User login
Vonoprazan-based therapy for resistant H. pylori superior to standard care
A look at the data behind the FDA approval
Vonoprazan, a potassium-competitive acid blocker, appears to be superior to standard proton pump inhibitor–based therapy in clarithromycin-resistant Helicobacter pylori strains, as well as noninferior to standard care in nonresistant infections, according to a recent study that supported a Food and Drug Administration approval of vonoprazan dual and triple therapies in May 2022.
For decades, H. pylori has been mostly treated by proton pump inhibitor–based triple therapy, which includes a proton pump inhibitor, clarithromycin, and amoxicillin or metronidazole. However, eradication rates have dropped below 80% in the United States and Europe, according to the authors, mainly because of rising rates of clarithromycin resistance.
Since H. pylori is a leading cause of peptic ulcer, gastric adenocarcinoma, and gastric mucosa–associated lymphoid tissue lymphoma, better eradication methods should be highlighted, researchers led by William Chey, MD, professor of medicine and director of the GI Physiology Laboratory at Michigan Medicine in Ann Arbor, wrote in Gastroenterology.
In a multicenter, randomized, controlled, phase 3 trial, the research team studied 1,046 treatment-naive adults with H. pylori infection at 103 sites in the U.S., the U.K., Bulgaria, the Czech Republic, Hungary, and Poland between December 2019 and January 2021.
The patients were randomized to receive open-label vonoprazan dual therapy or a double-blind triple therapy twice a day for 14 days. The vonoprazan dual therapy consisted of 20 mg of vonoprazan twice daily and 1 gram of amoxicillin three times per day. The triple therapy consisted of 20 mg of vonoprazan or 30 mg of lansoprazole (standard care), each given with 1 gram of amoxicillin and 500 mg of clarithromycin.
The primary outcome assessed noninferiority in eradication rates in patients without clarithromycin- and amoxicillin-resistant strains, with a noninferiority margin of 10%. Secondary outcomes assessed the superiority in eradication rates in clarithromycin-resistant infections, as well as in all patients.
Eradication rates for nonresistant strains were 84.7% for vonoprazan triple therapy and 78.5% for vonoprazan dual therapy, compared with 78.8% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered noninferior to standard therapy.
The eradication rates in clarithromycin-resistant infections were 65.8% for vonoprazan triple therapy and 69.6% in vonoprazan dual therapy, compared with 31.9% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered superior to standard therapy, with a difference of 33.9 percentage points for triple therapy and 37.7 percentage points for dual therapy.
In all patients, the eradication rates were 80.8% for vonoprazan triple therapy and 77.2% for vonoprazan dual therapy, compared with 68.5% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered superior, with a difference of 12.3 percentage points for triple therapy and 8.7 percentage points for dual therapy.
Treatment-emergent adverse events were reported in 34.1% of patients in the vonoprazan triple therapy group and 29.9% of patients in the vonoprazan dual therapy group, compared with 34.5% in the lansoprazole triple-therapy group. Most adverse events were mild to moderate.
Serious adverse events occurred in 1.3% of the overall study population, including 1.7% of the vonoprazan triple therapy group, 1.4% of the vonoprazan dual therapy group, and 0.9% of the lansoprazole triple therapy group. None were considered related to the study drugs.
Vonoprazan was approved for the treatment of H. pylori infections by the FDA in May 2022, and had already been approved for treatment of H. pylori infections and other acid-related diseases in several other countries. It decreases intragastric pH and maintains it to a greater degree than that of proton pump inhibitors, which has been associated with higher eradication rates, the authors wrote.
“Optimizing current regimens offers the potential to increase eradication rates and reduce additional antibiotic usage, thereby promoting and improving antimicrobial stewardship,” the study authors wrote.
The study was funded by Phathom Pharmaceuticals, which contributed to the design and conduct of the trial, collection and interpretation of the data, preparation and review of the manuscript, and the decision to submit the manuscript for publication. The study authors declared various conflicts of interest, including some who have received compensation as a consultant, advisory committee member, or employee for Phathom Pharmaceuticals.
Gastric acid inhibition plays a fundamental role for H. pylori eradication. Proton pump inhibitors (PPIs) are generally used, combined with antibiotics, in this scenario. More recently, vonoprazan, a potassium-competitive acid blocker, has been suggested to enhance H. pylori therapy by optimizing gastric acid suppression. However, clinical experience with vonoprazan has been limited to East Asian countries. The study by Chey et al. reports data from the first clinical trial from the United States and Europe, concluding that vonoprazan triple (together with amoxicillin and clarithromycin) and dual (together with amoxicillin) therapies were superior to PPI-based triple therapy, especially in clarithromycin-resistant strains.
However, some aspects deserve to be taken into consideration. The first one is that the cure rate with the standard triple therapy (with lansoprazole) was as low as 68%, underlining what has been known for a long time: This regimen should no longer be considered standard treatment in Europe or the United States and that it should not be recommended in areas with high (>15%) clarithromycin resistance, such as the United States and most European countries.
Secondly, the overall efficacy considering all patients (both with clarithromycin-susceptible and -resistant strains) with vonoprazan dual and triple regimens were of only 77% and 81%, not reaching the recommended target (≥ 90%) for first-line treatment. Therefore, the fair conclusion of the present article should have been not only that vonoprazan regimens are more effective than PPI ones, but also that all of them are insufficiently effective.
Finally, eradication rates in clarithromycin-resistant infections with the vonoprazan regimens (≤ 70%), although superior to those with lansoprazole (32%), were still clearly suboptimal, emphasizing that both PPI and vonoprazan based treatments would be inadequate if used in high-clarithromycin resistance regions.
Javier P. Gisbert, MD, PhD, is with the Hospital Universitario de La Princesa and the Universidad Autónoma de Madrid, both in Madrid. Dr. Gisbert has served as speaker, consultant, and advisory member for or has received research funding from Mayoly, Allergan, Diasorin, Gebro Pharma, and Richen.
A look at the data behind the FDA approval
A look at the data behind the FDA approval
Gastric acid inhibition plays a fundamental role for H. pylori eradication. Proton pump inhibitors (PPIs) are generally used, combined with antibiotics, in this scenario. More recently, vonoprazan, a potassium-competitive acid blocker, has been suggested to enhance H. pylori therapy by optimizing gastric acid suppression. However, clinical experience with vonoprazan has been limited to East Asian countries. The study by Chey et al. reports data from the first clinical trial from the United States and Europe, concluding that vonoprazan triple (together with amoxicillin and clarithromycin) and dual (together with amoxicillin) therapies were superior to PPI-based triple therapy, especially in clarithromycin-resistant strains.
However, some aspects deserve to be taken into consideration. The first one is that the cure rate with the standard triple therapy (with lansoprazole) was as low as 68%, underlining what has been known for a long time: This regimen should no longer be considered standard treatment in Europe or the United States and that it should not be recommended in areas with high (>15%) clarithromycin resistance, such as the United States and most European countries.
Secondly, the overall efficacy considering all patients (both with clarithromycin-susceptible and -resistant strains) with vonoprazan dual and triple regimens were of only 77% and 81%, not reaching the recommended target (≥ 90%) for first-line treatment. Therefore, the fair conclusion of the present article should have been not only that vonoprazan regimens are more effective than PPI ones, but also that all of them are insufficiently effective.
Finally, eradication rates in clarithromycin-resistant infections with the vonoprazan regimens (≤ 70%), although superior to those with lansoprazole (32%), were still clearly suboptimal, emphasizing that both PPI and vonoprazan based treatments would be inadequate if used in high-clarithromycin resistance regions.
Javier P. Gisbert, MD, PhD, is with the Hospital Universitario de La Princesa and the Universidad Autónoma de Madrid, both in Madrid. Dr. Gisbert has served as speaker, consultant, and advisory member for or has received research funding from Mayoly, Allergan, Diasorin, Gebro Pharma, and Richen.
Gastric acid inhibition plays a fundamental role for H. pylori eradication. Proton pump inhibitors (PPIs) are generally used, combined with antibiotics, in this scenario. More recently, vonoprazan, a potassium-competitive acid blocker, has been suggested to enhance H. pylori therapy by optimizing gastric acid suppression. However, clinical experience with vonoprazan has been limited to East Asian countries. The study by Chey et al. reports data from the first clinical trial from the United States and Europe, concluding that vonoprazan triple (together with amoxicillin and clarithromycin) and dual (together with amoxicillin) therapies were superior to PPI-based triple therapy, especially in clarithromycin-resistant strains.
However, some aspects deserve to be taken into consideration. The first one is that the cure rate with the standard triple therapy (with lansoprazole) was as low as 68%, underlining what has been known for a long time: This regimen should no longer be considered standard treatment in Europe or the United States and that it should not be recommended in areas with high (>15%) clarithromycin resistance, such as the United States and most European countries.
Secondly, the overall efficacy considering all patients (both with clarithromycin-susceptible and -resistant strains) with vonoprazan dual and triple regimens were of only 77% and 81%, not reaching the recommended target (≥ 90%) for first-line treatment. Therefore, the fair conclusion of the present article should have been not only that vonoprazan regimens are more effective than PPI ones, but also that all of them are insufficiently effective.
Finally, eradication rates in clarithromycin-resistant infections with the vonoprazan regimens (≤ 70%), although superior to those with lansoprazole (32%), were still clearly suboptimal, emphasizing that both PPI and vonoprazan based treatments would be inadequate if used in high-clarithromycin resistance regions.
Javier P. Gisbert, MD, PhD, is with the Hospital Universitario de La Princesa and the Universidad Autónoma de Madrid, both in Madrid. Dr. Gisbert has served as speaker, consultant, and advisory member for or has received research funding from Mayoly, Allergan, Diasorin, Gebro Pharma, and Richen.
Vonoprazan, a potassium-competitive acid blocker, appears to be superior to standard proton pump inhibitor–based therapy in clarithromycin-resistant Helicobacter pylori strains, as well as noninferior to standard care in nonresistant infections, according to a recent study that supported a Food and Drug Administration approval of vonoprazan dual and triple therapies in May 2022.
For decades, H. pylori has been mostly treated by proton pump inhibitor–based triple therapy, which includes a proton pump inhibitor, clarithromycin, and amoxicillin or metronidazole. However, eradication rates have dropped below 80% in the United States and Europe, according to the authors, mainly because of rising rates of clarithromycin resistance.
Since H. pylori is a leading cause of peptic ulcer, gastric adenocarcinoma, and gastric mucosa–associated lymphoid tissue lymphoma, better eradication methods should be highlighted, researchers led by William Chey, MD, professor of medicine and director of the GI Physiology Laboratory at Michigan Medicine in Ann Arbor, wrote in Gastroenterology.
In a multicenter, randomized, controlled, phase 3 trial, the research team studied 1,046 treatment-naive adults with H. pylori infection at 103 sites in the U.S., the U.K., Bulgaria, the Czech Republic, Hungary, and Poland between December 2019 and January 2021.
The patients were randomized to receive open-label vonoprazan dual therapy or a double-blind triple therapy twice a day for 14 days. The vonoprazan dual therapy consisted of 20 mg of vonoprazan twice daily and 1 gram of amoxicillin three times per day. The triple therapy consisted of 20 mg of vonoprazan or 30 mg of lansoprazole (standard care), each given with 1 gram of amoxicillin and 500 mg of clarithromycin.
The primary outcome assessed noninferiority in eradication rates in patients without clarithromycin- and amoxicillin-resistant strains, with a noninferiority margin of 10%. Secondary outcomes assessed the superiority in eradication rates in clarithromycin-resistant infections, as well as in all patients.
Eradication rates for nonresistant strains were 84.7% for vonoprazan triple therapy and 78.5% for vonoprazan dual therapy, compared with 78.8% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered noninferior to standard therapy.
The eradication rates in clarithromycin-resistant infections were 65.8% for vonoprazan triple therapy and 69.6% in vonoprazan dual therapy, compared with 31.9% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered superior to standard therapy, with a difference of 33.9 percentage points for triple therapy and 37.7 percentage points for dual therapy.
In all patients, the eradication rates were 80.8% for vonoprazan triple therapy and 77.2% for vonoprazan dual therapy, compared with 68.5% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered superior, with a difference of 12.3 percentage points for triple therapy and 8.7 percentage points for dual therapy.
