease likely predisposed formation of calcification through the creation of an environment susceptible to vascular injury via decreased synthesis of proteins C and S.3 Synthesis of fetuin-A, a protein that acts as a circulating inhibitor of vascular ossification/calcification, also is decreased in calcification. Another inhibitor of calcification, matrix Gla protein, is unable to undergo activation through vitamin K–dependent carboxylation secondary to liver disease–induced vitamin K deficiency.3 Microvascular calcification without calciphylaxis may occur in other conditions such as type 2 diabetes mellitus. Therefore, clinicopathologic correlation is important in determining the diagnosis.

Calciphylaxis has a variety of clinical presentations depending on the stage of disease. It begins as a fixed, indurated, livedo reticularis–like plaque. The lesions become increasingly violaceous with intermixed areas of light blanched skin secondary to ischemia and then develop retiform pupura.4 Eventually, affected sites can become bullous and ulcerate or form a necrotic eschar. Severe pain is a cardinal feature throughout all stages.4 Lesions in nonuremic calciphylaxis most commonly are located in the central and/or proximal areas of the body.2

Clinical suspicion is essential for diagnosis. Skin biopsy is the standard method for confirmation in unclear cases. The classic histologic features include intravascular and extravascular calcification, microthrombosis, and fibrointimal hyperplasia of the small dermal and subcutaneous arteries and arterioles, leading to ischemia and intense septal panniculitis.1 Von Kossa immunostaining is used to increase the detection of calcium deposits (Figure 1).1 In addition to the classic changes, our case demonstrated a rare histologic variant with pseudoxanthoma elasticum (PXE)–like changes (Figure 2), which are thought to occur secondary to pathologic elastin fibrogenesis or increased proteolytic activity resulting in abnormal remodeling of the extracellular matrix in the setting of increased calcification of elastin fibers.5 Detection of PXE-like changes may be a helpful clue when specimens lack other characteristic signs.

Von Kossa immunostaining showed calcifications in the subcutis that appeared black (original magnification ×200).
FIGURE 1. Von Kossa immunostaining showed calcifications in the subcutis that appeared black (original magnification ×200).

Wound care, pain control, and addressing underlying causes are mainstays of therapy. Sodium thiosulfate, an antioxidant with vasodilatory properties that also inhibits adipocyte calcification and blocks the ability of adipocytes to induce calcification of vascular smooth-muscle cells, also is useful. Antibiotic prophylaxis is not indicated.1

Scattered purple calcifications were interspersed in both dermal collagen and subcutaneous adipose tissue with pseudoxanthoma elasticum–like changes (H&E, original magnification ×100).
FIGURE 2. Scattered purple calcifications were interspersed in both dermal collagen and subcutaneous adipose tissue with pseudoxanthoma elasticum–like changes (H&E, original magnification ×100).

Even with treatment, both uremic and nonuremic calciphylaxis have a dismal prognosis; 1-year mortality is approximately 50% to 60% and rises to 80% at 2 years.4 Lesion location affects prognosis, and more proximal lesions portend worse outcomes. In patients with both proximal and distal lesions, there is a 90% mortality rate within 1 year. Ulceration also portends worse outcomes, as the wounds often are resistant to healing and act as nidi for infection.4 Septicemia is the most common cause of death.1

Ecthyma gangrenosum is a cutaneous manifestation secondary to an infection most commonly associated with Pseudomonas aeruginosa.6 It often presents in immunocompromised patients with an underlying gramnegative septicemia.7 The clinical presentation initially begins with painless macules that rapidly progress into necrotic ulcers, usually accompanied by associated systemic symptoms such as fever, chills, and hypotension. Histopathology reveals numerous gram-negative rods around necrotic vessels.7

Idiopathic purpura fulminans is the rarest form of purpura fulminans. It is caused by autoantibody formation against protein S, resulting in protein S depletion and subsequent hypercoagulability.8 It usually occurs 7 to 10 days after the onset of a precipitating infection. Lesions begin as erythematous macules that progress within hours to painful, sharply defined areas of purpura and hemorrhagic cutaneous necrosis that may extend to deeper tissues.8 Secondary infection of gangrenous tissue may occur. Distribution usually is diffuse and signs of septic shock and disseminated intravascular coagulation usually are present.

Hughes syndrome, also known as antiphospholipid syndrome, is an acquired autoimmune disorder that manifests clinically as recurrent arterial or venous thrombosis.9 Cutaneous manifestations consist of livedo reticularis, arterial and venous ulcers, and superficial thrombophlebitis.10 Laboratory testing for antiphospholipid antibodies and obtaining a detailed history of the patient’s cardiovascular health are crucial for diagnosis.9

Necrotizing fasciitis typically begins as an inconspicuous superficial cutaneous infection that rapidly is transmitted to the fascia. Infection can spread along fascial planes for several days without affecting the overlying skin, leading to delayed diagnosis.11 The first signs to appear are disproportionate pain and a change in skin color to reddish-purple or bluish-gray. Next, the skin will become indurated, swollen, shiny, and more painful.11 Skin breakdown will begin in 3 to 5 days and is accompanied by bullae and cutaneous gangrene. The involved area becomes painless due to thrombosis of the small vessels that supply the superficial nerves.12 Septic shock ultimately will develop if untreated.

We present a rare case of nonuremic calciphylaxis. We encourage dermatologists to include calciphylaxis in the differential when evaluating any patient with a painful retiform rash or ulcerated eschar, even in the absence of renal disease.

References
  1. Nigwekar SU, Thadhani R, Brandenburg VM. Calciphylaxis. N Engl J Med. 2018;378:1704-1714.
  2. Nigwekar SU, Wolf M, Sterns RH, et al. Calciphylaxis from nonuremic causes: a systematic review. Clin J Am Soc Nephrol. 2008;3:1139-1143.
  3. Sammour YM, Saleh HM, Gad MM, et al. Non-uremic calciphylaxis associated with alcoholic hepatitis: a case report. World J Hepatol. 2019;11:127-132.
  4. James WD, Elston DM, Treat J, et al, eds. Cutaneous vascular diseases. Andrews’ Diseases of the Skin: Clinical Dermatology. Elsevier; 2020:813-861.
  5. Nathoo RK, Harb JN, Auerbach J, et al. Pseudoxanthoma elasticum-like changes in nonuremic calciphylaxis: case series and brief review of a helpful diagnostic clue. J Cutan Pathol. 2017;44:1064-1069.
  6. Vaiman M, Lazarovitch T, Heller L, et al. Ecthyma gangrenosum and ecthyma-like lesions: review article. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2015;34:633-639.
  7. Greene SL, Su WP, Muller SA. Ecthyma gangrenosum: report of clinical, histopathologic, and bacteriologic aspects of eight cases. J Am Acad Dermatol. 1984;11(5 pt 1):781-787.
  8. Levin M, Eley BS, Louis J, et al. Postinfectious purpura fulminans caused by an autoantibody directed against protein S. J Pediatr. 1995;127:355-363.
  9. Hughes G. Hughes syndrome: the antiphospholipid syndrome—a clinical overview. Clin Rev Allergy Immunol. 2007;32:3-12.
  10. Chang Y, Dabiri G, Damstetter E, et al. Coagulation disorders and their cutaneous presentations: pathophysiology. J Am Acad Dermatol. 2016;74:783-792; quiz 793-794.
  11. Fais P, Viero A, Viel G, et al. Necrotizing fasciitis: case series and review of the literature on clinical and medico-legal diagnostic challenges. Int J Legal Med. 2018;132:1357-1366.
  12. Brook I. Microbiology and management of soft tissue and muscle infections. Int J Surg Lond Engl. 2008;6:328-338.
References
  1. Nigwekar SU, Thadhani R, Brandenburg VM. Calciphylaxis. N Engl J Med. 2018;378:1704-1714.
  2. Nigwekar SU, Wolf M, Sterns RH, et al. Calciphylaxis from nonuremic causes: a systematic review. Clin J Am Soc Nephrol. 2008;3:1139-1143.
  3. Sammour YM, Saleh HM, Gad MM, et al. Non-uremic calciphylaxis associated with alcoholic hepatitis: a case report. World J Hepatol. 2019;11:127-132.
  4. James WD, Elston DM, Treat J, et al, eds. Cutaneous vascular diseases. Andrews’ Diseases of the Skin: Clinical Dermatology. Elsevier; 2020:813-861.
  5. Nathoo RK, Harb JN, Auerbach J, et al. Pseudoxanthoma elasticum-like changes in nonuremic calciphylaxis: case series and brief review of a helpful diagnostic clue. J Cutan Pathol. 2017;44:1064-1069.
  6. Vaiman M, Lazarovitch T, Heller L, et al. Ecthyma gangrenosum and ecthyma-like lesions: review article. Eur J Clin Microbiol Infect Dis Off Publ Eur Soc Clin Microbiol. 2015;34:633-639.
  7. Greene SL, Su WP, Muller SA. Ecthyma gangrenosum: report of clinical, histopathologic, and bacteriologic aspects of eight cases. J Am Acad Dermatol. 1984;11(5 pt 1):781-787.
  8. Levin M, Eley BS, Louis J, et al. Postinfectious purpura fulminans caused by an autoantibody directed against protein S. J Pediatr. 1995;127:355-363.
  9. Hughes G. Hughes syndrome: the antiphospholipid syndrome—a clinical overview. Clin Rev Allergy Immunol. 2007;32:3-12.
  10. Chang Y, Dabiri G, Damstetter E, et al. Coagulation disorders and their cutaneous presentations: pathophysiology. J Am Acad Dermatol. 2016;74:783-792; quiz 793-794.
  11. Fais P, Viero A, Viel G, et al. Necrotizing fasciitis: case series and review of the literature on clinical and medico-legal diagnostic challenges. Int J Legal Med. 2018;132:1357-1366.
  12. Brook I. Microbiology and management of soft tissue and muscle infections. Int J Surg Lond Engl. 2008;6:328-338.
Issue
Cutis - 110(3)
Issue
Cutis - 110(3)
Page Number
E7-E9
Page Number
E7-E9
Publications
Publications
Topics
Article Type
Display Headline
Tender Nonhealing Lesion on the Leg
Display Headline
Tender Nonhealing Lesion on the Leg
Sections
Questionnaire Body