Treatment-emergent adverse events were reported in 34.1% of patients in the vonoprazan triple therapy group and 29.9% of patients in the vonoprazan dual therapy group, compared with 34.5% in the lansoprazole triple-therapy group. Most adverse events were mild to moderate.
Serious adverse events occurred in 1.3% of the overall study population, including 1.7% of the vonoprazan triple therapy group, 1.4% of the vonoprazan dual therapy group, and 0.9% of the lansoprazole triple therapy group. None were considered related to the study drugs.
Vonoprazan was approved for the treatment of H. pylori infections by the FDA in May 2022, and had already been approved for treatment of H. pylori infections and other acid-related diseases in several other countries. It decreases intragastric pH and maintains it to a greater degree than that of proton pump inhibitors, which has been associated with higher eradication rates, the authors wrote.
“Optimizing current regimens offers the potential to increase eradication rates and reduce additional antibiotic usage, thereby promoting and improving antimicrobial stewardship,” the study authors wrote.
The study was funded by Phathom Pharmaceuticals, which contributed to the design and conduct of the trial, collection and interpretation of the data, preparation and review of the manuscript, and the decision to submit the manuscript for publication. The study authors declared various conflicts of interest, including some who have received compensation as a consultant, advisory committee member, or employee for Phathom Pharmaceuticals.
Vonoprazan, a potassium-competitive acid blocker, appears to be superior to standard proton pump inhibitor–based therapy in clarithromycin-resistant Helicobacter pylori strains, as well as noninferior to standard care in nonresistant infections, according to a recent study that supported a Food and Drug Administration approval of vonoprazan dual and triple therapies in May 2022.
For decades, H. pylori has been mostly treated by proton pump inhibitor–based triple therapy, which includes a proton pump inhibitor, clarithromycin, and amoxicillin or metronidazole. However, eradication rates have dropped below 80% in the United States and Europe, according to the authors, mainly because of rising rates of clarithromycin resistance.
Since H. pylori is a leading cause of peptic ulcer, gastric adenocarcinoma, and gastric mucosa–associated lymphoid tissue lymphoma, better eradication methods should be highlighted, researchers led by William Chey, MD, professor of medicine and director of the GI Physiology Laboratory at Michigan Medicine in Ann Arbor, wrote in Gastroenterology.
In a multicenter, randomized, controlled, phase 3 trial, the research team studied 1,046 treatment-naive adults with H. pylori infection at 103 sites in the U.S., the U.K., Bulgaria, the Czech Republic, Hungary, and Poland between December 2019 and January 2021.
The patients were randomized to receive open-label vonoprazan dual therapy or a double-blind triple therapy twice a day for 14 days. The vonoprazan dual therapy consisted of 20 mg of vonoprazan twice daily and 1 gram of amoxicillin three times per day. The triple therapy consisted of 20 mg of vonoprazan or 30 mg of lansoprazole (standard care), each given with 1 gram of amoxicillin and 500 mg of clarithromycin.
The primary outcome assessed noninferiority in eradication rates in patients without clarithromycin- and amoxicillin-resistant strains, with a noninferiority margin of 10%. Secondary outcomes assessed the superiority in eradication rates in clarithromycin-resistant infections, as well as in all patients.
Eradication rates for nonresistant strains were 84.7% for vonoprazan triple therapy and 78.5% for vonoprazan dual therapy, compared with 78.8% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered noninferior to standard therapy.
The eradication rates in clarithromycin-resistant infections were 65.8% for vonoprazan triple therapy and 69.6% in vonoprazan dual therapy, compared with 31.9% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered superior to standard therapy, with a difference of 33.9 percentage points for triple therapy and 37.7 percentage points for dual therapy.
In all patients, the eradication rates were 80.8% for vonoprazan triple therapy and 77.2% for vonoprazan dual therapy, compared with 68.5% for lansoprazole triple therapy. The rates for both vonoprazan therapies were considered superior, with a difference of 12.3 percentage points for triple therapy and 8.7 percentage points for dual therapy.
Treatment-emergent adverse events were reported in 34.1% of patients in the vonoprazan triple therapy group and 29.9% of patients in the vonoprazan dual therapy group, compared with 34.5% in the lansoprazole triple-therapy group. Most adverse events were mild to moderate.
Serious adverse events occurred in 1.3% of the overall study population, including 1.7% of the vonoprazan triple therapy group, 1.4% of the vonoprazan dual therapy group, and 0.9% of the lansoprazole triple therapy group. None were considered related to the study drugs.
Vonoprazan was approved for the treatment of H. pylori infections by the FDA in May 2022, and had already been approved for treatment of H. pylori infections and other acid-related diseases in several other countries. It decreases intragastric pH and maintains it to a greater degree than that of proton pump inhibitors, which has been associated with higher eradication rates, the authors wrote.
“Optimizing current regimens offers the potential to increase eradication rates and reduce additional antibiotic usage, thereby promoting and improving antimicrobial stewardship,” the study authors wrote.
The study was funded by Phathom Pharmaceuticals, which contributed to the design and conduct of the trial, collection and interpretation of the data, preparation and review of the manuscript, and the decision to submit the manuscript for publication. The study authors declared various conflicts of interest, including some who have received compensation as a consultant, advisory committee member, or employee for Phathom Pharmaceuticals.
FROM GASTROENTEROLOGY
Jury out on synbiotics for kids with GI disorders
That’s the conclusion of a position paper from the European Society for Pediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) special interest group on gut microbiota and modifications and its working group for pre- and probiotics.
Based on their review of available data, the group could not offer a recommendation on use of any specific synbiotic preparation for treatment of acute gastroenteritis, Helicobacter pylori (H. pylori) infection, inflammatory bowel disease (IBD), infantile colic, functional abdominal pain, irritable bowel syndrome (IBS), or constipation.
No recommendation can be formulated on their use in the prevention of food allergies, the group also says.
The same goes for prevention of necrotizing enterocolitis (NEC) in preterm infants and newborns with cyanotic congenital heart disease, as well as prevention of food allergies.
The position paper was published online in the Journal of Pediatric Gastroenterology and Nutrition.
Few studies, major limitations
A synbiotic mixture comprises probiotics and prebiotics selectively utilized by host microorganisms that confers a health benefit on the host.
While the number of studies evaluating the effect of different synbiotics is increasing, the results to date are “ambiguous,” report first author Iva Hojsak, with Children’s Hospital Zagreb, Croatia, and colleagues. Well-designed studies using the same outcome measures for specific clinical indications are needed to allow comparison between studies, they write.
To gauge their effect on pediatric GI disorders, the group searched the literature for studies in English that compared the use of synbiotics, in all delivery vehicles and formulations, at any dose, with no synbiotic (placebo or no treatment or other interventions).
They found very few studies that addressed the specific indications of interest, ranging from two randomized controlled trials (RCTs) each for infantile colic and IBD to five RCTs for acute gastroenteritis.
There were only two indications (acute gastroenteritis and H. pylori) where two synbiotic preparations were tested.
The studies often included a limited number of participants, had significant methodological biases, scarcely reported on side effects or adverse events, and reported different outcomes, making inter-study comparisons tough.
“Comparison of studies was further limited by the synbiotic preparation used, where dose effect was not assessed,” the group writes. Also, few studies used the same synbiotic preparation for a specific clinical indication or the same amount of live bacteria and prebiotic in the preparation.
The authors made note of the newly stringent recommendations for direct evidence proposed by the International Scientific Association for Probiotics and Prebiotics, which state RCTs need to compare the synergistic synbiotic combination, the substrate alone, the live microorganisms alone, and a control.
Outside experts weigh in
Offering outside perspective, Gail Cresci, PhD, microbiome researcher, department of pediatric gastroenterology, hepatology, and nutrition, Cleveland Clinic Children’s Hospital, said what’s “most notable with this review is that there is an issue with studies that incorporate a prebiotic and probiotic, in that there is much heterogeneity with the probiotic strains and prebiotic substrates that are investigated.”
Dr. Cresci also noted that “both pre- and probiotics have specific mechanisms of action based on the substrate and strain, respectively, so to pull the trials together and analyze as a ‘synbiotic’ treating all the combinations the same is not accurate [and] also limits the ability to do meta-analyses and make recommendations in a position paper.”
Geoffrey Preidis, MD, PhD, spokesperson for the American Gastroenterological Association (AGA), also reviewed the paper for this news organization.
He noted that few studies tested the exact same synbiotic preparation for a given clinical indication.
“For the majority of GI disorders examined in this review, the total number of studies testing a particular synbiotic formulation is exactly one. Clinical recommendations rarely can be made based on the results of a single trial,” said Dr. Preidis, a pediatric gastroenterologist with Texas Children’s Hospital, Houston.
“Perhaps most importantly, most studies do not report safety data as rigorously as these data are reported in pharmaceutical trials, so the risk of side effects could be higher than we think,” he noted.
“Millions of Americans take probiotics. They are the third most commonly used dietary supplement behind vitamins and minerals,” Dr. Preidis added. “Prebiotics and synbiotics also are increasing in popularity. They can be found almost everywhere – in supermarkets, drugstores, health food stores, and online – in pill or powder form and in some foods and beverages.
None of these products have been approved by the [U.S. Food and Drug Administration] to treat, cure, or prevent any disease. In most circumstances, there is not enough clinical evidence to suggest a clear value to be gained for most consumers,” he said.
Dr. Preidis said he agrees with the conclusions of this “thoughtfully written position paper” on whether synbiotics have a role in the management of GI disorders in children.
“Synbiotics should not be given routinely to infants or children to manage GI disorders at this time,” he said in an interview. “Potential beneficial effects are not yet confirmed in multiple well-designed, adequately powered trials that test the same synbiotic combination and dose, measure the same outcomes, and rigorously document all adverse effects.”
This research had no specific funding. Dr. Hojsak received payment/honorarium for lectures from BioGaia, Nutricia, Biocodex, AbelaPharm, Nestle, Abbott, Sandoz, Oktal Pharma, and Takeda. Dr. Cresci and Dr. Preidis report no relevant disclosures.
A version of this article first appeared on Medscape.com.
That’s the conclusion of a position paper from the European Society for Pediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) special interest group on gut microbiota and modifications and its working group for pre- and probiotics.
Based on their review of available data, the group could not offer a recommendation on use of any specific synbiotic preparation for treatment of acute gastroenteritis, Helicobacter pylori (H. pylori) infection, inflammatory bowel disease (IBD), infantile colic, functional abdominal pain, irritable bowel syndrome (IBS), or constipation.
No recommendation can be formulated on their use in the prevention of food allergies, the group also says.
The same goes for prevention of necrotizing enterocolitis (NEC) in preterm infants and newborns with cyanotic congenital heart disease, as well as prevention of food allergies.
The position paper was published online in the Journal of Pediatric Gastroenterology and Nutrition.
Few studies, major limitations
A synbiotic mixture comprises probiotics and prebiotics selectively utilized by host microorganisms that confers a health benefit on the host.
While the number of studies evaluating the effect of different synbiotics is increasing, the results to date are “ambiguous,” report first author Iva Hojsak, with Children’s Hospital Zagreb, Croatia, and colleagues. Well-designed studies using the same outcome measures for specific clinical indications are needed to allow comparison between studies, they write.
To gauge their effect on pediatric GI disorders, the group searched the literature for studies in English that compared the use of synbiotics, in all delivery vehicles and formulations, at any dose, with no synbiotic (placebo or no treatment or other interventions).
They found very few studies that addressed the specific indications of interest, ranging from two randomized controlled trials (RCTs) each for infantile colic and IBD to five RCTs for acute gastroenteritis.
There were only two indications (acute gastroenteritis and H. pylori) where two synbiotic preparations were tested.
The studies often included a limited number of participants, had significant methodological biases, scarcely reported on side effects or adverse events, and reported different outcomes, making inter-study comparisons tough.
“Comparison of studies was further limited by the synbiotic preparation used, where dose effect was not assessed,” the group writes. Also, few studies used the same synbiotic preparation for a specific clinical indication or the same amount of live bacteria and prebiotic in the preparation.
The authors made note of the newly stringent recommendations for direct evidence proposed by the International Scientific Association for Probiotics and Prebiotics, which state RCTs need to compare the synergistic synbiotic combination, the substrate alone, the live microorganisms alone, and a control.