A 50-year-old woman presented to our dermatology clinic with an exquisitely tender, nonhealing lesion on the left leg of 2 weeks’ duration that began as a small red-purplish spot. She applied a triple antibiotic ointment and wrapped the area with gauze daily but reported that it continued to enlarge and darken in color before forming a “scab.” She noted occasional seropurulent discharge and denied any trauma or new exposures to the area. She was seen at a local emergency department 3 days prior to presentation and was prescribed oral clindamycin for suspected cellulitis, but she denied any improvement with the initiation of antibiotics. Her medical history was notable for obesity, depression, hypothyroidism, and liver disease secondary to alcohol use disorder. She reported that she drank a pint of vodka daily. Her medications included pantoprazole, spironolactone, bumetanide, citalopram, levothyroxine, naltrexone, tramadol, and a multivitamin. Physical examination revealed violaceous mottling with areas of superficial erythema and ulceration with necrotic eschars on the proximal left thigh that were extremely painful. A biopsy was obtained for confirmation of diagnosis, but the patient died before the results were returned.

Tender nonhealing lesion on the leg

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 09/28/2022 - 11:30
Un-Gate On Date
Wed, 09/28/2022 - 11:30
Use ProPublica
CFC Schedule Remove Status
Wed, 09/28/2022 - 11:30
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Article PDF Media

Commentary: Evaluating HCC Treatments, October 2022

Article Type
Changed
Wed, 09/28/2022 - 10:52
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD
Patients with hepatocellular carcinoma (HCC) may benefit from liver-directed therapies or palliative systemic therapies. This month we will review articles that address management of patients who were treated with different standard treatment options.

Komatsu and colleagues performed a case-matched analysis to evaluate the best first-line treatment for HCC in patients with macroscopic portal vein tumor thrombus (PVTT). Patients had advanced HCC and macroscopic PVTT that invaded an ipsilateral first-order portal branch, main trunk, or contralateral portal vein. The propensity score–matched groups underwent either hepatectomy (n = 36) or received sorafenib (n = 36). To be considered for resection, patients had to have Child-Pugh (CP) grade A or B liver function, an Eastern Cooperative Oncology Group Performance Status score of ≤ 1, life expectancy of > 3 months, and the macroscopic resection of the targeted tumor could be planned with an estimated remnant liver volume ≥ 35%.

Out of 36 patients who underwent surgery, 23 underwent reductive hepatectomy, and 13 underwent complete resection of tumor. Out of 36 patients who received sorafenib, 21 underwent subsequent treatments. The median overall survival (OS) of patients who underwent hepatectomy was 15.1 months, significantly longer than the 4.5 months for patients who were treated with sorafenib. The authors concluded that selected patients who underwent tumor resection lived longer than patients who received systemic therapy with sorafenib first, despite the presence of macroscopic PVTT.

Lenvatinib is an approved treatment for patients with HCC. Because most patients with HCC have underlying cirrhosis, monitoring the underlying liver function is an important facet of patient management. Huynh and colleagues reported on patients in the REFLECT trial whose liver function deteriorated from CP-A to CP-B while receiving systemic therapy. This post hoc analysis included patients whose liver function deteriorated to CP-B or remained CP-A within 8 weeks of randomization to lenvatinib (CP-B: n = 60; CP-A: n = 413) or sorafenib (CP-B: n = 47; CP-A: n = 427). Patients receiving lenvatinib who developed CP-B cirrhosis compared with patients who maintained CP-A cirrhosis had a median progression-free survival (PFS) of 3.7 months (95% CI 1.8-7.4) vs 6.5 months (95% CI 5.6-7.4) and OS of 6.8 months (95% CI 2.6-10.3) vs 13.3 months (95% CI 11.6-16.1). CP-B patients receiving sorafenib had a median PFS and OS of only 0.5 months (95% CI 0.1-3.6) and 4.5 months (95% CI 2.9-6.1), respectively. No new safety signals were reported in CP-B patients. The investigators concluded that deterioration of liver function to CP-B does not require the discontinuation of lenvatinib therapy.

Finally, Brown and colleagues performed a meta-analysis of studies that evaluated transarterial radioembolization (TARE) and transarterial chemoembolization (TACE) in patients with HCC by reviewing 17 studies involving 2465 patients that directly compared TACE and TARE. TARE significantly prolonged the mean time to progression (17.5 vs 9.8 months; 95% CI 1.3-8.3 months) but resulted in comparable OS (absolute difference −0.55 months; 95% CI −1.95 to 3.05 months). Safety profiles appeared to favor TARE. The authors concluded that TACE and TARE should be compared in larger prospective studies to better compare survival, progression, and safety data.

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Publications
Topics
Sections
Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Dr. Damjanov scans the journals, so you don’t have to!
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD
Patients with hepatocellular carcinoma (HCC) may benefit from liver-directed therapies or palliative systemic therapies. This month we will review articles that address management of patients who were treated with different standard treatment options.

Komatsu and colleagues performed a case-matched analysis to evaluate the best first-line treatment for HCC in patients with macroscopic portal vein tumor thrombus (PVTT). Patients had advanced HCC and macroscopic PVTT that invaded an ipsilateral first-order portal branch, main trunk, or contralateral portal vein. The propensity score–matched groups underwent either hepatectomy (n = 36) or received sorafenib (n = 36). To be considered for resection, patients had to have Child-Pugh (CP) grade A or B liver function, an Eastern Cooperative Oncology Group Performance Status score of ≤ 1, life expectancy of > 3 months, and the macroscopic resection of the targeted tumor could be planned with an estimated remnant liver volume ≥ 35%.

Out of 36 patients who underwent surgery, 23 underwent reductive hepatectomy, and 13 underwent complete resection of tumor. Out of 36 patients who received sorafenib, 21 underwent subsequent treatments. The median overall survival (OS) of patients who underwent hepatectomy was 15.1 months, significantly longer than the 4.5 months for patients who were treated with sorafenib. The authors concluded that selected patients who underwent tumor resection lived longer than patients who received systemic therapy with sorafenib first, despite the presence of macroscopic PVTT.

Lenvatinib is an approved treatment for patients with HCC. Because most patients with HCC have underlying cirrhosis, monitoring the underlying liver function is an important facet of patient management. Huynh and colleagues reported on patients in the REFLECT trial whose liver function deteriorated from CP-A to CP-B while receiving systemic therapy. This post hoc analysis included patients whose liver function deteriorated to CP-B or remained CP-A within 8 weeks of randomization to lenvatinib (CP-B: n = 60; CP-A: n = 413) or sorafenib (CP-B: n = 47; CP-A: n = 427). Patients receiving lenvatinib who developed CP-B cirrhosis compared with patients who maintained CP-A cirrhosis had a median progression-free survival (PFS) of 3.7 months (95% CI 1.8-7.4) vs 6.5 months (95% CI 5.6-7.4) and OS of 6.8 months (95% CI 2.6-10.3) vs 13.3 months (95% CI 11.6-16.1). CP-B patients receiving sorafenib had a median PFS and OS of only 0.5 months (95% CI 0.1-3.6) and 4.5 months (95% CI 2.9-6.1), respectively. No new safety signals were reported in CP-B patients. The investigators concluded that deterioration of liver function to CP-B does not require the discontinuation of lenvatinib therapy.

Finally, Brown and colleagues performed a meta-analysis of studies that evaluated transarterial radioembolization (TARE) and transarterial chemoembolization (TACE) in patients with HCC by reviewing 17 studies involving 2465 patients that directly compared TACE and TARE. TARE significantly prolonged the mean time to progression (17.5 vs 9.8 months; 95% CI 1.3-8.3 months) but resulted in comparable OS (absolute difference −0.55 months; 95% CI −1.95 to 3.05 months). Safety profiles appeared to favor TARE. The authors concluded that TACE and TARE should be compared in larger prospective studies to better compare survival, progression, and safety data.

Nevena Damjanov, MD
Patients with hepatocellular carcinoma (HCC) may benefit from liver-directed therapies or palliative systemic therapies. This month we will review articles that address management of patients who were treated with different standard treatment options.

Komatsu and colleagues performed a case-matched analysis to evaluate the best first-line treatment for HCC in patients with macroscopic portal vein tumor thrombus (PVTT). Patients had advanced HCC and macroscopic PVTT that invaded an ipsilateral first-order portal branch, main trunk, or contralateral portal vein. The propensity score–matched groups underwent either hepatectomy (n = 36) or received sorafenib (n = 36). To be considered for resection, patients had to have Child-Pugh (CP) grade A or B liver function, an Eastern Cooperative Oncology Group Performance Status score of ≤ 1, life expectancy of > 3 months, and the macroscopic resection of the targeted tumor could be planned with an estimated remnant liver volume ≥ 35%.

Out of 36 patients who underwent surgery, 23 underwent reductive hepatectomy, and 13 underwent complete resection of tumor. Out of 36 patients who received sorafenib, 21 underwent subsequent treatments. The median overall survival (OS) of patients who underwent hepatectomy was 15.1 months, significantly longer than the 4.5 months for patients who were treated with sorafenib. The authors concluded that selected patients who underwent tumor resection lived longer than patients who received systemic therapy with sorafenib first, despite the presence of macroscopic PVTT.