Outside experts weigh in
Offering outside perspective, Gail Cresci, PhD, microbiome researcher, department of pediatric gastroenterology, hepatology, and nutrition, Cleveland Clinic Children’s Hospital, said what’s “most notable with this review is that there is an issue with studies that incorporate a prebiotic and probiotic, in that there is much heterogeneity with the probiotic strains and prebiotic substrates that are investigated.”
Dr. Cresci also noted that “both pre- and probiotics have specific mechanisms of action based on the substrate and strain, respectively, so to pull the trials together and analyze as a ‘synbiotic’ treating all the combinations the same is not accurate [and] also limits the ability to do meta-analyses and make recommendations in a position paper.”
Geoffrey Preidis, MD, PhD, spokesperson for the American Gastroenterological Association (AGA), also reviewed the paper for this news organization.
He noted that few studies tested the exact same synbiotic preparation for a given clinical indication.
“For the majority of GI disorders examined in this review, the total number of studies testing a particular synbiotic formulation is exactly one. Clinical recommendations rarely can be made based on the results of a single trial,” said Dr. Preidis, a pediatric gastroenterologist with Texas Children’s Hospital, Houston.
“Perhaps most importantly, most studies do not report safety data as rigorously as these data are reported in pharmaceutical trials, so the risk of side effects could be higher than we think,” he noted.
“Millions of Americans take probiotics. They are the third most commonly used dietary supplement behind vitamins and minerals,” Dr. Preidis added. “Prebiotics and synbiotics also are increasing in popularity. They can be found almost everywhere – in supermarkets, drugstores, health food stores, and online – in pill or powder form and in some foods and beverages.
None of these products have been approved by the [U.S. Food and Drug Administration] to treat, cure, or prevent any disease. In most circumstances, there is not enough clinical evidence to suggest a clear value to be gained for most consumers,” he said.
Dr. Preidis said he agrees with the conclusions of this “thoughtfully written position paper” on whether synbiotics have a role in the management of GI disorders in children.
“Synbiotics should not be given routinely to infants or children to manage GI disorders at this time,” he said in an interview. “Potential beneficial effects are not yet confirmed in multiple well-designed, adequately powered trials that test the same synbiotic combination and dose, measure the same outcomes, and rigorously document all adverse effects.”
This research had no specific funding. Dr. Hojsak received payment/honorarium for lectures from BioGaia, Nutricia, Biocodex, AbelaPharm, Nestle, Abbott, Sandoz, Oktal Pharma, and Takeda. Dr. Cresci and Dr. Preidis report no relevant disclosures.
A version of this article first appeared on Medscape.com.
That’s the conclusion of a position paper from the European Society for Pediatric Gastroenterology Hepatology and Nutrition (ESPGHAN) special interest group on gut microbiota and modifications and its working group for pre- and probiotics.
Based on their review of available data, the group could not offer a recommendation on use of any specific synbiotic preparation for treatment of acute gastroenteritis, Helicobacter pylori (H. pylori) infection, inflammatory bowel disease (IBD), infantile colic, functional abdominal pain, irritable bowel syndrome (IBS), or constipation.
No recommendation can be formulated on their use in the prevention of food allergies, the group also says.
The same goes for prevention of necrotizing enterocolitis (NEC) in preterm infants and newborns with cyanotic congenital heart disease, as well as prevention of food allergies.
The position paper was published online in the Journal of Pediatric Gastroenterology and Nutrition.
Few studies, major limitations
A synbiotic mixture comprises probiotics and prebiotics selectively utilized by host microorganisms that confers a health benefit on the host.
While the number of studies evaluating the effect of different synbiotics is increasing, the results to date are “ambiguous,” report first author Iva Hojsak, with Children’s Hospital Zagreb, Croatia, and colleagues. Well-designed studies using the same outcome measures for specific clinical indications are needed to allow comparison between studies, they write.
To gauge their effect on pediatric GI disorders, the group searched the literature for studies in English that compared the use of synbiotics, in all delivery vehicles and formulations, at any dose, with no synbiotic (placebo or no treatment or other interventions).
They found very few studies that addressed the specific indications of interest, ranging from two randomized controlled trials (RCTs) each for infantile colic and IBD to five RCTs for acute gastroenteritis.
There were only two indications (acute gastroenteritis and H. pylori) where two synbiotic preparations were tested.
The studies often included a limited number of participants, had significant methodological biases, scarcely reported on side effects or adverse events, and reported different outcomes, making inter-study comparisons tough.
“Comparison of studies was further limited by the synbiotic preparation used, where dose effect was not assessed,” the group writes. Also, few studies used the same synbiotic preparation for a specific clinical indication or the same amount of live bacteria and prebiotic in the preparation.
The authors made note of the newly stringent recommendations for direct evidence proposed by the International Scientific Association for Probiotics and Prebiotics, which state RCTs need to compare the synergistic synbiotic combination, the substrate alone, the live microorganisms alone, and a control.
Outside experts weigh in
Offering outside perspective, Gail Cresci, PhD, microbiome researcher, department of pediatric gastroenterology, hepatology, and nutrition, Cleveland Clinic Children’s Hospital, said what’s “most notable with this review is that there is an issue with studies that incorporate a prebiotic and probiotic, in that there is much heterogeneity with the probiotic strains and prebiotic substrates that are investigated.”
Dr. Cresci also noted that “both pre- and probiotics have specific mechanisms of action based on the substrate and strain, respectively, so to pull the trials together and analyze as a ‘synbiotic’ treating all the combinations the same is not accurate [and] also limits the ability to do meta-analyses and make recommendations in a position paper.”
Geoffrey Preidis, MD, PhD, spokesperson for the American Gastroenterological Association (AGA), also reviewed the paper for this news organization.
He noted that few studies tested the exact same synbiotic preparation for a given clinical indication.
“For the majority of GI disorders examined in this review, the total number of studies testing a particular synbiotic formulation is exactly one. Clinical recommendations rarely can be made based on the results of a single trial,” said Dr. Preidis, a pediatric gastroenterologist with Texas Children’s Hospital, Houston.
“Perhaps most importantly, most studies do not report safety data as rigorously as these data are reported in pharmaceutical trials, so the risk of side effects could be higher than we think,” he noted.
“Millions of Americans take probiotics. They are the third most commonly used dietary supplement behind vitamins and minerals,” Dr. Preidis added. “Prebiotics and synbiotics also are increasing in popularity. They can be found almost everywhere – in supermarkets, drugstores, health food stores, and online – in pill or powder form and in some foods and beverages.
None of these products have been approved by the [U.S. Food and Drug Administration] to treat, cure, or prevent any disease. In most circumstances, there is not enough clinical evidence to suggest a clear value to be gained for most consumers,” he said.
Dr. Preidis said he agrees with the conclusions of this “thoughtfully written position paper” on whether synbiotics have a role in the management of GI disorders in children.
“Synbiotics should not be given routinely to infants or children to manage GI disorders at this time,” he said in an interview. “Potential beneficial effects are not yet confirmed in multiple well-designed, adequately powered trials that test the same synbiotic combination and dose, measure the same outcomes, and rigorously document all adverse effects.”
This research had no specific funding. Dr. Hojsak received payment/honorarium for lectures from BioGaia, Nutricia, Biocodex, AbelaPharm, Nestle, Abbott, Sandoz, Oktal Pharma, and Takeda. Dr. Cresci and Dr. Preidis report no relevant disclosures.
A version of this article first appeared on Medscape.com.
FROM JOURNAL OF PEDIATRIC GASTROENTEROLOGY AND NUTRITION
Genetic counseling for cancer often costs patients nothing
But even among those who do, the cost is $16 or less, a cohort study shows.
“The findings highlight the relatively low financial costs of genetic counseling, a form of care with potentially substantial implications for cancer treatment,” lead author Mya Roberson, PhD, Vanderbilt University, Nashville, Tenn., and colleagues explained.
The study was published online in JAMA Health Forum.
Genetic counseling is an important feature of cancer care that can affect treatment decisions and surveillance. But coverage of genetic counseling services varies across insurance types.
To understand the costs to patients, the investigators used data from IBM Watson Health MarketScan to create a cohort of privately insured patients with breast, prostate, endometrial, ovarian, colorectal, and pancreatic cancer who had at least one genetic counseling session from 2013 to the end of 2019.
Dr. Roberson and colleagues then calculated out-of-pocket costs – including coinsurance, copayments, and deductibles – and total costs paid on claims for genetic counseling encounters. The cohort included 16,791 patients, the majority of whom had breast cancer.
Although the median insurance payment for genetic counseling encounters was $118 ($58-$211), most patients paid nothing out of pocket for these services. Among the 31% of patients with an out-of-pocket expense, the cost was $16 or less.
Compared with breast cancer patients, men with prostate cancer were 28% more likely to have out-of-pocket costs for genetic counseling, which may “reflect a lack of awareness about the medical necessity of genetic counseling,” the authors suggested.
Overall, the study highlights the value of genetic counseling in cancer care.
“Cancer genetic counseling not only promotes informed decision-making about genetic testing and cancer treatment in the era of precision medicine, but it also is a form of low-cost, high-value care,” the authors wrote.
The study was funded by a grant from the National Cancer Institute. Dr. Roberson disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
But even among those who do, the cost is $16 or less, a cohort study shows.
“The findings highlight the relatively low financial costs of genetic counseling, a form of care with potentially substantial implications for cancer treatment,” lead author Mya Roberson, PhD, Vanderbilt University, Nashville, Tenn., and colleagues explained.
The study was published online in JAMA Health Forum.
Genetic counseling is an important feature of cancer care that can affect treatment decisions and surveillance. But coverage of genetic counseling services varies across insurance types.
To understand the costs to patients, the investigators used data from IBM Watson Health MarketScan to create a cohort of privately insured patients with breast, prostate, endometrial, ovarian, colorectal, and pancreatic cancer who had at least one genetic counseling session from 2013 to the end of 2019.
Dr. Roberson and colleagues then calculated out-of-pocket costs – including coinsurance, copayments, and deductibles – and total costs paid on claims for genetic counseling encounters. The cohort included 16,791 patients, the majority of whom had breast cancer.
Although the median insurance payment for genetic counseling encounters was $118 ($58-$211), most patients paid nothing out of pocket for these services. Among the 31% of patients with an out-of-pocket expense, the cost was $16 or less.
Compared with breast cancer patients, men with prostate cancer were 28% more likely to have out-of-pocket costs for genetic counseling, which may “reflect a lack of awareness about the medical necessity of genetic counseling,” the authors suggested.
Overall, the study highlights the value of genetic counseling in cancer care.
“Cancer genetic counseling not only promotes informed decision-making about genetic testing and cancer treatment in the era of precision medicine, but it also is a form of low-cost, high-value care,” the authors wrote.
The study was funded by a grant from the National Cancer Institute. Dr. Roberson disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
But even among those who do, the cost is $16 or less, a cohort study shows.
“The findings highlight the relatively low financial costs of genetic counseling, a form of care with potentially substantial implications for cancer treatment,” lead author Mya Roberson, PhD, Vanderbilt University, Nashville, Tenn., and colleagues explained.
The study was published online in JAMA Health Forum.
Genetic counseling is an important feature of cancer care that can affect treatment decisions and surveillance. But coverage of genetic counseling services varies across insurance types.
To understand the costs to patients, the investigators used data from IBM Watson Health MarketScan to create a cohort of privately insured patients with breast, prostate, endometrial, ovarian, colorectal, and pancreatic cancer who had at least one genetic counseling session from 2013 to the end of 2019.
Dr. Roberson and colleagues then calculated out-of-pocket costs – including coinsurance, copayments, and deductibles – and total costs paid on claims for genetic counseling encounters. The cohort included 16,791 patients, the majority of whom had breast cancer.
Although the median insurance payment for genetic counseling encounters was $118 ($58-$211), most patients paid nothing out of pocket for these services. Among the 31% of patients with an out-of-pocket expense, the cost was $16 or less.
Compared with breast cancer patients, men with prostate cancer were 28% more likely to have out-of-pocket costs for genetic counseling, which may “reflect a lack of awareness about the medical necessity of genetic counseling,” the authors suggested.
Overall, the study highlights the value of genetic counseling in cancer care.
“Cancer genetic counseling not only promotes informed decision-making about genetic testing and cancer treatment in the era of precision medicine, but it also is a form of low-cost, high-value care,” the authors wrote.