Lenvatinib is an approved treatment for patients with HCC. Because most patients with HCC have underlying cirrhosis, monitoring the underlying liver function is an important facet of patient management. Huynh and colleagues reported on patients in the REFLECT trial whose liver function deteriorated from CP-A to CP-B while receiving systemic therapy. This post hoc analysis included patients whose liver function deteriorated to CP-B or remained CP-A within 8 weeks of randomization to lenvatinib (CP-B: n = 60; CP-A: n = 413) or sorafenib (CP-B: n = 47; CP-A: n = 427). Patients receiving lenvatinib who developed CP-B cirrhosis compared with patients who maintained CP-A cirrhosis had a median progression-free survival (PFS) of 3.7 months (95% CI 1.8-7.4) vs 6.5 months (95% CI 5.6-7.4) and OS of 6.8 months (95% CI 2.6-10.3) vs 13.3 months (95% CI 11.6-16.1). CP-B patients receiving sorafenib had a median PFS and OS of only 0.5 months (95% CI 0.1-3.6) and 4.5 months (95% CI 2.9-6.1), respectively. No new safety signals were reported in CP-B patients. The investigators concluded that deterioration of liver function to CP-B does not require the discontinuation of lenvatinib therapy.

Finally, Brown and colleagues performed a meta-analysis of studies that evaluated transarterial radioembolization (TARE) and transarterial chemoembolization (TACE) in patients with HCC by reviewing 17 studies involving 2465 patients that directly compared TACE and TARE. TARE significantly prolonged the mean time to progression (17.5 vs 9.8 months; 95% CI 1.3-8.3 months) but resulted in comparable OS (absolute difference −0.55 months; 95% CI −1.95 to 3.05 months). Safety profiles appeared to favor TARE. The authors concluded that TACE and TARE should be compared in larger prospective studies to better compare survival, progression, and safety data.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: HCC October 2022
Gate On Date
Thu, 01/20/2022 - 12:15
Un-Gate On Date
Thu, 01/20/2022 - 12:15
Use ProPublica
CFC Schedule Remove Status
Thu, 01/20/2022 - 12:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
332967.1
Activity ID
83146
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Exact Sciences Corporate [ 6025 ]

Commentary: Evaluating HCC Treatments, October 2022

Article Type
Changed
Wed, 09/28/2022 - 10:48
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD
Patients with hepatocellular carcinoma (HCC) may benefit from liver-directed therapies or palliative systemic therapies. This month we will review articles that address management of patients who were treated with different standard treatment options.

Komatsu and colleagues performed a case-matched analysis to evaluate the best first-line treatment for HCC in patients with macroscopic portal vein tumor thrombus (PVTT). Patients had advanced HCC and macroscopic PVTT that invaded an ipsilateral first-order portal branch, main trunk, or contralateral portal vein. The propensity score–matched groups underwent either hepatectomy (n = 36) or received sorafenib (n = 36). To be considered for resection, patients had to have Child-Pugh (CP) grade A or B liver function, an Eastern Cooperative Oncology Group Performance Status score of ≤ 1, life expectancy of > 3 months, and the macroscopic resection of the targeted tumor could be planned with an estimated remnant liver volume ≥ 35%.

Out of 36 patients who underwent surgery, 23 underwent reductive hepatectomy, and 13 underwent complete resection of tumor. Out of 36 patients who received sorafenib, 21 underwent subsequent treatments. The median overall survival (OS) of patients who underwent hepatectomy was 15.1 months, significantly longer than the 4.5 months for patients who were treated with sorafenib. The authors concluded that selected patients who underwent tumor resection lived longer than patients who received systemic therapy with sorafenib first, despite the presence of macroscopic PVTT.

Lenvatinib is an approved treatment for patients with HCC. Because most patients with HCC have underlying cirrhosis, monitoring the underlying liver function is an important facet of patient management. Huynh and colleagues reported on patients in the REFLECT trial whose liver function deteriorated from CP-A to CP-B while receiving systemic therapy. This post hoc analysis included patients whose liver function deteriorated to CP-B or remained CP-A within 8 weeks of randomization to lenvatinib (CP-B: n = 60; CP-A: n = 413) or sorafenib (CP-B: n = 47; CP-A: n = 427). Patients receiving lenvatinib who developed CP-B cirrhosis compared with patients who maintained CP-A cirrhosis had a median progression-free survival (PFS) of 3.7 months (95% CI 1.8-7.4) vs 6.5 months (95% CI 5.6-7.4) and OS of 6.8 months (95% CI 2.6-10.3) vs 13.3 months (95% CI 11.6-16.1). CP-B patients receiving sorafenib had a median PFS and OS of only 0.5 months (95% CI 0.1-3.6) and 4.5 months (95% CI 2.9-6.1), respectively. No new safety signals were reported in CP-B patients. The investigators concluded that deterioration of liver function to CP-B does not require the discontinuation of lenvatinib therapy.

Finally, Brown and colleagues performed a meta-analysis of studies that evaluated transarterial radioembolization (TARE) and transarterial chemoembolization (TACE) in patients with HCC by reviewing 17 studies involving 2465 patients that directly compared TACE and TARE. TARE significantly prolonged the mean time to progression (17.5 vs 9.8 months; 95% CI 1.3-8.3 months) but resulted in comparable OS (absolute difference −0.55 months; 95% CI −1.95 to 3.05 months). Safety profiles appeared to favor TARE. The authors concluded that TACE and TARE should be compared in larger prospective studies to better compare survival, progression, and safety data.

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Publications
Topics
Sections
Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Author and Disclosure Information

Nevena Damjanov, MD, Professor, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania; Chief, Department of Hematology-Oncology, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania

Nevena Damjanov, MD, has disclosed the following relevant financial relationships:

Serve(d) as a director, officer, partner, employee, advisor, consultant, or trustee for: QED; Eisai

Received research grant from: Basilea; Bristol-Myers Squibb; Merck

Dr. Damjanov scans the journals, so you don’t have to!
Dr. Damjanov scans the journals, so you don’t have to!

Nevena Damjanov, MD
Patients with hepatocellular carcinoma (HCC) may benefit from liver-directed therapies or palliative systemic therapies. This month we will review articles that address management of patients who were treated with different standard treatment options.

Komatsu and colleagues performed a case-matched analysis to evaluate the best first-line treatment for HCC in patients with macroscopic portal vein tumor thrombus (PVTT). Patients had advanced HCC and macroscopic PVTT that invaded an ipsilateral first-order portal branch, main trunk, or contralateral portal vein. The propensity score–matched groups underwent either hepatectomy (n = 36) or received sorafenib (n = 36). To be considered for resection, patients had to have Child-Pugh (CP) grade A or B liver function, an Eastern Cooperative Oncology Group Performance Status score of ≤ 1, life expectancy of > 3 months, and the macroscopic resection of the targeted tumor could be planned with an estimated remnant liver volume ≥ 35%.

Out of 36 patients who underwent surgery, 23 underwent reductive hepatectomy, and 13 underwent complete resection of tumor. Out of 36 patients who received sorafenib, 21 underwent subsequent treatments. The median overall survival (OS) of patients who underwent hepatectomy was 15.1 months, significantly longer than the 4.5 months for patients who were treated with sorafenib. The authors concluded that selected patients who underwent tumor resection lived longer than patients who received systemic therapy with sorafenib first, despite the presence of macroscopic PVTT.

Lenvatinib is an approved treatment for patients with HCC. Because most patients with HCC have underlying cirrhosis, monitoring the underlying liver function is an important facet of patient management. Huynh and colleagues reported on patients in the REFLECT trial whose liver function deteriorated from CP-A to CP-B while receiving systemic therapy. This post hoc analysis included patients whose liver function deteriorated to CP-B or remained CP-A within 8 weeks of randomization to lenvatinib (CP-B: n = 60; CP-A: n = 413) or sorafenib (CP-B: n = 47; CP-A: n = 427). Patients receiving lenvatinib who developed CP-B cirrhosis compared with patients who maintained CP-A cirrhosis had a median progression-free survival (PFS) of 3.7 months (95% CI 1.8-7.4) vs 6.5 months (95% CI 5.6-7.4) and OS of 6.8 months (95% CI 2.6-10.3) vs 13.3 months (95% CI 11.6-16.1). CP-B patients receiving sorafenib had a median PFS and OS of only 0.5 months (95% CI 0.1-3.6) and 4.5 months (95% CI 2.9-6.1), respectively. No new safety signals were reported in CP-B patients. The investigators concluded that deterioration of liver function to CP-B does not require the discontinuation of lenvatinib therapy.

Finally, Brown and colleagues performed a meta-analysis of studies that evaluated transarterial radioembolization (TARE) and transarterial chemoembolization (TACE) in patients with HCC by reviewing 17 studies involving 2465 patients that directly compared TACE and TARE. TARE significantly prolonged the mean time to progression (17.5 vs 9.8 months; 95% CI 1.3-8.3 months) but resulted in comparable OS (absolute difference −0.55 months; 95% CI −1.95 to 3.05 months). Safety profiles appeared to favor TARE. The authors concluded that TACE and TARE should be compared in larger prospective studies to better compare survival, progression, and safety data.

Nevena Damjanov, MD
Patients with hepatocellular carcinoma (HCC) may benefit from liver-directed therapies or palliative systemic therapies. This month we will review articles that address management of patients who were treated with different standard treatment options.

Komatsu and colleagues performed a case-matched analysis to evaluate the best first-line treatment for HCC in patients with macroscopic portal vein tumor thrombus (PVTT). Patients had advanced HCC and macroscopic PVTT that invaded an ipsilateral first-order portal branch, main trunk, or contralateral portal vein. The propensity score–matched groups underwent either hepatectomy (n = 36) or received sorafenib (n = 36). To be considered for resection, patients had to have Child-Pugh (CP) grade A or B liver function, an Eastern Cooperative Oncology Group Performance Status score of ≤ 1, life expectancy of > 3 months, and the macroscopic resection of the targeted tumor could be planned with an estimated remnant liver volume ≥ 35%.