The study was funded by a grant from the National Cancer Institute. Dr. Roberson disclosed no relevant financial relationships.
A version of this article first appeared on Medscape.com.
FROM JAMA HEALTH FORUM
Updates on treatment/prevention of VTE in cancer patients
Updated clinical practice guidelines for the treatment and prevention of venous thromboembolism for patients with cancer, including those with cancer and COVID-19, have been released by the International Initiative on Thrombosis and Cancer (ITAC), an academic working group of VTE experts.
“Because patients with cancer have a baseline increased risk of VTE, compared with patients without cancer, the combination of both COVID-19 and cancer – and its effect on VTE risk and treatment – is of concern,” said the authors, led by Dominique Farge, MD, PhD, Nord Universite de Paris.
they added.
The new guidelines were published online in The Lancet Oncology.
“Cancer-associated VTE remains an important clinical problem, associated with increased morbidity and mortality,” Dr. Farge and colleagues observed.
“The ITAC guidelines’ companion free web-based mobile application will assist the practicing clinician with decision making at various levels to provide optimal care of patients with cancer to treat and prevent VTE,” they emphasized. More information is available at itaccme.com.
Cancer patients with COVID
The new section of the guidelines notes that the treatment and prevention of VTE for cancer patients infected with SARS-CoV-2 remain the same as for patients without COVID.
Whether or not cancer patients with COVID-19 are hospitalized, have been discharged, or are ambulatory, they should be assessed for the risk of VTE, as should any other patient. For cancer patients with COVID-19 who are hospitalized, pharmacologic prophylaxis should be given at the same dose and anticoagulant type as for hospitalized cancer patients who do not have COVID-19.
Following discharge, VTE prophylaxis is not advised for cancer patients infected with SARS-CoV-2, and routine primary pharmacologic prophylaxis of VTE for ambulatory patients with COVID-19 is also not recommended, the authors noted.
Initial treatment of established VTE
Initial treatment of established VTE for up to 10 days of anticoagulation should include low-molecular-weight heparin (LMWH) when creatinine clearance is at least 30 mL/min.
“A regimen of LMWH, taken once per day, is recommended unless a twice-per-day regimen is required because of patients’ characteristics,” the authors noted. These characteristics include a high risk of bleeding, moderate renal failure, and the need for technical intervention, including surgery.
If a twice-a-day regimen is required, only enoxaparin at a dose of 1 mg/kg twice daily can be used, the authors cautioned.
For patients with a low risk of gastrointestinal or genitourinary bleeding, rivaroxaban (Xarelto) or apixaban (Eliquis) can be given in the first 10 days, as well as edoxaban (Lixiana). The latter should be started after at least 5 days of parenteral anticoagulation, provided creatinine clearance is at least 30 mL/min.
“Unfractionated heparin as well as fondaparinux (GlaxoSmithKline) can be also used for the initial treatment of established VTE when LMWH or direct oral anticoagulants are contraindicated,” Dr. Farge and colleagues wrote.
Thrombolysis can be considered on a case-by-case basis, although physicians must pay attention to specific contraindications, especially bleeding risk.
“In the initial treatment of VTE, inferior vena cava filters might be considered when anticoagulant treatment is contraindicated or, in the case of pulmonary embolism, when recurrence occurs under optimal anticoagulation,” the authors noted.
Maintenance VTE treatment
For maintenance therapy, which the authors define as early maintenance for up to 6 months and long-term maintenance beyond 6 months, they point out that LMWHs are preferred over vitamin K antagonists for the treatment of VTE when the creatinine clearance is again at least 30 mL/min.
Any of the direct oral anticoagulants (DOAs) – edoxaban, rivaroxaban, or apixaban – is also recommended for the same patients, provided there is no risk of inducing a strong drug-drug interaction or GI absorption is impaired.
However, the DOAs should be used with caution for patients with GI malignancies, especially upper GI cancers, because data show there is an increased risk of GI bleeding with both edoxaban and rivaroxaban.
“LMWH or direct oral anticoagulants should be used for a minimum of 6 months to treat established VTE in patients with cancer,” the authors wrote.
“After 6 months, termination or continuation of anticoagulation (LMWH, direct oral anticoagulants, or vitamin K antagonists) should be based on individual evaluation of the benefit-risk ratio,” they added.
Treatment of VTE recurrence
The guideline authors explain that three options can be considered in the event of VTE recurrence. These include an increase in the LMWH dose by 20%-25%, or a switch to a DOA, or, if patients are taking a DOA, a switch to an LMWH. If the patient is taking a vitamin K antagonist, it can be switched to either an LMWH or a DOA.
For treatment of catheter-related thrombosis, anticoagulant treatment is recommended for a minimum of 3 months and as long as the central venous catheter is in place. In this setting, the LMWHs are recommended.
The central venous catheter can be kept in place if it is functional, well positioned, and is not infected, provided there is good resolution of symptoms under close surveillance while anticoagulants are being administered.
In surgically treated patients, the LMWH, given once a day, to patients with a serum creatinine concentration of at least 30 mL/min can be used to prevent VTE. Alternatively, VTE can be prevented by the use low-dose unfractionated heparin, given three times a day.
“Pharmacological prophylaxis should be started 2-12 h preoperatively and continued for at least 7–10 days,” Dr. Farge and colleagues advised. In this setting, there is insufficient evidence to support the use of fondaparinux or a DOA as an alternative to an LMWH for the prophylaxis of postoperative VTE. “Use of the highest prophylactic dose of LMWH to prevent postoperative VTE in patients with cancer is recommended,” the authors advised.
Furthermore, extended prophylaxis of at least 4 weeks with LMWH is advised to prevent postoperative VTE after major abdominal or pelvic surgery. Mechanical methods are not recommended except when pharmacologic methods are contraindicated. Inferior vena cava filters are also not recommended for routine prophylaxis.
Patients with reduced mobility
For medically treated hospitalized patients with cancer whose mobility is reduced, the authors recommend prophylaxis with either an LMWH or fondaparinux, provided their creatinine clearance is at least 30 mL/min. These patients can also be treated with unfractionated heparin, they add.
In contrast, DOAs are not recommended – at least not routinely – in this setting, the authors cautioned. Primary pharmacologic prophylaxis of VTE with either LMWH or DOAs – either rivaroxaban or apixaban – is indicated in ambulatory patients with locally advanced or metastatic pancreatic cancer who are receiving systemic anticancer therapy, provided they are at low risk of bleeding.
However, primary pharmacologic prophylaxis with LMWH is not recommended outside of a clinical trial for patients with locally advanced or metastatic lung cancer who are undergoing systemic anticancer therapy, even for patients who are at low risk of bleeding.
For ambulatory patients who are receiving systemic anticancer therapy and who are at intermediate risk of VTE, primary prophylaxis with rivaroxaban or apixaban is recommended for those with myeloma who are receiving immunomodulatory therapy plus steroids or other systemic therapies.
In this setting, oral anticoagulants should consist of a vitamin K antagonist, given at low or therapeutic doses, or apixaban, given at prophylactic doses. Alternatively, LMWH, given at prophylactic doses, or low-dose aspirin, given at a dose of 100 mg/day, can be used.
Catheter-related thrombosis
Use of anticoagulation for routine prophylaxis of catheter-related thrombosis is not recommended. Catheters should be inserted on the right side in the jugular vein, and the distal extremity of the central catheter should be located at the junction of the superior vena cava and the right atrium. “In patients requiring central venous catheters, we suggest the use of implanted ports over peripheral inserted central catheter lines,” the authors noted.
The authors described a number of unique situations regarding the treatment of VTE. These situations include patients with a brain tumor, for whom treatment of established VTE should favor either LMWH or a DOA. The authors also recommended the use of LMWH or unfractionated heparin, started postoperatively, for the prevention of VTE for patients undergoing neurosurgery.
In contrast, pharmacologic prophylaxis of VTE in medically treated patients with a brain tumor who are not undergoing neurosurgery is not recommended. “In the presence of severe renal failure...we suggest using unfractionated heparin followed by early vitamin K antagonists (possibly from day 1) or LMWH adjusted to anti-Xa concentration of the treatment of established VTE,” Dr. Farge and colleagues wrote.
Anticoagulant treatment is also recommended for a minimum of 3 months for children with symptomatic catheter-related thrombosis and as long as the central venous catheter is in place. For children with acute lymphoblastic leukemia who are undergoing induction chemotherapy, LMWH is also recommended as thromboprophylaxis.
For children who require a central venous catheter, the authors suggested that physicians use implanted ports over peripherally inserted central lines.
A version of this article first appeared on Medscape.com.
Updated clinical practice guidelines for the treatment and prevention of venous thromboembolism for patients with cancer, including those with cancer and COVID-19, have been released by the International Initiative on Thrombosis and Cancer (ITAC), an academic working group of VTE experts.
“Because patients with cancer have a baseline increased risk of VTE, compared with patients without cancer, the combination of both COVID-19 and cancer – and its effect on VTE risk and treatment – is of concern,” said the authors, led by Dominique Farge, MD, PhD, Nord Universite de Paris.
they added.
The new guidelines were published online in The Lancet Oncology.
“Cancer-associated VTE remains an important clinical problem, associated with increased morbidity and mortality,” Dr. Farge and colleagues observed.
“The ITAC guidelines’ companion free web-based mobile application will assist the practicing clinician with decision making at various levels to provide optimal care of patients with cancer to treat and prevent VTE,” they emphasized. More information is available at itaccme.com.
Cancer patients with COVID
The new section of the guidelines notes that the treatment and prevention of VTE for cancer patients infected with SARS-CoV-2 remain the same as for patients without COVID.
Whether or not cancer patients with COVID-19 are hospitalized, have been discharged, or are ambulatory, they should be assessed for the risk of VTE, as should any other patient. For cancer patients with COVID-19 who are hospitalized, pharmacologic prophylaxis should be given at the same dose and anticoagulant type as for hospitalized cancer patients who do not have COVID-19.
Following discharge, VTE prophylaxis is not advised for cancer patients infected with SARS-CoV-2, and routine primary pharmacologic prophylaxis of VTE for ambulatory patients with COVID-19 is also not recommended, the authors noted.
Initial treatment of established VTE
Initial treatment of established VTE for up to 10 days of anticoagulation should include low-molecular-weight heparin (LMWH) when creatinine clearance is at least 30 mL/min.
“A regimen of LMWH, taken once per day, is recommended unless a twice-per-day regimen is required because of patients’ characteristics,” the authors noted. These characteristics include a high risk of bleeding, moderate renal failure, and the need for technical intervention, including surgery.
If a twice-a-day regimen is required, only enoxaparin at a dose of 1 mg/kg twice daily can be used, the authors cautioned.
For patients with a low risk of gastrointestinal or genitourinary bleeding, rivaroxaban (Xarelto) or apixaban (Eliquis) can be given in the first 10 days, as well as edoxaban (Lixiana). The latter should be started after at least 5 days of parenteral anticoagulation, provided creatinine clearance is at least 30 mL/min.
“Unfractionated heparin as well as fondaparinux (GlaxoSmithKline) can be also used for the initial treatment of established VTE when LMWH or direct oral anticoagulants are contraindicated,” Dr. Farge and colleagues wrote.
Thrombolysis can be considered on a case-by-case basis, although physicians must pay attention to specific contraindications, especially bleeding risk.
“In the initial treatment of VTE, inferior vena cava filters might be considered when anticoagulant treatment is contraindicated or, in the case of pulmonary embolism, when recurrence occurs under optimal anticoagulation,” the authors noted.
Maintenance VTE treatment
For maintenance therapy, which the authors define as early maintenance for up to 6 months and long-term maintenance beyond 6 months, they point out that LMWHs are preferred over vitamin K antagonists for the treatment of VTE when the creatinine clearance is again at least 30 mL/min.
Any of the direct oral anticoagulants (DOAs) – edoxaban, rivaroxaban, or apixaban – is also recommended for the same patients, provided there is no risk of inducing a strong drug-drug interaction or GI absorption is impaired.
However, the DOAs should be used with caution for patients with GI malignancies, especially upper GI cancers, because data show there is an increased risk of GI bleeding with both edoxaban and rivaroxaban.
“LMWH or direct oral anticoagulants should be used for a minimum of 6 months to treat established VTE in patients with cancer,” the authors wrote.
“After 6 months, termination or continuation of anticoagulation (LMWH, direct oral anticoagulants, or vitamin K antagonists) should be based on individual evaluation of the benefit-risk ratio,” they added.
Treatment of VTE recurrence
The guideline authors explain that three options can be considered in the event of VTE recurrence. These include an increase in the LMWH dose by 20%-25%, or a switch to a DOA, or, if patients are taking a DOA, a switch to an LMWH. If the patient is taking a vitamin K antagonist, it can be switched to either an LMWH or a DOA.
For treatment of catheter-related thrombosis, anticoagulant treatment is recommended for a minimum of 3 months and as long as the central venous catheter is in place. In this setting, the LMWHs are recommended.
The central venous catheter can be kept in place if it is functional, well positioned, and is not infected, provided there is good resolution of symptoms under close surveillance while anticoagulants are being administered.
In surgically treated patients, the LMWH, given once a day, to patients with a serum creatinine concentration of at least 30 mL/min can be used to prevent VTE. Alternatively, VTE can be prevented by the use low-dose unfractionated heparin, given three times a day.
“Pharmacological prophylaxis should be started 2-12 h preoperatively and continued for at least 7–10 days,” Dr. Farge and colleagues advised. In this setting, there is insufficient evidence to support the use of fondaparinux or a DOA as an alternative to an LMWH for the prophylaxis of postoperative VTE. “Use of the highest prophylactic dose of LMWH to prevent postoperative VTE in patients with cancer is recommended,” the authors advised.
Furthermore, extended prophylaxis of at least 4 weeks with LMWH is advised to prevent postoperative VTE after major abdominal or pelvic surgery. Mechanical methods are not recommended except when pharmacologic methods are contraindicated. Inferior vena cava filters are also not recommended for routine prophylaxis.
Patients with reduced mobility
For medically treated hospitalized patients with cancer whose mobility is reduced, the authors recommend prophylaxis with either an LMWH or fondaparinux, provided their creatinine clearance is at least 30 mL/min. These patients can also be treated with unfractionated heparin, they add.
In contrast, DOAs are not recommended – at least not routinely – in this setting, the authors cautioned. Primary pharmacologic prophylaxis of VTE with either LMWH or DOAs – either rivaroxaban or apixaban – is indicated in ambulatory patients with locally advanced or metastatic pancreatic cancer who are receiving systemic anticancer therapy, provided they are at low risk of bleeding.
However, primary pharmacologic prophylaxis with LMWH is not recommended outside of a clinical trial for patients with locally advanced or metastatic lung cancer who are undergoing systemic anticancer therapy, even for patients who are at low risk of bleeding.
For ambulatory patients who are receiving systemic anticancer therapy and who are at intermediate risk of VTE, primary prophylaxis with rivaroxaban or apixaban is recommended for those with myeloma who are receiving immunomodulatory therapy plus steroids or other systemic therapies.
In this setting, oral anticoagulants should consist of a vitamin K antagonist, given at low or therapeutic doses, or apixaban, given at prophylactic doses. Alternatively, LMWH, given at prophylactic doses, or low-dose aspirin, given at a dose of 100 mg/day, can be used.
Catheter-related thrombosis
Use of anticoagulation for routine prophylaxis of catheter-related thrombosis is not recommended. Catheters should be inserted on the right side in the jugular vein, and the distal extremity of the central catheter should be located at the junction of the superior vena cava and the right atrium. “In patients requiring central venous catheters, we suggest the use of implanted ports over peripheral inserted central catheter lines,” the authors noted.
The authors described a number of unique situations regarding the treatment of VTE. These situations include patients with a brain tumor, for whom treatment of established VTE should favor either LMWH or a DOA. The authors also recommended the use of LMWH or unfractionated heparin, started postoperatively, for the prevention of VTE for patients undergoing neurosurgery.
In contrast, pharmacologic prophylaxis of VTE in medically treated patients with a brain tumor who are not undergoing neurosurgery is not recommended. “In the presence of severe renal failure...we suggest using unfractionated heparin followed by early vitamin K antagonists (possibly from day 1) or LMWH adjusted to anti-Xa concentration of the treatment of established VTE,” Dr. Farge and colleagues wrote.
Anticoagulant treatment is also recommended for a minimum of 3 months for children with symptomatic catheter-related thrombosis and as long as the central venous catheter is in place. For children with acute lymphoblastic leukemia who are undergoing induction chemotherapy, LMWH is also recommended as thromboprophylaxis.
For children who require a central venous catheter, the authors suggested that physicians use implanted ports over peripherally inserted central lines.
A version of this article first appeared on Medscape.com.
Updated clinical practice guidelines for the treatment and prevention of venous thromboembolism for patients with cancer, including those with cancer and COVID-19, have been released by the International Initiative on Thrombosis and Cancer (ITAC), an academic working group of VTE experts.
“Because patients with cancer have a baseline increased risk of VTE, compared with patients without cancer, the combination of both COVID-19 and cancer – and its effect on VTE risk and treatment – is of concern,” said the authors, led by Dominique Farge, MD, PhD, Nord Universite de Paris.
they added.
The new guidelines were published online in The Lancet Oncology.
“Cancer-associated VTE remains an important clinical problem, associated with increased morbidity and mortality,” Dr. Farge and colleagues observed.
“The ITAC guidelines’ companion free web-based mobile application will assist the practicing clinician with decision making at various levels to provide optimal care of patients with cancer to treat and prevent VTE,” they emphasized. More information is available at itaccme.com.
Cancer patients with COVID
The new section of the guidelines notes that the treatment and prevention of VTE for cancer patients infected with SARS-CoV-2 remain the same as for patients without COVID.
Whether or not cancer patients with COVID-19 are hospitalized, have been discharged, or are ambulatory, they should be assessed for the risk of VTE, as should any other patient. For cancer patients with COVID-19 who are hospitalized, pharmacologic prophylaxis should be given at the same dose and anticoagulant type as for hospitalized cancer patients who do not have COVID-19.
Following discharge, VTE prophylaxis is not advised for cancer patients infected with SARS-CoV-2, and routine primary pharmacologic prophylaxis of VTE for ambulatory patients with COVID-19 is also not recommended, the authors noted.
Initial treatment of established VTE
Initial treatment of established VTE for up to 10 days of anticoagulation should include low-molecular-weight heparin (LMWH) when creatinine clearance is at least 30 mL/min.
“A regimen of LMWH, taken once per day, is recommended unless a twice-per-day regimen is required because of patients’ characteristics,” the authors noted. These characteristics include a high risk of bleeding, moderate renal failure, and the need for technical intervention, including surgery.
If a twice-a-day regimen is required, only enoxaparin at a dose of 1 mg/kg twice daily can be used, the authors cautioned.
For patients with a low risk of gastrointestinal or genitourinary bleeding, rivaroxaban (Xarelto) or apixaban (Eliquis) can be given in the first 10 days, as well as edoxaban (Lixiana). The latter should be started after at least 5 days of parenteral anticoagulation, provided creatinine clearance is at least 30 mL/min.
“Unfractionated heparin as well as fondaparinux (GlaxoSmithKline) can be also used for the initial treatment of established VTE when LMWH or direct oral anticoagulants are contraindicated,” Dr. Farge and colleagues wrote.
Thrombolysis can be considered on a case-by-case basis, although physicians must pay attention to specific contraindications, especially bleeding risk.
“In the initial treatment of VTE, inferior vena cava filters might be considered when anticoagulant treatment is contraindicated or, in the case of pulmonary embolism, when recurrence occurs under optimal anticoagulation,” the authors noted.
Maintenance VTE treatment
For maintenance therapy, which the authors define as early maintenance for up to 6 months and long-term maintenance beyond 6 months, they point out that LMWHs are preferred over vitamin K antagonists for the treatment of VTE when the creatinine clearance is again at least 30 mL/min.
Any of the direct oral anticoagulants (DOAs) – edoxaban, rivaroxaban, or apixaban – is also recommended for the same patients, provided there is no risk of inducing a strong drug-drug interaction or GI absorption is impaired.
However, the DOAs should be used with caution for patients with GI malignancies, especially upper GI cancers, because data show there is an increased risk of GI bleeding with both edoxaban and rivaroxaban.
“LMWH or direct oral anticoagulants should be used for a minimum of 6 months to treat established VTE in patients with cancer,” the authors wrote.
“After 6 months, termination or continuation of anticoagulation (LMWH, direct oral anticoagulants, or vitamin K antagonists) should be based on individual evaluation of the benefit-risk ratio,” they added.
Treatment of VTE recurrence
The guideline authors explain that three options can be considered in the event of VTE recurrence. These include an increase in the LMWH dose by 20%-25%, or a switch to a DOA, or, if patients are taking a DOA, a switch to an LMWH. If the patient is taking a vitamin K antagonist, it can be switched to either an LMWH or a DOA.
For treatment of catheter-related thrombosis, anticoagulant treatment is recommended for a minimum of 3 months and as long as the central venous catheter is in place. In this setting, the LMWHs are recommended.
The central venous catheter can be kept in place if it is functional, well positioned, and is not infected, provided there is good resolution of symptoms under close surveillance while anticoagulants are being administered.
In surgically treated patients, the LMWH, given once a day, to patients with a serum creatinine concentration of at least 30 mL/min can be used to prevent VTE. Alternatively, VTE can be prevented by the use low-dose unfractionated heparin, given three times a day.
“Pharmacological prophylaxis should be started 2-12 h preoperatively and continued for at least 7–10 days,” Dr. Farge and colleagues advised. In this setting, there is insufficient evidence to support the use of fondaparinux or a DOA as an alternative to an LMWH for the prophylaxis of postoperative VTE. “Use of the highest prophylactic dose of LMWH to prevent postoperative VTE in patients with cancer is recommended,” the authors advised.
Furthermore, extended prophylaxis of at least 4 weeks with LMWH is advised to prevent postoperative VTE after major abdominal or pelvic surgery. Mechanical methods are not recommended except when pharmacologic methods are contraindicated. Inferior vena cava filters are also not recommended for routine prophylaxis.
Patients with reduced mobility
For medically treated hospitalized patients with cancer whose mobility is reduced, the authors recommend prophylaxis with either an LMWH or fondaparinux, provided their creatinine clearance is at least 30 mL/min. These patients can also be treated with unfractionated heparin, they add.
In contrast, DOAs are not recommended – at least not routinely – in this setting, the authors cautioned. Primary pharmacologic prophylaxis of VTE with either LMWH or DOAs – either rivaroxaban or apixaban – is indicated in ambulatory patients with locally advanced or metastatic pancreatic cancer who are receiving systemic anticancer therapy, provided they are at low risk of bleeding.
However, primary pharmacologic prophylaxis with LMWH is not recommended outside of a clinical trial for patients with locally advanced or metastatic lung cancer who are undergoing systemic anticancer therapy, even for patients who are at low risk of bleeding.
For ambulatory patients who are receiving systemic anticancer therapy and who are at intermediate risk of VTE, primary prophylaxis with rivaroxaban or apixaban is recommended for those with myeloma who are receiving immunomodulatory therapy plus steroids or other systemic therapies.
In this setting, oral anticoagulants should consist of a vitamin K antagonist, given at low or therapeutic doses, or apixaban, given at prophylactic doses. Alternatively, LMWH, given at prophylactic doses, or low-dose aspirin, given at a dose of 100 mg/day, can be used.
Catheter-related thrombosis
Use of anticoagulation for routine prophylaxis of catheter-related thrombosis is not recommended. Catheters should be inserted on the right side in the jugular vein, and the distal extremity of the central catheter should be located at the junction of the superior vena cava and the right atrium. “In patients requiring central venous catheters, we suggest the use of implanted ports over peripheral inserted central catheter lines,” the authors noted.
The authors described a number of unique situations regarding the treatment of VTE. These situations include patients with a brain tumor, for whom treatment of established VTE should favor either LMWH or a DOA. The authors also recommended the use of LMWH or unfractionated heparin, started postoperatively, for the prevention of VTE for patients undergoing neurosurgery.
In contrast, pharmacologic prophylaxis of VTE in medically treated patients with a brain tumor who are not undergoing neurosurgery is not recommended. “In the presence of severe renal failure...we suggest using unfractionated heparin followed by early vitamin K antagonists (possibly from day 1) or LMWH adjusted to anti-Xa concentration of the treatment of established VTE,” Dr. Farge and colleagues wrote.