Out of 36 patients who underwent surgery, 23 underwent reductive hepatectomy, and 13 underwent complete resection of tumor. Out of 36 patients who received sorafenib, 21 underwent subsequent treatments. The median overall survival (OS) of patients who underwent hepatectomy was 15.1 months, significantly longer than the 4.5 months for patients who were treated with sorafenib. The authors concluded that selected patients who underwent tumor resection lived longer than patients who received systemic therapy with sorafenib first, despite the presence of macroscopic PVTT.

Lenvatinib is an approved treatment for patients with HCC. Because most patients with HCC have underlying cirrhosis, monitoring the underlying liver function is an important facet of patient management. Huynh and colleagues reported on patients in the REFLECT trial whose liver function deteriorated from CP-A to CP-B while receiving systemic therapy. This post hoc analysis included patients whose liver function deteriorated to CP-B or remained CP-A within 8 weeks of randomization to lenvatinib (CP-B: n = 60; CP-A: n = 413) or sorafenib (CP-B: n = 47; CP-A: n = 427). Patients receiving lenvatinib who developed CP-B cirrhosis compared with patients who maintained CP-A cirrhosis had a median progression-free survival (PFS) of 3.7 months (95% CI 1.8-7.4) vs 6.5 months (95% CI 5.6-7.4) and OS of 6.8 months (95% CI 2.6-10.3) vs 13.3 months (95% CI 11.6-16.1). CP-B patients receiving sorafenib had a median PFS and OS of only 0.5 months (95% CI 0.1-3.6) and 4.5 months (95% CI 2.9-6.1), respectively. No new safety signals were reported in CP-B patients. The investigators concluded that deterioration of liver function to CP-B does not require the discontinuation of lenvatinib therapy.

Finally, Brown and colleagues performed a meta-analysis of studies that evaluated transarterial radioembolization (TARE) and transarterial chemoembolization (TACE) in patients with HCC by reviewing 17 studies involving 2465 patients that directly compared TACE and TARE. TARE significantly prolonged the mean time to progression (17.5 vs 9.8 months; 95% CI 1.3-8.3 months) but resulted in comparable OS (absolute difference −0.55 months; 95% CI −1.95 to 3.05 months). Safety profiles appeared to favor TARE. The authors concluded that TACE and TARE should be compared in larger prospective studies to better compare survival, progression, and safety data.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: HCC October 2022
Gate On Date
Thu, 01/20/2022 - 12:15
Un-Gate On Date
Thu, 01/20/2022 - 12:15
Use ProPublica
CFC Schedule Remove Status
Thu, 01/20/2022 - 12:15
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
332967.1
Activity ID
83146
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Exact Sciences Corporate [ 6025 ]

Commentary: New Treatments and Fertility Preservation in BC, October 2022

Article Type
Changed
Wed, 01/04/2023 - 17:23
Dr. Roesch scans the journals, so you don't have to!

Erin Roesch, MD
The combination of endocrine therapy plus cyclin-dependent kinase (CDK) 4/6 inhibitors has led to significant survival benefits in the first-line setting for hormone receptor–positive (HR+)/ human epidermal growth factor receptor 2–negative (HER2-) metastatic breast cancer. However, the development of endocrine resistance poses a future challenge because sequential single-agent chemotherapy has historically been pursued in later lines of treatment.


The phase 3 TROPiCS-02 trial compared the trophoblast cell-surface antigen 2 (Trop-2)–directed antibody-drug conjugate sacituzumab govitecan with physician's choice of chemotherapy. There were 543 patients with HR+/HER2- locally recurrent inoperable or metastatic breast cancer that was also endocrine resistant and had been treated with two to four prior lines of chemotherapy in the advanced setting (Rugo et al). Sacituzumab govitecan led to a 34% reduction in risk for progression or death vs physician's choice of chemotherapy (hazard ratio 0.66; P = .0003; median progression-free survival [PFS], 5.5 months vs 4.0 months, respectively). The PFS at 6 and 12 months was 46% vs 30% and 21% vs 7% for sacituzumab govitecan and physician's choice chemotherapy, respectively. Grade ≥ 3 neutropenia and diarrhea were more common with sacituzumab govitecan than with physician's choice of chemotherapy (51% vs 9%) and were managed with supportive care measures.

Sacituzumab govitecan has previously proven an active drug for metastatic triple-negative breast cancer, and the final results from the phase 3 ASCENT study1 confirmed a significant survival benefit with sacituzumab govitecan vs single-agent chemotherapy for patients with metastatic triple-negative breast cancer in the second-line or greater setting (median overall survival [OS] 12.1 vs 6.7 months; hazard ratio 0.48; P < .0001). Sacituzumab govitecan and other antibody-drug conjugates are emerging as active therapies for all subtypes of breast cancer, and more treatment options will inevitably yield future questions surrounding sequencing and resistance mechanisms.

The phase 3 NALA trial2 demonstrated superior outcomes with the combination of neratinib plus capecitabine vs lapatinib plus capecitabine among patients with previously treated HER2+ metastatic breast cancer (hazard ratio 0.76; 1-year PFS 29% vs 15%). Findings from a single-center retrospective study including 72 patients with HER2+ advanced breast cancer who received either neratinib plus capecitabine or neratinib alone support efficacy and tolerability in the real-world setting (Cunningham et al). Among all patients, the median PFS was 5.9 months and median OS was 15.0 months; for those with brain metastases (n = 38), median PFS and median OS were 5.7 and 12.5 months, respectively. The gastrointestinal toxicity of neratinib can affect its clinical use, and a total of 64% of patients in this study reported diarrhea (10% reported grade 3) despite using antidiarrheal prophylaxis.

The treatment algorithm for HER2+ metastatic breast cancer has been evolving at a rapid pace, specifically for second-line and beyond. Neratinib remains a relevant therapy choice for these patients. The central nervous system activity of neratinib and other tyrosine kinase inhibitors, such as tucatinib, often make these the preferred treatment options for patients with brain metastases and stimulate the idea of prevention of brain metastases at an earlier time point.

Young women with breast cancer encounter unique challenges related to the stage of life during which they are diagnosed. It is essential to consider the effect of cancer treatment on fertility, including direct effects of chemotherapy and the duration of endocrine therapy (5-10 years) that can delay attempts at conceiving. Potential concerns surrounding fertility preservation (FP) include the theoretical risk of increased estradiol levels and treatment delay to allow these procedures to occur; however, various studies have supported the safety of FP.3

A prospective cohort study including 1257 women of reproductive age who were diagnosed with breast cancer and underwent FP treatment demonstrated similar disease-specific mortality in women who underwent hormonal FP (adjusted hazard ratio [aHR] 0.59; 95% CI 0.32-1.09), those who underwent nonhormonal FP (aHR 0.51, 95% CI 0.20-1.29), and women who did not pursue FP (Marklund et al). Furthermore, among 723 women with detailed information on relapse there was no significant difference in rate of relapse or death among those who underwent hormonal FP (aHR 0.81; 95% CI 0.49-1.37) vs those who underwent nonhormonal FP (aHR 0.75; 95% CI 0.35-1.62).

The growing body of evidence in this field highlights the importance of oncofertility awareness for both patients and providers. Young women diagnosed with breast cancer should be offered referrals to fertility specialists when interested and educated on the safety of these approaches as it relates to breast cancer outcomes.

A multicenter retrospective study compared the efficacy and safety of controlled ovarian hyperstimulation with letrozole (LetCOH) or without letrozole (cCOH) among 97 young women (≤ 40 years) diagnosed with early-stage breast cancer (Goldrat et al). The LetCOH group had lower peak estradiol levels (343 pg/mL vs 1009 pg/mL; P < .001) and higher oocyte maturation rates compared with the cCOH group, but a similar number of mature oocytes collected (P = .281). Disease recurrence occurred more frequently in the LetCOH group than in the cCOH group (17% vs 7.2%), and five patients in total had a distant recurrence (four undergoing LetCOH vs one undergoing cCOH).

The LetCOH group did have larger tumors and a higher number of HER2+ cancers. These findings suggest that a COH protocol using letrozole can yield FP outcomes similar to those of the conventional protocol while minimizing exposure to high levels of estradiol. Extended follow-up and future prospective studies will be essential to gain survival data and further define the roles of various FP procedures.

Additional References

  1. Bardia A, Tolaney SM, Loirat D, et al. Sacituzumab govitecan (SG) versus treatment of physician's choice (TPC) in patients (pts) with previously treated, metastatic triple-negative breast cancer (mTNBC): final results from the phase 3 ASCENT study. J Clin Oncol. 2022;40(16 Suppl):107 Doi: 10.1200/JCO.2022.40.16_suppl.1071
  2. Saura C, Oliveira M, Feng YH, et al. Neratinib plus capecitabine versus lapatinib plus capecitabine in HER2-positive metastatic breast cancer previously treated with ≥ 2 HER2-directed regimens: phase III NALA trial. J Clin Oncol. 2020;38:3138-3149. Doi: 10.1200/JCO.20.00147
  3. Moravek MB, Confino R, Lawson AK, et al. Predictors and outcomes in breast cancer patients who did or did not pursue fertility preservation. Breast Cancer Res Treat. 2021;186:429-437. Doi: 10.1007/s10549-020-06031-4

 

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Publications
Topics
Sections
Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Author and Disclosure Information

Erin E. Roesch, MD, Associate Staff, Department of Medical Oncology, Cleveland Clinic, Cleveland, Ohio
Erin E. Roesch, MD, has disclosed the following relevant financial relationships:
Serve(d) as a speaker or a member of a speakers bureau for: Puma Biotechnology

Dr. Roesch scans the journals, so you don't have to!
Dr. Roesch scans the journals, so you don't have to!

Erin Roesch, MD
The combination of endocrine therapy plus cyclin-dependent kinase (CDK) 4/6 inhibitors has led to significant survival benefits in the first-line setting for hormone receptor–positive (HR+)/ human epidermal growth factor receptor 2–negative (HER2-) metastatic breast cancer. However, the development of endocrine resistance poses a future challenge because sequential single-agent chemotherapy has historically been pursued in later lines of treatment.


The phase 3 TROPiCS-02 trial compared the trophoblast cell-surface antigen 2 (Trop-2)–directed antibody-drug conjugate sacituzumab govitecan with physician's choice of chemotherapy. There were 543 patients with HR+/HER2- locally recurrent inoperable or metastatic breast cancer that was also endocrine resistant and had been treated with two to four prior lines of chemotherapy in the advanced setting (Rugo et al). Sacituzumab govitecan led to a 34% reduction in risk for progression or death vs physician's choice of chemotherapy (hazard ratio 0.66; P = .0003; median progression-free survival [PFS], 5.5 months vs 4.0 months, respectively). The PFS at 6 and 12 months was 46% vs 30% and 21% vs 7% for sacituzumab govitecan and physician's choice chemotherapy, respectively. Grade ≥ 3 neutropenia and diarrhea were more common with sacituzumab govitecan than with physician's choice of chemotherapy (51% vs 9%) and were managed with supportive care measures.

Sacituzumab govitecan has previously proven an active drug for metastatic triple-negative breast cancer, and the final results from the phase 3 ASCENT study1 confirmed a significant survival benefit with sacituzumab govitecan vs single-agent chemotherapy for patients with metastatic triple-negative breast cancer in the second-line or greater setting (median overall survival [OS] 12.1 vs 6.7 months; hazard ratio 0.48; P < .0001). Sacituzumab govitecan and other antibody-drug conjugates are emerging as active therapies for all subtypes of breast cancer, and more treatment options will inevitably yield future questions surrounding sequencing and resistance mechanisms.

The phase 3 NALA trial2 demonstrated superior outcomes with the combination of neratinib plus capecitabine vs lapatinib plus capecitabine among patients with previously treated HER2+ metastatic breast cancer (hazard ratio 0.76; 1-year PFS 29% vs 15%). Findings from a single-center retrospective study including 72 patients with HER2+ advanced breast cancer who received either neratinib plus capecitabine or neratinib alone support efficacy and tolerability in the real-world setting (Cunningham et al). Among all patients, the median PFS was 5.9 months and median OS was 15.0 months; for those with brain metastases (n = 38), median PFS and median OS were 5.7 and 12.5 months, respectively. The gastrointestinal toxicity of neratinib can affect its clinical use, and a total of 64% of patients in this study reported diarrhea (10% reported grade 3) despite using antidiarrheal prophylaxis.

The treatment algorithm for HER2+ metastatic breast cancer has been evolving at a rapid pace, specifically for second-line and beyond. Neratinib remains a relevant therapy choice for these patients. The central nervous system activity of neratinib and other tyrosine kinase inhibitors, such as tucatinib, often make these the preferred treatment options for patients with brain metastases and stimulate the idea of prevention of brain metastases at an earlier time point.

Young women with breast cancer encounter unique challenges related to the stage of life during which they are diagnosed. It is essential to consider the effect of cancer treatment on fertility, including direct effects of chemotherapy and the duration of endocrine therapy (5-10 years) that can delay attempts at conceiving. Potential concerns surrounding fertility preservation (FP) include the theoretical risk of increased estradiol levels and treatment delay to allow these procedures to occur; however, various studies have supported the safety of FP.3

A prospective cohort study including 1257 women of reproductive age who were diagnosed with breast cancer and underwent FP treatment demonstrated similar disease-specific mortality in women who underwent hormonal FP (adjusted hazard ratio [aHR] 0.59; 95% CI 0.32-1.09), those who underwent nonhormonal FP (aHR 0.51, 95% CI 0.20-1.29), and women who did not pursue FP (Marklund et al). Furthermore, among 723 women with detailed information on relapse there was no significant difference in rate of relapse or death among those who underwent hormonal FP (aHR 0.81; 95% CI 0.49-1.37) vs those who underwent nonhormonal FP (aHR 0.75; 95% CI 0.35-1.62).

The growing body of evidence in this field highlights the importance of oncofertility awareness for both patients and providers. Young women diagnosed with breast cancer should be offered referrals to fertility specialists when interested and educated on the safety of these approaches as it relates to breast cancer outcomes.

A multicenter retrospective study compared the efficacy and safety of controlled ovarian hyperstimulation with letrozole (LetCOH) or without letrozole (cCOH) among 97 young women (≤ 40 years) diagnosed with early-stage breast cancer (Goldrat et al). The LetCOH group had lower peak estradiol levels (343 pg/mL vs 1009 pg/mL; P < .001) and higher oocyte maturation rates compared with the cCOH group, but a similar number of mature oocytes collected (P = .281). Disease recurrence occurred more frequently in the LetCOH group than in the cCOH group (17% vs 7.2%), and five patients in total had a distant recurrence (four undergoing LetCOH vs one undergoing cCOH).

The LetCOH group did have larger tumors and a higher number of HER2+ cancers. These findings suggest that a COH protocol using letrozole can yield FP outcomes similar to those of the conventional protocol while minimizing exposure to high levels of estradiol. Extended follow-up and future prospective studies will be essential to gain survival data and further define the roles of various FP procedures.

Additional References

  1. Bardia A, Tolaney SM, Loirat D, et al. Sacituzumab govitecan (SG) versus treatment of physician's choice (TPC) in patients (pts) with previously treated, metastatic triple-negative breast cancer (mTNBC): final results from the phase 3 ASCENT study. J Clin Oncol. 2022;40(16 Suppl):107 Doi: 10.1200/JCO.2022.40.16_suppl.1071
  2. Saura C, Oliveira M, Feng YH, et al. Neratinib plus capecitabine versus lapatinib plus capecitabine in HER2-positive metastatic breast cancer previously treated with ≥ 2 HER2-directed regimens: phase III NALA trial. J Clin Oncol. 2020;38:3138-3149. Doi: 10.1200/JCO.20.00147
  3. Moravek MB, Confino R, Lawson AK, et al. Predictors and outcomes in breast cancer patients who did or did not pursue fertility preservation. Breast Cancer Res Treat. 2021;186:429-437. Doi: 10.1007/s10549-020-06031-4

 

Erin Roesch, MD
The combination of endocrine therapy plus cyclin-dependent kinase (CDK) 4/6 inhibitors has led to significant survival benefits in the first-line setting for hormone receptor–positive (HR+)/ human epidermal growth factor receptor 2–negative (HER2-) metastatic breast cancer. However, the development of endocrine resistance poses a future challenge because sequential single-agent chemotherapy has historically been pursued in later lines of treatment.


The phase 3 TROPiCS-02 trial compared the trophoblast cell-surface antigen 2 (Trop-2)–directed antibody-drug conjugate sacituzumab govitecan with physician's choice of chemotherapy. There were 543 patients with HR+/HER2- locally recurrent inoperable or metastatic breast cancer that was also endocrine resistant and had been treated with two to four prior lines of chemotherapy in the advanced setting (Rugo et al). Sacituzumab govitecan led to a 34% reduction in risk for progression or death vs physician's choice of chemotherapy (hazard ratio 0.66; P = .0003; median progression-free survival [PFS], 5.5 months vs 4.0 months, respectively). The PFS at 6 and 12 months was 46% vs 30% and 21% vs 7% for sacituzumab govitecan and physician's choice chemotherapy, respectively. Grade ≥ 3 neutropenia and diarrhea were more common with sacituzumab govitecan than with physician's choice of chemotherapy (51% vs 9%) and were managed with supportive care measures.

Sacituzumab govitecan has previously proven an active drug for metastatic triple-negative breast cancer, and the final results from the phase 3 ASCENT study1 confirmed a significant survival benefit with sacituzumab govitecan vs single-agent chemotherapy for patients with metastatic triple-negative breast cancer in the second-line or greater setting (median overall survival [OS] 12.1 vs 6.7 months; hazard ratio 0.48; P < .0001). Sacituzumab govitecan and other antibody-drug conjugates are emerging as active therapies for all subtypes of breast cancer, and more treatment options will inevitably yield future questions surrounding sequencing and resistance mechanisms.

The phase 3 NALA trial2 demonstrated superior outcomes with the combination of neratinib plus capecitabine vs lapatinib plus capecitabine among patients with previously treated HER2+ metastatic breast cancer (hazard ratio 0.76; 1-year PFS 29% vs 15%). Findings from a single-center retrospective study including 72 patients with HER2+ advanced breast cancer who received either neratinib plus capecitabine or neratinib alone support efficacy and tolerability in the real-world setting (Cunningham et al). Among all patients, the median PFS was 5.9 months and median OS was 15.0 months; for those with brain metastases (n = 38), median PFS and median OS were 5.7 and 12.5 months, respectively. The gastrointestinal toxicity of neratinib can affect its clinical use, and a total of 64% of patients in this study reported diarrhea (10% reported grade 3) despite using antidiarrheal prophylaxis.

The treatment algorithm for HER2+ metastatic breast cancer has been evolving at a rapid pace, specifically for second-line and beyond. Neratinib remains a relevant therapy choice for these patients. The central nervous system activity of neratinib and other tyrosine kinase inhibitors, such as tucatinib, often make these the preferred treatment options for patients with brain metastases and stimulate the idea of prevention of brain metastases at an earlier time point.