Anticoagulant treatment is also recommended for a minimum of 3 months for children with symptomatic catheter-related thrombosis and as long as the central venous catheter is in place. For children with acute lymphoblastic leukemia who are undergoing induction chemotherapy, LMWH is also recommended as thromboprophylaxis.
For children who require a central venous catheter, the authors suggested that physicians use implanted ports over peripherally inserted central lines.
A version of this article first appeared on Medscape.com.
FROM THE LANCET ONCOLOGY
Firm Exophytic Tumor on the Shin
The Diagnosis: Leiomyosarcoma
Cutaneous leiomyosarcomas are relatively rare neoplasms that favor the head, neck, and extremities of older adults.1 Dermal leiomyosarcomas originate from arrector pili and are locally aggressive, whereas subcutaneous leiomyosarcomas arise from vascular smooth muscle and metastasize in 30% to 60% of cases.2 Clinically, leiomyosarcomas present as solitary, firm, well-circumscribed nodules with possible ulceration and crusting.3 Histopathology of leiomyosarcoma shows fascicles of atypical spindle cells with blunt-ended nuclei and perinuclear glycogen vacuoles, variable atypia, and mitotic figures (quiz images). Definitive diagnosis is based on positive immunohistochemical staining for desmin and smooth muscle actin.4 Treatment entails complete removal via wide local excision or Mohs micrographic surgery.5
Atypical fibroxanthoma (AFX) is a malignant fibrohistiocytic neoplasm that arises in the dermis and preferentially affects the head and neck in older individuals.3 Atypical fibroxanthoma presents as a nonspecific, pinkred, sometimes ulcerated papule on sun-damaged skin that may clinically resemble a squamous cell carcinoma (SCC) or basal cell carcinoma.6 Histopathology shows pleomorphic spindle cells with hyperchromatic nuclei and abundant cytoplasm mixed with multinucleated giant cells and scattered mitotic figures (Figure 1). Immunohistochemistry is essential for distinguishing AFX from other spindle cell neoplasms. Atypical fibroxanthoma stains positively for vimentin, procollagen-1, CD10, and CD68 but is negative for S-100, human melanoma black 45, Melan-A, desmin, cytokeratin, p40, and p63.6 Treatment includes wide local excision or Mohs micrographic surgery.
Melanoma is an aggressive cancer with the propensity to metastasize. Both desmoplastic and spindle cell variants demonstrate atypical spindled melanocytes on histology, and desmoplasia is seen in the desmoplastic variant (Figure 2). In some cases, evaluation of the epidermis for melanoma in situ may aid in diagnosis.7 Clinical and prognostic features differ between the 2 variants. Desmoplastic melanomas usually present on the head and neck as scarlike nodules with a low rate of nodal involvement, while spindle cell melanomas can occur anywhere on the body, often are amelanotic, and are associated with widespread metastatic disease at the time of presentation.8 SOX10 (SRY-box transcription factor 10) and S-100 may be the only markers that are positive in desmoplastic melanoma.9,10 Treatment depends on the thickness of the lesion.11
Spindle cell SCC is a histologic variant of SCC characterized by spindled epithelial cells. Spindle cell SCC typically presents as an ulcerated or exophytic mass in sun-exposed areas or areas exposed to ionizing radiation, or in immunocompromised individuals. Histopathology shows spindled pleomorphic keratinocytes with elongated nuclei infiltrating the dermis and minimal keratinization (Figure 3).12 Immunohistochemistry is necessary to distinguish spindle cell SCC from other spindle cell tumors such as spindle cell melanoma, AFX, and leiomyosarcoma. Spindle cell SCC is positive for high-molecular-weight cytokeratin, p40, and p63. Mohs micrographic surgery provides the highest cure rate, and radiation therapy may be considered when clear surgical margins cannot be obtained.6
Undifferentiated pleomorphic sarcoma (UPS) (formerly known as malignant fibrous histiocytoma) describes tumors that resemble AFX but are more invasive. They commonly involve the soft tissue with a higher risk for both recurrence and metastasis than AFX.13 Histopathology shows marked cytologic pleomorphism, bizarre cellular forms, atypical mitoses, and ulceration (Figure 4).14 Diagnosis of UPS is by exclusion and is dependent on immunohistochemical studies. In contrast to AFX, UPS is more likely to be positive for LN-2 (CD74).6 Undifferentiated pleomorphic sarcoma has been treated with surgical excision in combination with chemical and radiation therapy, but due to limited data, optimal management is less clear compared to AFX.15 There is a substantial risk for local recurrence and metastasis, and the lungs are the most common sites of distant metastasis.13 In a study of 23 individuals with high-grade UPS, 5-year metastasis-free survival and local recurrence-free survival were 26% and 16%, respectively.10
- Massi D, Franchi A, Alos L, et al. Primary cutaneous leiomyosarcoma: clinicopathological analysis of 36 cases. Histopathology. 2010;56: 251-262. doi:10.1111/j.1365-2559.2009.03471.x
- Ciurea ME, Georgescu CV, Radu CC, et al. Cutaneous leiomyosarcoma—case report [published online June 25, 2014]. J Med Life. 2014;7:270-273.
- Fleury LFF, Sanches JA. Primary cutaneous sarcomas. An Bras Dermatol. 2006;81:207-221. doi:10.1590/s0365-05962006000300002
- Murback NDN, de Castro BC, Takita LC, et al. Cutaneous leiomyosarcoma on the face. An Bras Dermatol. 2018;93:262-264. doi:10.1590 /abd1806-4841.20186715
- Winchester DS, Hocker TL, Brewer JD, et al. Leiomyosarcoma of the skin: clinical, histopathologic, and prognostic factors that influence outcomes. J Am Acad Dermatol. 2014;71:919-925. doi:10.1016/j .jaad.2014.07.020
- Hollmig ST, Sachdev R, Cockerell CJ, et al. Spindle cell neoplasms encountered in dermatologic surgery: a review. Dermatol Surg. 2012;38:825-850. doi:10.1111/j.1524-4725.2012.02296.x
- De Almeida LS, Requena L, Rütten A, et al. Desmoplastic malignant melanoma: a clinicopathologic analysis of 113 cases. Am J Dermatopathol. 2008;30:207-215. doi:10.1097/DAD.0B013E3181716E6B
- Weissinger SE, Keil P, Silvers DN, et al. A diagnostic algorithm to distinguish desmoplastic from spindle cell melanoma. Mod Pathol. 2014;27:524-534. doi:10.1038/modpathol.2013.162
- Ohsie SJ, Sarantopoulos GP, Cochran AJ, et al. Immunohistochemical characteristics of melanoma. J Cutan Pathol. 2008;35:433-444. doi:10.1111/j.1600-0560.2007.00891.x
- Delisca GO, Mesko NW, Alamanda VK, et al. MFH and highgrade undifferentiated pleomorphic sarcoma—what’s in a name? [published online September 12, 2014]. J Surg Oncol. 2015;111:173-177. doi:10.1002/jso.23787
- Baron PL, Nguyen CL. Malignant of melanoma. In: Holzheimer RG, Mannick JA, eds. Surgical Treatment: Evidence-Based and Problem- Oriented. Zuckschwerdt; 2001. https://www.ncbi.nlm.nih.gov/books /NBK6877
- Wernheden E, Trøstrup H, Pedersen Pilt A. Unusual presentation of cutaneous spindle cell squamous cell carcinoma: a case report. Case Rep Dermatol. 2020;12:70-75. doi:10.1159/000507358
- Ramsey JK, Chen JL, Schoenfield L, et al. Undifferentiated pleomorphic sarcoma metastatic to the orbit. Ophthal Plast Reconstr Surg. 2018;34:E193-E195. doi:10.1097/IOP.0000000000001240
- Winchester D, Lehman J, Tello T, et al. Undifferentiated pleomorphic sarcoma: factors predictive of adverse outcomes. J Am Acad Dermatol. 2018;79:853-859. doi:10.1016/j.jaad.2018.05.022
- Soleymani T, Tyler Hollmig S. Conception and management of a poorly understood spectrum of dermatologic neoplasms: atypical fibroxanthoma, pleomorphic dermal sarcoma, and undifferentiated pleomorphic sarcoma. Curr Treat Options Oncol. 2017;18:50. doi:10.1007 /s11864-017-0489-6
The Diagnosis: Leiomyosarcoma
Cutaneous leiomyosarcomas are relatively rare neoplasms that favor the head, neck, and extremities of older adults.1 Dermal leiomyosarcomas originate from arrector pili and are locally aggressive, whereas subcutaneous leiomyosarcomas arise from vascular smooth muscle and metastasize in 30% to 60% of cases.2 Clinically, leiomyosarcomas present as solitary, firm, well-circumscribed nodules with possible ulceration and crusting.3 Histopathology of leiomyosarcoma shows fascicles of atypical spindle cells with blunt-ended nuclei and perinuclear glycogen vacuoles, variable atypia, and mitotic figures (quiz images). Definitive diagnosis is based on positive immunohistochemical staining for desmin and smooth muscle actin.4 Treatment entails complete removal via wide local excision or Mohs micrographic surgery.5
Atypical fibroxanthoma (AFX) is a malignant fibrohistiocytic neoplasm that arises in the dermis and preferentially affects the head and neck in older individuals.3 Atypical fibroxanthoma presents as a nonspecific, pinkred, sometimes ulcerated papule on sun-damaged skin that may clinically resemble a squamous cell carcinoma (SCC) or basal cell carcinoma.6 Histopathology shows pleomorphic spindle cells with hyperchromatic nuclei and abundant cytoplasm mixed with multinucleated giant cells and scattered mitotic figures (Figure 1). Immunohistochemistry is essential for distinguishing AFX from other spindle cell neoplasms. Atypical fibroxanthoma stains positively for vimentin, procollagen-1, CD10, and CD68 but is negative for S-100, human melanoma black 45, Melan-A, desmin, cytokeratin, p40, and p63.6 Treatment includes wide local excision or Mohs micrographic surgery.
Melanoma is an aggressive cancer with the propensity to metastasize. Both desmoplastic and spindle cell variants demonstrate atypical spindled melanocytes on histology, and desmoplasia is seen in the desmoplastic variant (Figure 2). In some cases, evaluation of the epidermis for melanoma in situ may aid in diagnosis.7 Clinical and prognostic features differ between the 2 variants. Desmoplastic melanomas usually present on the head and neck as scarlike nodules with a low rate of nodal involvement, while spindle cell melanomas can occur anywhere on the body, often are amelanotic, and are associated with widespread metastatic disease at the time of presentation.8 SOX10 (SRY-box transcription factor 10) and S-100 may be the only markers that are positive in desmoplastic melanoma.9,10 Treatment depends on the thickness of the lesion.11
Spindle cell SCC is a histologic variant of SCC characterized by spindled epithelial cells. Spindle cell SCC typically presents as an ulcerated or exophytic mass in sun-exposed areas or areas exposed to ionizing radiation, or in immunocompromised individuals. Histopathology shows spindled pleomorphic keratinocytes with elongated nuclei infiltrating the dermis and minimal keratinization (Figure 3).12 Immunohistochemistry is necessary to distinguish spindle cell SCC from other spindle cell tumors such as spindle cell melanoma, AFX, and leiomyosarcoma. Spindle cell SCC is positive for high-molecular-weight cytokeratin, p40, and p63. Mohs micrographic surgery provides the highest cure rate, and radiation therapy may be considered when clear surgical margins cannot be obtained.6
Undifferentiated pleomorphic sarcoma (UPS) (formerly known as malignant fibrous histiocytoma) describes tumors that resemble AFX but are more invasive. They commonly involve the soft tissue with a higher risk for both recurrence and metastasis than AFX.13 Histopathology shows marked cytologic pleomorphism, bizarre cellular forms, atypical mitoses, and ulceration (Figure 4).14 Diagnosis of UPS is by exclusion and is dependent on immunohistochemical studies. In contrast to AFX, UPS is more likely to be positive for LN-2 (CD74).6 Undifferentiated pleomorphic sarcoma has been treated with surgical excision in combination with chemical and radiation therapy, but due to limited data, optimal management is less clear compared to AFX.15 There is a substantial risk for local recurrence and metastasis, and the lungs are the most common sites of distant metastasis.13 In a study of 23 individuals with high-grade UPS, 5-year metastasis-free survival and local recurrence-free survival were 26% and 16%, respectively.10
The Diagnosis: Leiomyosarcoma
Cutaneous leiomyosarcomas are relatively rare neoplasms that favor the head, neck, and extremities of older adults.1 Dermal leiomyosarcomas originate from arrector pili and are locally aggressive, whereas subcutaneous leiomyosarcomas arise from vascular smooth muscle and metastasize in 30% to 60% of cases.2 Clinically, leiomyosarcomas present as solitary, firm, well-circumscribed nodules with possible ulceration and crusting.3 Histopathology of leiomyosarcoma shows fascicles of atypical spindle cells with blunt-ended nuclei and perinuclear glycogen vacuoles, variable atypia, and mitotic figures (quiz images). Definitive diagnosis is based on positive immunohistochemical staining for desmin and smooth muscle actin.4 Treatment entails complete removal via wide local excision or Mohs micrographic surgery.5
Atypical fibroxanthoma (AFX) is a malignant fibrohistiocytic neoplasm that arises in the dermis and preferentially affects the head and neck in older individuals.3 Atypical fibroxanthoma presents as a nonspecific, pinkred, sometimes ulcerated papule on sun-damaged skin that may clinically resemble a squamous cell carcinoma (SCC) or basal cell carcinoma.6 Histopathology shows pleomorphic spindle cells with hyperchromatic nuclei and abundant cytoplasm mixed with multinucleated giant cells and scattered mitotic figures (Figure 1). Immunohistochemistry is essential for distinguishing AFX from other spindle cell neoplasms. Atypical fibroxanthoma stains positively for vimentin, procollagen-1, CD10, and CD68 but is negative for S-100, human melanoma black 45, Melan-A, desmin, cytokeratin, p40, and p63.6 Treatment includes wide local excision or Mohs micrographic surgery.