Young women with breast cancer encounter unique challenges related to the stage of life during which they are diagnosed. It is essential to consider the effect of cancer treatment on fertility, including direct effects of chemotherapy and the duration of endocrine therapy (5-10 years) that can delay attempts at conceiving. Potential concerns surrounding fertility preservation (FP) include the theoretical risk of increased estradiol levels and treatment delay to allow these procedures to occur; however, various studies have supported the safety of FP.3

A prospective cohort study including 1257 women of reproductive age who were diagnosed with breast cancer and underwent FP treatment demonstrated similar disease-specific mortality in women who underwent hormonal FP (adjusted hazard ratio [aHR] 0.59; 95% CI 0.32-1.09), those who underwent nonhormonal FP (aHR 0.51, 95% CI 0.20-1.29), and women who did not pursue FP (Marklund et al). Furthermore, among 723 women with detailed information on relapse there was no significant difference in rate of relapse or death among those who underwent hormonal FP (aHR 0.81; 95% CI 0.49-1.37) vs those who underwent nonhormonal FP (aHR 0.75; 95% CI 0.35-1.62).

The growing body of evidence in this field highlights the importance of oncofertility awareness for both patients and providers. Young women diagnosed with breast cancer should be offered referrals to fertility specialists when interested and educated on the safety of these approaches as it relates to breast cancer outcomes.

A multicenter retrospective study compared the efficacy and safety of controlled ovarian hyperstimulation with letrozole (LetCOH) or without letrozole (cCOH) among 97 young women (≤ 40 years) diagnosed with early-stage breast cancer (Goldrat et al). The LetCOH group had lower peak estradiol levels (343 pg/mL vs 1009 pg/mL; P < .001) and higher oocyte maturation rates compared with the cCOH group, but a similar number of mature oocytes collected (P = .281). Disease recurrence occurred more frequently in the LetCOH group than in the cCOH group (17% vs 7.2%), and five patients in total had a distant recurrence (four undergoing LetCOH vs one undergoing cCOH).

The LetCOH group did have larger tumors and a higher number of HER2+ cancers. These findings suggest that a COH protocol using letrozole can yield FP outcomes similar to those of the conventional protocol while minimizing exposure to high levels of estradiol. Extended follow-up and future prospective studies will be essential to gain survival data and further define the roles of various FP procedures.

Additional References

  1. Bardia A, Tolaney SM, Loirat D, et al. Sacituzumab govitecan (SG) versus treatment of physician's choice (TPC) in patients (pts) with previously treated, metastatic triple-negative breast cancer (mTNBC): final results from the phase 3 ASCENT study. J Clin Oncol. 2022;40(16 Suppl):107 Doi: 10.1200/JCO.2022.40.16_suppl.1071
  2. Saura C, Oliveira M, Feng YH, et al. Neratinib plus capecitabine versus lapatinib plus capecitabine in HER2-positive metastatic breast cancer previously treated with ≥ 2 HER2-directed regimens: phase III NALA trial. J Clin Oncol. 2020;38:3138-3149. Doi: 10.1200/JCO.20.00147
  3. Moravek MB, Confino R, Lawson AK, et al. Predictors and outcomes in breast cancer patients who did or did not pursue fertility preservation. Breast Cancer Res Treat. 2021;186:429-437. Doi: 10.1007/s10549-020-06031-4

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Breast Cancer October 2022
Gate On Date
Mon, 05/03/2021 - 14:45
Un-Gate On Date
Mon, 05/03/2021 - 14:45
Use ProPublica
CFC Schedule Remove Status
Mon, 05/03/2021 - 14:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
329444.1
Activity ID
77844
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
Kadcyla [ 3564 ]

Commentary: IBS and comorbid conditions, October 2022

Article Type
Changed
Mon, 10/17/2022 - 14:48
APN Kametas scans the journals, so you don’t have to!

Marita Kametas, MSN, FNP-BC, CMSRN, COCN
Three studies this month highlight the increased incidence of irritable bowel syndrome (IBS) in the setting of other chronic diseases, specifically diseases with an effect on the abdomen and pelvis:

  • Fairbrass et al: Natural history and impact of IBS-type symptoms in inflammatory bowel disease during 6 years of longitudinal follow-up
  • Nabi et al: Endometriosis and IBS — a systematic review and meta-analyses
  • Wang et al: Factors related to IBS and differences among subtypes — a cross-sectional study in the UK Biobank

Combined diseases can make assessment and treatment very difficult for clinicians. Having a thorough understanding of the pathophysiology and phenotype of each of these diseases is imperative to ensuring that they are managed to the standard of care. When these diseases are in remission and abdominal symptoms persist, it begs the question what is the cause? IBS should always be in the differential diagnosis. The dynamic relationship between the varying presentations of IBS and co-occurring conditions can greatly affect the patient's quality of life and experience with the healthcare system. Thus, it is vital to implement an interdisciplinary approach in order to comprehensively care for the patient and build a therapeutic relationship with the patient. Establishing short-term and long-term goals through shared decision-making will create a foundation of trust and allow for improved patient experience.

Author and Disclosure Information

Marita Kametas, MSN,  FNP-BC, CMSRN, COCN, Advanced Practice Nurse, Department of Digestive Diseases, The University of Chicago Medicine, Chicage, Illinois

APN Kametas is an active member of the Abbvie Speakers’ Bureau

Publications
Topics
Sections
Author and Disclosure Information

Marita Kametas, MSN,  FNP-BC, CMSRN, COCN, Advanced Practice Nurse, Department of Digestive Diseases, The University of Chicago Medicine, Chicage, Illinois

APN Kametas is an active member of the Abbvie Speakers’ Bureau

Author and Disclosure Information

Marita Kametas, MSN,  FNP-BC, CMSRN, COCN, Advanced Practice Nurse, Department of Digestive Diseases, The University of Chicago Medicine, Chicage, Illinois

APN Kametas is an active member of the Abbvie Speakers’ Bureau

APN Kametas scans the journals, so you don’t have to!
APN Kametas scans the journals, so you don’t have to!

Marita Kametas, MSN, FNP-BC, CMSRN, COCN
Three studies this month highlight the increased incidence of irritable bowel syndrome (IBS) in the setting of other chronic diseases, specifically diseases with an effect on the abdomen and pelvis:

  • Fairbrass et al: Natural history and impact of IBS-type symptoms in inflammatory bowel disease during 6 years of longitudinal follow-up
  • Nabi et al: Endometriosis and IBS — a systematic review and meta-analyses
  • Wang et al: Factors related to IBS and differences among subtypes — a cross-sectional study in the UK Biobank

Combined diseases can make assessment and treatment very difficult for clinicians. Having a thorough understanding of the pathophysiology and phenotype of each of these diseases is imperative to ensuring that they are managed to the standard of care. When these diseases are in remission and abdominal symptoms persist, it begs the question what is the cause? IBS should always be in the differential diagnosis. The dynamic relationship between the varying presentations of IBS and co-occurring conditions can greatly affect the patient's quality of life and experience with the healthcare system. Thus, it is vital to implement an interdisciplinary approach in order to comprehensively care for the patient and build a therapeutic relationship with the patient. Establishing short-term and long-term goals through shared decision-making will create a foundation of trust and allow for improved patient experience.

Marita Kametas, MSN, FNP-BC, CMSRN, COCN
Three studies this month highlight the increased incidence of irritable bowel syndrome (IBS) in the setting of other chronic diseases, specifically diseases with an effect on the abdomen and pelvis:

  • Fairbrass et al: Natural history and impact of IBS-type symptoms in inflammatory bowel disease during 6 years of longitudinal follow-up
  • Nabi et al: Endometriosis and IBS — a systematic review and meta-analyses
  • Wang et al: Factors related to IBS and differences among subtypes — a cross-sectional study in the UK Biobank

Combined diseases can make assessment and treatment very difficult for clinicians. Having a thorough understanding of the pathophysiology and phenotype of each of these diseases is imperative to ensuring that they are managed to the standard of care. When these diseases are in remission and abdominal symptoms persist, it begs the question what is the cause? IBS should always be in the differential diagnosis. The dynamic relationship between the varying presentations of IBS and co-occurring conditions can greatly affect the patient's quality of life and experience with the healthcare system. Thus, it is vital to implement an interdisciplinary approach in order to comprehensively care for the patient and build a therapeutic relationship with the patient. Establishing short-term and long-term goals through shared decision-making will create a foundation of trust and allow for improved patient experience.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: IBS October 2022
Gate On Date
Wed, 06/29/2022 - 18:45
Un-Gate On Date
Wed, 06/29/2022 - 18:45
Use ProPublica
CFC Schedule Remove Status
Wed, 06/29/2022 - 18:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
334012
Activity ID
87195
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
IBSRELA [6066]

Commentary: New Topical Approaches Hit the Spots (and the Itch) for AD, October 2022

Article Type
Changed
Tue, 09/27/2022 - 12:18
Dr. Silverberg scans the journals, so you don’t have to!

Jonathan Silverberg, MD, PhD, MPH
Over the past few months, I have commented on cutting-edge data supporting the use of multiple agents that were recently approved for the treatment of atopic dermatitis (AD), including injectable dupilumab and tralokinumab, oral abrocitinib, baricitinib and upadacitinib, and topical ruxolitinib. In this month's Clinical Edge Journal Scan commentary, I would like to address recent data for several novel topical treatments in AD.

The Janus kinase (JAK) family has become a popular target for novel drug development. There are four JAK subunits, JAK1, JAK2, JAK3, and TYK2, each playing different roles in vital immunologic, hematologic, and homeostatic functions. A wide array of topical JAK-inhibitors has been or are currently being investigated for the treatment of AD, each with different profiles of selectivity across the JAK family.

Ruxolitinib is a preferential JAK1 and JAK2 inhibitor. Oral ruxolitinib is currently approved for the treatment of myelofibrosis. Topical ruxolitinib cream is also approved in the United States for the treatment of mild-to-moderate AD and, more recently, vitiligo.

Blauvelt and colleagues published the findings from a post hoc analysis of data related to itch responses from two randomized, double-blind, vehicle-controlled, phase 3 studies of patients with mild-to-moderate AD. Topical ruxolitinib applied twice daily led to a significant increase in the proportion of patients achieving at least a 2-point or 4-point reduction in Peak Pruritus Numeric Rating Scale (PP-NRS) within 12 hours of initial application and continued improvements out to week 8. Patients treated with ruxolitinib cream were also significantly more likely to achieve an itch-free state and faster reductions in itch compared with vehicle. Rapid improvement of itch is an important feature for any topical therapy that may be used as needed for AD flares.

Brepocitinib is a preferential JAK1 and tyrosine kinase 2 inhibitor. Oral brepocitinib is currently under investigation for several immune-mediated disorders, including dermatomyositis and lupus. Topical brepocitinib cream was also studied in a randomized, double-blind, vehicle-controlled, phase 2 study of patients with mild-to-moderate AD. Brepocitinib cream applied daily or twice daily led to significant decreases in the Eczema Area and Severity Index (EASI), increases in the proportion of patients who achieved Investigator Global Assessment (IGA) scores of clear or almost clear, EASI-75 responses, and 2-point or 4-point reductions in PP-NRS. Overall, topical brepocitinib was well tolerated. Though more studies are needed, topical brepocitinib may become an important addition to our toolbox for managing AD and perhaps other chronic inflammatory skin diseases.

Let's move along to yet another novel mechanism studied in AD: ectoine. Ectoine is a naturally derived chemical from some bacteria that can act as an osmolyte and protect organisms from osmotic stress. It is used in some skin care products because it can protect against xerosis and ultraviolet exposure. Alexopoulos and colleagues published the results of a randomized, single-blind, vehicle-controlled study of a novel cream containing 1% ectoine and 0.1% hyaluronic acid (EHA) in children with mild-to-moderate AD. At week 4, application of EHA cream led to significant decreases in SCORing AD (SCORAD) and IGA scores compared with vehicle cream as well all secondary outcome measures. EHA cream was well-tolerated overall, with most adverse events being cutaneous and mild.

All of these topical agents were studied as "reactive" therapies, ie, to be applied to active AD lesions. It would be nice to have approaches that can also prevent AD. Ní Chaoimh and colleagues published findings from the STOP AD trial that examined whether routine application of emollients in the first 8 weeks of life can prevent AD at age 12 months. They found that early emollient use resulted in significantly lower incidence of AD and similar rates of skin infections at age 12 months. Previous studies found mixed results regarding the efficacy of routine application of emollients in newborns. Though more studies are needed, these results are promising and suggest that early life use of emollients could be a cheap, feasible, and effective option to prevent AD.

Author and Disclosure Information

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC

Publications
Topics
Sections
Author and Disclosure Information

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC

Author and Disclosure Information

Jonathan Silverberg, MD, PHD, MPH
George Washington University School of Medicine and Health Sciences
Washington, DC

Dr. Silverberg scans the journals, so you don’t have to!
Dr. Silverberg scans the journals, so you don’t have to!

Jonathan Silverberg, MD, PhD, MPH
Over the past few months, I have commented on cutting-edge data supporting the use of multiple agents that were recently approved for the treatment of atopic dermatitis (AD), including injectable dupilumab and tralokinumab, oral abrocitinib, baricitinib and upadacitinib, and topical ruxolitinib. In this month's Clinical Edge Journal Scan commentary, I would like to address recent data for several novel topical treatments in AD.

The Janus kinase (JAK) family has become a popular target for novel drug development. There are four JAK subunits, JAK1, JAK2, JAK3, and TYK2, each playing different roles in vital immunologic, hematologic, and homeostatic functions. A wide array of topical JAK-inhibitors has been or are currently being investigated for the treatment of AD, each with different profiles of selectivity across the JAK family.

Ruxolitinib is a preferential JAK1 and JAK2 inhibitor. Oral ruxolitinib is currently approved for the treatment of myelofibrosis. Topical ruxolitinib cream is also approved in the United States for the treatment of mild-to-moderate AD and, more recently, vitiligo.

Blauvelt and colleagues published the findings from a post hoc analysis of data related to itch responses from two randomized, double-blind, vehicle-controlled, phase 3 studies of patients with mild-to-moderate AD. Topical ruxolitinib applied twice daily led to a significant increase in the proportion of patients achieving at least a 2-point or 4-point reduction in Peak Pruritus Numeric Rating Scale (PP-NRS) within 12 hours of initial application and continued improvements out to week 8. Patients treated with ruxolitinib cream were also significantly more likely to achieve an itch-free state and faster reductions in itch compared with vehicle. Rapid improvement of itch is an important feature for any topical therapy that may be used as needed for AD flares.

Brepocitinib is a preferential JAK1 and tyrosine kinase 2 inhibitor. Oral brepocitinib is currently under investigation for several immune-mediated disorders, including dermatomyositis and lupus. Topical brepocitinib cream was also studied in a randomized, double-blind, vehicle-controlled, phase 2 study of patients with mild-to-moderate AD. Brepocitinib cream applied daily or twice daily led to significant decreases in the Eczema Area and Severity Index (EASI), increases in the proportion of patients who achieved Investigator Global Assessment (IGA) scores of clear or almost clear, EASI-75 responses, and 2-point or 4-point reductions in PP-NRS. Overall, topical brepocitinib was well tolerated. Though more studies are needed, topical brepocitinib may become an important addition to our toolbox for managing AD and perhaps other chronic inflammatory skin diseases.

Let's move along to yet another novel mechanism studied in AD: ectoine. Ectoine is a naturally derived chemical from some bacteria that can act as an osmolyte and protect organisms from osmotic stress. It is used in some skin care products because it can protect against xerosis and ultraviolet exposure. Alexopoulos and colleagues published the results of a randomized, single-blind, vehicle-controlled study of a novel cream containing 1% ectoine and 0.1% hyaluronic acid (EHA) in children with mild-to-moderate AD. At week 4, application of EHA cream led to significant decreases in SCORing AD (SCORAD) and IGA scores compared with vehicle cream as well all secondary outcome measures. EHA cream was well-tolerated overall, with most adverse events being cutaneous and mild.

All of these topical agents were studied as "reactive" therapies, ie, to be applied to active AD lesions. It would be nice to have approaches that can also prevent AD. Ní Chaoimh and colleagues published findings from the STOP AD trial that examined whether routine application of emollients in the first 8 weeks of life can prevent AD at age 12 months. They found that early emollient use resulted in significantly lower incidence of AD and similar rates of skin infections at age 12 months. Previous studies found mixed results regarding the efficacy of routine application of emollients in newborns. Though more studies are needed, these results are promising and suggest that early life use of emollients could be a cheap, feasible, and effective option to prevent AD.

Jonathan Silverberg, MD, PhD, MPH
Over the past few months, I have commented on cutting-edge data supporting the use of multiple agents that were recently approved for the treatment of atopic dermatitis (AD), including injectable dupilumab and tralokinumab, oral abrocitinib, baricitinib and upadacitinib, and topical ruxolitinib. In this month's Clinical Edge Journal Scan commentary, I would like to address recent data for several novel topical treatments in AD.

The Janus kinase (JAK) family has become a popular target for novel drug development. There are four JAK subunits, JAK1, JAK2, JAK3, and TYK2, each playing different roles in vital immunologic, hematologic, and homeostatic functions. A wide array of topical JAK-inhibitors has been or are currently being investigated for the treatment of AD, each with different profiles of selectivity across the JAK family.

Ruxolitinib is a preferential JAK1 and JAK2 inhibitor. Oral ruxolitinib is currently approved for the treatment of myelofibrosis. Topical ruxolitinib cream is also approved in the United States for the treatment of mild-to-moderate AD and, more recently, vitiligo.

Blauvelt and colleagues published the findings from a post hoc analysis of data related to itch responses from two randomized, double-blind, vehicle-controlled, phase 3 studies of patients with mild-to-moderate AD. Topical ruxolitinib applied twice daily led to a significant increase in the proportion of patients achieving at least a 2-point or 4-point reduction in Peak Pruritus Numeric Rating Scale (PP-NRS) within 12 hours of initial application and continued improvements out to week 8. Patients treated with ruxolitinib cream were also significantly more likely to achieve an itch-free state and faster reductions in itch compared with vehicle. Rapid improvement of itch is an important feature for any topical therapy that may be used as needed for AD flares.

Brepocitinib is a preferential JAK1 and tyrosine kinase 2 inhibitor. Oral brepocitinib is currently under investigation for several immune-mediated disorders, including dermatomyositis and lupus. Topical brepocitinib cream was also studied in a randomized, double-blind, vehicle-controlled, phase 2 study of patients with mild-to-moderate AD. Brepocitinib cream applied daily or twice daily led to significant decreases in the Eczema Area and Severity Index (EASI), increases in the proportion of patients who achieved Investigator Global Assessment (IGA) scores of clear or almost clear, EASI-75 responses, and 2-point or 4-point reductions in PP-NRS. Overall, topical brepocitinib was well tolerated. Though more studies are needed, topical brepocitinib may become an important addition to our toolbox for managing AD and perhaps other chronic inflammatory skin diseases.

Let's move along to yet another novel mechanism studied in AD: ectoine. Ectoine is a naturally derived chemical from some bacteria that can act as an osmolyte and protect organisms from osmotic stress. It is used in some skin care products because it can protect against xerosis and ultraviolet exposure. Alexopoulos and colleagues published the results of a randomized, single-blind, vehicle-controlled study of a novel cream containing 1% ectoine and 0.1% hyaluronic acid (EHA) in children with mild-to-moderate AD. At week 4, application of EHA cream led to significant decreases in SCORing AD (SCORAD) and IGA scores compared with vehicle cream as well all secondary outcome measures. EHA cream was well-tolerated overall, with most adverse events being cutaneous and mild.