Melanoma is an aggressive cancer with the propensity to metastasize. Both desmoplastic and spindle cell variants demonstrate atypical spindled melanocytes on histology, and desmoplasia is seen in the desmoplastic variant (Figure 2). In some cases, evaluation of the epidermis for melanoma in situ may aid in diagnosis.7 Clinical and prognostic features differ between the 2 variants. Desmoplastic melanomas usually present on the head and neck as scarlike nodules with a low rate of nodal involvement, while spindle cell melanomas can occur anywhere on the body, often are amelanotic, and are associated with widespread metastatic disease at the time of presentation.8 SOX10 (SRY-box transcription factor 10) and S-100 may be the only markers that are positive in desmoplastic melanoma.9,10 Treatment depends on the thickness of the lesion.11
Spindle cell SCC is a histologic variant of SCC characterized by spindled epithelial cells. Spindle cell SCC typically presents as an ulcerated or exophytic mass in sun-exposed areas or areas exposed to ionizing radiation, or in immunocompromised individuals. Histopathology shows spindled pleomorphic keratinocytes with elongated nuclei infiltrating the dermis and minimal keratinization (Figure 3).12 Immunohistochemistry is necessary to distinguish spindle cell SCC from other spindle cell tumors such as spindle cell melanoma, AFX, and leiomyosarcoma. Spindle cell SCC is positive for high-molecular-weight cytokeratin, p40, and p63. Mohs micrographic surgery provides the highest cure rate, and radiation therapy may be considered when clear surgical margins cannot be obtained.6
Undifferentiated pleomorphic sarcoma (UPS) (formerly known as malignant fibrous histiocytoma) describes tumors that resemble AFX but are more invasive. They commonly involve the soft tissue with a higher risk for both recurrence and metastasis than AFX.13 Histopathology shows marked cytologic pleomorphism, bizarre cellular forms, atypical mitoses, and ulceration (Figure 4).14 Diagnosis of UPS is by exclusion and is dependent on immunohistochemical studies. In contrast to AFX, UPS is more likely to be positive for LN-2 (CD74).6 Undifferentiated pleomorphic sarcoma has been treated with surgical excision in combination with chemical and radiation therapy, but due to limited data, optimal management is less clear compared to AFX.15 There is a substantial risk for local recurrence and metastasis, and the lungs are the most common sites of distant metastasis.13 In a study of 23 individuals with high-grade UPS, 5-year metastasis-free survival and local recurrence-free survival were 26% and 16%, respectively.10
- Massi D, Franchi A, Alos L, et al. Primary cutaneous leiomyosarcoma: clinicopathological analysis of 36 cases. Histopathology. 2010;56: 251-262. doi:10.1111/j.1365-2559.2009.03471.x
- Ciurea ME, Georgescu CV, Radu CC, et al. Cutaneous leiomyosarcoma—case report [published online June 25, 2014]. J Med Life. 2014;7:270-273.
- Fleury LFF, Sanches JA. Primary cutaneous sarcomas. An Bras Dermatol. 2006;81:207-221. doi:10.1590/s0365-05962006000300002
- Murback NDN, de Castro BC, Takita LC, et al. Cutaneous leiomyosarcoma on the face. An Bras Dermatol. 2018;93:262-264. doi:10.1590 /abd1806-4841.20186715
- Winchester DS, Hocker TL, Brewer JD, et al. Leiomyosarcoma of the skin: clinical, histopathologic, and prognostic factors that influence outcomes. J Am Acad Dermatol. 2014;71:919-925. doi:10.1016/j .jaad.2014.07.020
- Hollmig ST, Sachdev R, Cockerell CJ, et al. Spindle cell neoplasms encountered in dermatologic surgery: a review. Dermatol Surg. 2012;38:825-850. doi:10.1111/j.1524-4725.2012.02296.x
- De Almeida LS, Requena L, Rütten A, et al. Desmoplastic malignant melanoma: a clinicopathologic analysis of 113 cases. Am J Dermatopathol. 2008;30:207-215. doi:10.1097/DAD.0B013E3181716E6B
- Weissinger SE, Keil P, Silvers DN, et al. A diagnostic algorithm to distinguish desmoplastic from spindle cell melanoma. Mod Pathol. 2014;27:524-534. doi:10.1038/modpathol.2013.162
- Ohsie SJ, Sarantopoulos GP, Cochran AJ, et al. Immunohistochemical characteristics of melanoma. J Cutan Pathol. 2008;35:433-444. doi:10.1111/j.1600-0560.2007.00891.x
- Delisca GO, Mesko NW, Alamanda VK, et al. MFH and highgrade undifferentiated pleomorphic sarcoma—what’s in a name? [published online September 12, 2014]. J Surg Oncol. 2015;111:173-177. doi:10.1002/jso.23787
- Baron PL, Nguyen CL. Malignant of melanoma. In: Holzheimer RG, Mannick JA, eds. Surgical Treatment: Evidence-Based and Problem- Oriented. Zuckschwerdt; 2001. https://www.ncbi.nlm.nih.gov/books /NBK6877
- Wernheden E, Trøstrup H, Pedersen Pilt A. Unusual presentation of cutaneous spindle cell squamous cell carcinoma: a case report. Case Rep Dermatol. 2020;12:70-75. doi:10.1159/000507358
- Ramsey JK, Chen JL, Schoenfield L, et al. Undifferentiated pleomorphic sarcoma metastatic to the orbit. Ophthal Plast Reconstr Surg. 2018;34:E193-E195. doi:10.1097/IOP.0000000000001240
- Winchester D, Lehman J, Tello T, et al. Undifferentiated pleomorphic sarcoma: factors predictive of adverse outcomes. J Am Acad Dermatol. 2018;79:853-859. doi:10.1016/j.jaad.2018.05.022
- Soleymani T, Tyler Hollmig S. Conception and management of a poorly understood spectrum of dermatologic neoplasms: atypical fibroxanthoma, pleomorphic dermal sarcoma, and undifferentiated pleomorphic sarcoma. Curr Treat Options Oncol. 2017;18:50. doi:10.1007 /s11864-017-0489-6
- Massi D, Franchi A, Alos L, et al. Primary cutaneous leiomyosarcoma: clinicopathological analysis of 36 cases. Histopathology. 2010;56: 251-262. doi:10.1111/j.1365-2559.2009.03471.x
- Ciurea ME, Georgescu CV, Radu CC, et al. Cutaneous leiomyosarcoma—case report [published online June 25, 2014]. J Med Life. 2014;7:270-273.
- Fleury LFF, Sanches JA. Primary cutaneous sarcomas. An Bras Dermatol. 2006;81:207-221. doi:10.1590/s0365-05962006000300002
- Murback NDN, de Castro BC, Takita LC, et al. Cutaneous leiomyosarcoma on the face. An Bras Dermatol. 2018;93:262-264. doi:10.1590 /abd1806-4841.20186715
- Winchester DS, Hocker TL, Brewer JD, et al. Leiomyosarcoma of the skin: clinical, histopathologic, and prognostic factors that influence outcomes. J Am Acad Dermatol. 2014;71:919-925. doi:10.1016/j .jaad.2014.07.020
- Hollmig ST, Sachdev R, Cockerell CJ, et al. Spindle cell neoplasms encountered in dermatologic surgery: a review. Dermatol Surg. 2012;38:825-850. doi:10.1111/j.1524-4725.2012.02296.x
- De Almeida LS, Requena L, Rütten A, et al. Desmoplastic malignant melanoma: a clinicopathologic analysis of 113 cases. Am J Dermatopathol. 2008;30:207-215. doi:10.1097/DAD.0B013E3181716E6B
- Weissinger SE, Keil P, Silvers DN, et al. A diagnostic algorithm to distinguish desmoplastic from spindle cell melanoma. Mod Pathol. 2014;27:524-534. doi:10.1038/modpathol.2013.162
- Ohsie SJ, Sarantopoulos GP, Cochran AJ, et al. Immunohistochemical characteristics of melanoma. J Cutan Pathol. 2008;35:433-444. doi:10.1111/j.1600-0560.2007.00891.x
- Delisca GO, Mesko NW, Alamanda VK, et al. MFH and highgrade undifferentiated pleomorphic sarcoma—what’s in a name? [published online September 12, 2014]. J Surg Oncol. 2015;111:173-177. doi:10.1002/jso.23787
- Baron PL, Nguyen CL. Malignant of melanoma. In: Holzheimer RG, Mannick JA, eds. Surgical Treatment: Evidence-Based and Problem- Oriented. Zuckschwerdt; 2001. https://www.ncbi.nlm.nih.gov/books /NBK6877
- Wernheden E, Trøstrup H, Pedersen Pilt A. Unusual presentation of cutaneous spindle cell squamous cell carcinoma: a case report. Case Rep Dermatol. 2020;12:70-75. doi:10.1159/000507358
- Ramsey JK, Chen JL, Schoenfield L, et al. Undifferentiated pleomorphic sarcoma metastatic to the orbit. Ophthal Plast Reconstr Surg. 2018;34:E193-E195. doi:10.1097/IOP.0000000000001240
- Winchester D, Lehman J, Tello T, et al. Undifferentiated pleomorphic sarcoma: factors predictive of adverse outcomes. J Am Acad Dermatol. 2018;79:853-859. doi:10.1016/j.jaad.2018.05.022
- Soleymani T, Tyler Hollmig S. Conception and management of a poorly understood spectrum of dermatologic neoplasms: atypical fibroxanthoma, pleomorphic dermal sarcoma, and undifferentiated pleomorphic sarcoma. Curr Treat Options Oncol. 2017;18:50. doi:10.1007 /s11864-017-0489-6
A 62-year-old man presented with a firm, exophytic, 2.8×1.5-cm tumor on the left shin of 6 to 7 years’ duration. An excisional biopsy was obtained for histopathologic evaluation.
Blood test for cancer available, but is it ready for prime time?
The Galleri blood test is being now offered by a number of United States health networks.
The company marketing the test, GRAIL, has established partnerships with the U.S. Department of Veterans Affairs, Mercy Health, Ochsner Health, Intermountain Healthcare, Community Health Network, Knight Cancer Institute at Oregon Health & Science University, Premier, and Cleveland Clinic, among others.
Cleveland Clinic’s Eric Klein, MD, emeritus chair of the Glickman Urological Kidney Institute, is enthusiastic about the test, describing it as a “game-changer” and emphasizing that it can detect many different cancers and at a very early stage.