All of these topical agents were studied as "reactive" therapies, ie, to be applied to active AD lesions. It would be nice to have approaches that can also prevent AD. Ní Chaoimh and colleagues published findings from the STOP AD trial that examined whether routine application of emollients in the first 8 weeks of life can prevent AD at age 12 months. They found that early emollient use resulted in significantly lower incidence of AD and similar rates of skin infections at age 12 months. Previous studies found mixed results regarding the efficacy of routine application of emollients in newborns. Though more studies are needed, these results are promising and suggest that early life use of emollients could be a cheap, feasible, and effective option to prevent AD.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Atopic Dermatitis October 2022
Gate On Date
Thu, 07/29/2021 - 18:45
Un-Gate On Date
Thu, 07/29/2021 - 18:45
Use ProPublica
CFC Schedule Remove Status
Thu, 07/29/2021 - 18:45
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article
Activity Salesforce Deliverable ID
325140.4
Activity ID
77941
Product Name
Clinical Edge Journal Scan
Product ID
124
Supporter Name /ID
RINVOQ [ 5260 ]

Hypertensive disorder during pregnancy increases risk for elevated blood pressure in offspring

Article Type
Changed
Tue, 10/18/2022 - 14:57

Key clinical point: Offspring who were exposed in utero to any subtype of hypertensive disorders during pregnancy (HDP) were at an increased risk for higher blood pressure (BP) than those with no exposure.

 

Major finding: In utero exposure vs no exposure to HDP was associated with higher systolic BP (mean difference 2.46 mm Hg; 95% CI 1.88-3.03 mm Hg) in offspring. Higher systolic BP was also observed in offspring exposed vs not exposed in utero to HDP subtypes, including pregnancy-associated hypertension, preeclampsia, gestational hypertension, and chronic hypertension.

 

Study details: Findings are from a systematic review and meta-analysis of 24 cohort studies including 3839 offspring who were exposed to HDP in utero and 57,977 offspring from normotensive mothers.

 

Disclosures: This study was partly supported by Sichuan Science and Technology Program, China. The authors declared no conflicts of interest.

 

Source: Yu H et al. Association between hypertensive disorders during pregnancy and elevated blood pressure in offspring: A systematic review and meta-analysis. J Clin Hypertens (Greenwich). 2022 (Sep 12). Doi: 10.1111/jch.14577

 

Publications
Topics
Sections

Key clinical point: Offspring who were exposed in utero to any subtype of hypertensive disorders during pregnancy (HDP) were at an increased risk for higher blood pressure (BP) than those with no exposure.

 

Major finding: In utero exposure vs no exposure to HDP was associated with higher systolic BP (mean difference 2.46 mm Hg; 95% CI 1.88-3.03 mm Hg) in offspring. Higher systolic BP was also observed in offspring exposed vs not exposed in utero to HDP subtypes, including pregnancy-associated hypertension, preeclampsia, gestational hypertension, and chronic hypertension.

 

Study details: Findings are from a systematic review and meta-analysis of 24 cohort studies including 3839 offspring who were exposed to HDP in utero and 57,977 offspring from normotensive mothers.

 

Disclosures: This study was partly supported by Sichuan Science and Technology Program, China. The authors declared no conflicts of interest.

 

Source: Yu H et al. Association between hypertensive disorders during pregnancy and elevated blood pressure in offspring: A systematic review and meta-analysis. J Clin Hypertens (Greenwich). 2022 (Sep 12). Doi: 10.1111/jch.14577

 

Key clinical point: Offspring who were exposed in utero to any subtype of hypertensive disorders during pregnancy (HDP) were at an increased risk for higher blood pressure (BP) than those with no exposure.

 

Major finding: In utero exposure vs no exposure to HDP was associated with higher systolic BP (mean difference 2.46 mm Hg; 95% CI 1.88-3.03 mm Hg) in offspring. Higher systolic BP was also observed in offspring exposed vs not exposed in utero to HDP subtypes, including pregnancy-associated hypertension, preeclampsia, gestational hypertension, and chronic hypertension.

 

Study details: Findings are from a systematic review and meta-analysis of 24 cohort studies including 3839 offspring who were exposed to HDP in utero and 57,977 offspring from normotensive mothers.

 

Disclosures: This study was partly supported by Sichuan Science and Technology Program, China. The authors declared no conflicts of interest.

 

Source: Yu H et al. Association between hypertensive disorders during pregnancy and elevated blood pressure in offspring: A systematic review and meta-analysis. J Clin Hypertens (Greenwich). 2022 (Sep 12). Doi: 10.1111/jch.14577

 

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Obstetric Emergencies October 2022
Gate On Date
Tue, 09/27/2022 - 10:00
Un-Gate On Date
Tue, 09/27/2022 - 10:00
Use ProPublica
CFC Schedule Remove Status
Tue, 09/27/2022 - 10:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article

Obstetrics injuries and management during shoulder dystocia

Article Type
Changed
Tue, 10/18/2022 - 14:57

Key clinical point: The risk for brachial plexus strain, injury, or tear can be minimized with prompt identification of shoulder dystocia (SD) accompanied by cessation of axial fetal head traction, while accurate obstetrical maneuvers can avoid permanent obstetric brachial palsy (OBP) or cerebral morbidity.

 

Major finding: SD was mostly unilateral anterior, with only 0.9% of cases diagnosed as the more difficult bilateral SD and 2% as recurrent SD. The majority (87.4%) of SD cases were managed by McRobert’s maneuver; the other management procedures included Barnum’s procedure (7.9%), Wood’s maneuver (3.9%), and Menticoglou procedure (0.4%). Only 7.5% of newborns were diagnosed with transient form of Duchenne Erb obstetrics brachioparesis (OBP), none with permanent OBP, and only 1 with cerebral morbidity.

 

Study details: This retrospective study analyzed the data of 45,687 singleton deliveries (vaginal deliveries, 78.9%; cesarean sections, 21.1%). Overall, 0.7% of vaginally delivered neonates had fetal SD.

Disclosures: No source of funding was reported. The authors declared no conflicts of interest.

 

Source: Habek D et al. Obstetrics injuries during shoulder dystocia in a tertiary perinatal center. Eur J Obstet Gynecol Reprod Biol. 2022;278:33-37 (Sep 10). Doi: 10.1016/j.ejogrb.2022.09.009

Publications
Topics
Sections

Key clinical point: The risk for brachial plexus strain, injury, or tear can be minimized with prompt identification of shoulder dystocia (SD) accompanied by cessation of axial fetal head traction, while accurate obstetrical maneuvers can avoid permanent obstetric brachial palsy (OBP) or cerebral morbidity.

 

Major finding: SD was mostly unilateral anterior, with only 0.9% of cases diagnosed as the more difficult bilateral SD and 2% as recurrent SD. The majority (87.4%) of SD cases were managed by McRobert’s maneuver; the other management procedures included Barnum’s procedure (7.9%), Wood’s maneuver (3.9%), and Menticoglou procedure (0.4%). Only 7.5% of newborns were diagnosed with transient form of Duchenne Erb obstetrics brachioparesis (OBP), none with permanent OBP, and only 1 with cerebral morbidity.

 

Study details: This retrospective study analyzed the data of 45,687 singleton deliveries (vaginal deliveries, 78.9%; cesarean sections, 21.1%). Overall, 0.7% of vaginally delivered neonates had fetal SD.

Disclosures: No source of funding was reported. The authors declared no conflicts of interest.

 

Source: Habek D et al. Obstetrics injuries during shoulder dystocia in a tertiary perinatal center. Eur J Obstet Gynecol Reprod Biol. 2022;278:33-37 (Sep 10). Doi: 10.1016/j.ejogrb.2022.09.009

Key clinical point: The risk for brachial plexus strain, injury, or tear can be minimized with prompt identification of shoulder dystocia (SD) accompanied by cessation of axial fetal head traction, while accurate obstetrical maneuvers can avoid permanent obstetric brachial palsy (OBP) or cerebral morbidity.

 

Major finding: SD was mostly unilateral anterior, with only 0.9% of cases diagnosed as the more difficult bilateral SD and 2% as recurrent SD. The majority (87.4%) of SD cases were managed by McRobert’s maneuver; the other management procedures included Barnum’s procedure (7.9%), Wood’s maneuver (3.9%), and Menticoglou procedure (0.4%). Only 7.5% of newborns were diagnosed with transient form of Duchenne Erb obstetrics brachioparesis (OBP), none with permanent OBP, and only 1 with cerebral morbidity.

 

Study details: This retrospective study analyzed the data of 45,687 singleton deliveries (vaginal deliveries, 78.9%; cesarean sections, 21.1%). Overall, 0.7% of vaginally delivered neonates had fetal SD.

Disclosures: No source of funding was reported. The authors declared no conflicts of interest.

 

Source: Habek D et al. Obstetrics injuries during shoulder dystocia in a tertiary perinatal center. Eur J Obstet Gynecol Reprod Biol. 2022;278:33-37 (Sep 10). Doi: 10.1016/j.ejogrb.2022.09.009

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Article Series
Clinical Edge Journal Scan: Obstetric Emergencies October 2022
Gate On Date
Tue, 09/27/2022 - 10:00
Un-Gate On Date
Tue, 09/27/2022 - 10:00
Use ProPublica
CFC Schedule Remove Status
Tue, 09/27/2022 - 10:00
Hide sidebar & use full width
render the right sidebar.
Conference Recap Checkbox
Not Conference Recap
Clinical Edge
Display the Slideshow in this Article
Medscape Article
Display survey writer
Reuters content
Disable Inline Native ads
WebMD Article