“It completely changes the way we think about screening for cancer,” commented Jeff Venstrom, MD, chief medical officer at GRAIL. He joined the company because “there are not many things in life where you can be part of a disruptive paradigm and disruptive technology, and this really is disruptive,” he said in an interview.
‘The devil is in the details’
But there is some concern among clinicians that widespread clinical use of the test may be premature.
Having a blood test for multiple cancers is a “very good idea, and the scientific basis for this platform is sound,” commented Timothy R. Rebbeck, PhD, professor of cancer prevention, Harvard T.H. Chan School of Public Health, and Division of Population Sciences, Dana-Farber Cancer Institute, both in Boston.
“But the devil is in the details to ensure the test can accurately detect very early cancers and there is a pathway for subsequent workup (diagnosis, monitoring, treatment, etc.),” Dr. Rebbeck told this news organization.
Galleri is offering the test to individuals who are older than 50 and have a family history of cancer or those who are high risk for cancer or immunocompromised. They suggest that interested individuals get in touch with their health care professional, who then needs to register with GRAIL and order the test.
As well as needing a prescription, interested individuals will have to pay for it out of pocket, around $950. The test is not covered by medical insurance and is not approved by the U.S. Food and Drug Administration.
Falls into primary care setting
Dr. Rebbeck commented that Galleri is a screening test for individuals who don’t have cancer, so the test is intended to fall into the primary care setting. But he warned that “clinical pathways are not yet in place (but are being developed) so that primary care providers can effectively use them.”
The test uses next-generation sequencing to analyze the arrangement of methyl groups on circulating tumor (or cell-free) DNA in a blood sample.
The methylation turns genes on or off, explains Cleveland Clinic’s Dr. Klein in his post. “It’s like fingerprints and how fingerprints tell the difference between two people,” he wrote. “The methylation patterns are fingerprints that are characteristic of each kind of cancer. They look one way for lung cancer and different for colon cancer.”
The test returns one of two possible results: either “positive, cancer signal detected” or “negative, no cancer signal detected.”
According to the company, when a cancer signal is detected, the Galleri test predicts the cancer signal origin “with high accuracy, to help guide the next steps to diagnosis.”
However, one problem for clinical practice is all the follow-up tests an individual may undergo if their test comes back positive, said Sameek Roychowdhury, MD, PhD, an oncologist with Ohio State University Comprehensive Cancer Center, Columbus.
“Not everybody will have an actual cancer, but they may undergo many tests, with a lot of stress and cost and still not find anything. I can tell you every time someone undergoes a test looking for cancer, that is not an easy day,” Dr. Roychowdhury said in an interview.
In a large-scale validation study, the Galleri test had a specificity of 99.5% (false-positive rate of 0.5%), meaning in roughly 200 people tested without cancer, only one person received a false-positive result (that is, “cancer signal detected” when cancer is not present).
The overall sensitivity of the test for any stage of cancer was 51.5%, although it was higher for later-stage cancers (77% for stage III and 90.1% for stage IV) and lower for early-stage cancers (16.8% for stage I and 40.4% for stage II).
Exacerbate health disparities?
In Dr. Rebbeck’s view, the characteristics of the test are still “relatively poor for detecting very early cancers, so it will need additional tweaking before it really achieves the goal of multi-cancer EARLY detection,” he said.
Dr. Venstrom acknowledges that the test is “not perfect yet” and says the company will continue to update and improve its performance. “We have some new data coming out in September,” he said.
Clinical data are being accumulated in the United Kingdom, where the Galleri test is being investigated in a large trial run by the National Health Service (NHS). The company recently announced that the enrollment of 140,000 healthy cancer-free volunteers aged 50-77 into this trial has now been completed and claimed this the largest-ever study of a multi-cancer early detection test.
Dr. Roychowdhury said he would encourage anyone interested in the test to join a clinical trial.
Another expert approached for comment last year, when GRAIL first started marketing the test, was in agreement. This test should be viewed as one that is still under clinical investigation, commented William Grady, MD, a member of the clinical research division and public health sciences division at the Fred Hutchinson Cancer Research Center, Seattle.
“The Galleri test is still unproven in the clinical care setting and ... I am concerned that many of the results will be false-positives and will cause many unnecessary follow-up tests and imaging studies as well as anxiety in the people getting the test done,” Dr. Grady said.
Dr. Rebbeck said another issue that needs to be addressed is whether all populations will have access to and benefit from these types of blood tests to screen for cancer, given that they are expensive.
“There is a great danger – as we have seen with many other technological innovations – that the wealthy and connected benefit, but the majority of the population, and particularly those who are underserved, do not,” Dr. Rebbeck said.
“As a result, health disparities are created or exacerbated. This is something that needs to be addressed so that the future use of these tests will provide equitable benefits,” he added.
Dr. Rebbeck and Dr. Roychowdhury have reported no relevant financial relationships. Dr. Venstrom is an employee of GRAIL.
A version of this article first appeared on Medscape.com.
The Galleri blood test is being now offered by a number of United States health networks.
The company marketing the test, GRAIL, has established partnerships with the U.S. Department of Veterans Affairs, Mercy Health, Ochsner Health, Intermountain Healthcare, Community Health Network, Knight Cancer Institute at Oregon Health & Science University, Premier, and Cleveland Clinic, among others.
Cleveland Clinic’s Eric Klein, MD, emeritus chair of the Glickman Urological Kidney Institute, is enthusiastic about the test, describing it as a “game-changer” and emphasizing that it can detect many different cancers and at a very early stage.
“It completely changes the way we think about screening for cancer,” commented Jeff Venstrom, MD, chief medical officer at GRAIL. He joined the company because “there are not many things in life where you can be part of a disruptive paradigm and disruptive technology, and this really is disruptive,” he said in an interview.
‘The devil is in the details’
But there is some concern among clinicians that widespread clinical use of the test may be premature.
Having a blood test for multiple cancers is a “very good idea, and the scientific basis for this platform is sound,” commented Timothy R. Rebbeck, PhD, professor of cancer prevention, Harvard T.H. Chan School of Public Health, and Division of Population Sciences, Dana-Farber Cancer Institute, both in Boston.
“But the devil is in the details to ensure the test can accurately detect very early cancers and there is a pathway for subsequent workup (diagnosis, monitoring, treatment, etc.),” Dr. Rebbeck told this news organization.
Galleri is offering the test to individuals who are older than 50 and have a family history of cancer or those who are high risk for cancer or immunocompromised. They suggest that interested individuals get in touch with their health care professional, who then needs to register with GRAIL and order the test.
As well as needing a prescription, interested individuals will have to pay for it out of pocket, around $950. The test is not covered by medical insurance and is not approved by the U.S. Food and Drug Administration.
Falls into primary care setting
Dr. Rebbeck commented that Galleri is a screening test for individuals who don’t have cancer, so the test is intended to fall into the primary care setting. But he warned that “clinical pathways are not yet in place (but are being developed) so that primary care providers can effectively use them.”
The test uses next-generation sequencing to analyze the arrangement of methyl groups on circulating tumor (or cell-free) DNA in a blood sample.
The methylation turns genes on or off, explains Cleveland Clinic’s Dr. Klein in his post. “It’s like fingerprints and how fingerprints tell the difference between two people,” he wrote. “The methylation patterns are fingerprints that are characteristic of each kind of cancer. They look one way for lung cancer and different for colon cancer.”
The test returns one of two possible results: either “positive, cancer signal detected” or “negative, no cancer signal detected.”
According to the company, when a cancer signal is detected, the Galleri test predicts the cancer signal origin “with high accuracy, to help guide the next steps to diagnosis.”
However, one problem for clinical practice is all the follow-up tests an individual may undergo if their test comes back positive, said Sameek Roychowdhury, MD, PhD, an oncologist with Ohio State University Comprehensive Cancer Center, Columbus.
“Not everybody will have an actual cancer, but they may undergo many tests, with a lot of stress and cost and still not find anything. I can tell you every time someone undergoes a test looking for cancer, that is not an easy day,” Dr. Roychowdhury said in an interview.
In a large-scale validation study, the Galleri test had a specificity of 99.5% (false-positive rate of 0.5%), meaning in roughly 200 people tested without cancer, only one person received a false-positive result (that is, “cancer signal detected” when cancer is not present).
The overall sensitivity of the test for any stage of cancer was 51.5%, although it was higher for later-stage cancers (77% for stage III and 90.1% for stage IV) and lower for early-stage cancers (16.8% for stage I and 40.4% for stage II).
Exacerbate health disparities?
In Dr. Rebbeck’s view, the characteristics of the test are still “relatively poor for detecting very early cancers, so it will need additional tweaking before it really achieves the goal of multi-cancer EARLY detection,” he said.
Dr. Venstrom acknowledges that the test is “not perfect yet” and says the company will continue to update and improve its performance. “We have some new data coming out in September,” he said.
Clinical data are being accumulated in the United Kingdom, where the Galleri test is being investigated in a large trial run by the National Health Service (NHS). The company recently announced that the enrollment of 140,000 healthy cancer-free volunteers aged 50-77 into this trial has now been completed and claimed this the largest-ever study of a multi-cancer early detection test.
Dr. Roychowdhury said he would encourage anyone interested in the test to join a clinical trial.
Another expert approached for comment last year, when GRAIL first started marketing the test, was in agreement. This test should be viewed as one that is still under clinical investigation, commented William Grady, MD, a member of the clinical research division and public health sciences division at the Fred Hutchinson Cancer Research Center, Seattle.
“The Galleri test is still unproven in the clinical care setting and ... I am concerned that many of the results will be false-positives and will cause many unnecessary follow-up tests and imaging studies as well as anxiety in the people getting the test done,” Dr. Grady said.
Dr. Rebbeck said another issue that needs to be addressed is whether all populations will have access to and benefit from these types of blood tests to screen for cancer, given that they are expensive.
“There is a great danger – as we have seen with many other technological innovations – that the wealthy and connected benefit, but the majority of the population, and particularly those who are underserved, do not,” Dr. Rebbeck said.
“As a result, health disparities are created or exacerbated. This is something that needs to be addressed so that the future use of these tests will provide equitable benefits,” he added.
Dr. Rebbeck and Dr. Roychowdhury have reported no relevant financial relationships. Dr. Venstrom is an employee of GRAIL.
A version of this article first appeared on Medscape.com.
The Galleri blood test is being now offered by a number of United States health networks.
The company marketing the test, GRAIL, has established partnerships with the U.S. Department of Veterans Affairs, Mercy Health, Ochsner Health, Intermountain Healthcare, Community Health Network, Knight Cancer Institute at Oregon Health & Science University, Premier, and Cleveland Clinic, among others.
Cleveland Clinic’s Eric Klein, MD, emeritus chair of the Glickman Urological Kidney Institute, is enthusiastic about the test, describing it as a “game-changer” and emphasizing that it can detect many different cancers and at a very early stage.
“It completely changes the way we think about screening for cancer,” commented Jeff Venstrom, MD, chief medical officer at GRAIL. He joined the company because “there are not many things in life where you can be part of a disruptive paradigm and disruptive technology, and this really is disruptive,” he said in an interview.
‘The devil is in the details’
But there is some concern among clinicians that widespread clinical use of the test may be premature.
Having a blood test for multiple cancers is a “very good idea, and the scientific basis for this platform is sound,” commented Timothy R. Rebbeck, PhD, professor of cancer prevention, Harvard T.H. Chan School of Public Health, and Division of Population Sciences, Dana-Farber Cancer Institute, both in Boston.
“But the devil is in the details to ensure the test can accurately detect very early cancers and there is a pathway for subsequent workup (diagnosis, monitoring, treatment, etc.),” Dr. Rebbeck told this news organization.
Galleri is offering the test to individuals who are older than 50 and have a family history of cancer or those who are high risk for cancer or immunocompromised. They suggest that interested individuals get in touch with their health care professional, who then needs to register with GRAIL and order the test.
As well as needing a prescription, interested individuals will have to pay for it out of pocket, around $950. The test is not covered by medical insurance and is not approved by the U.S. Food and Drug Administration.
Falls into primary care setting
Dr. Rebbeck commented that Galleri is a screening test for individuals who don’t have cancer, so the test is intended to fall into the primary care setting. But he warned that “clinical pathways are not yet in place (but are being developed) so that primary care providers can effectively use them.”
The test uses next-generation sequencing to analyze the arrangement of methyl groups on circulating tumor (or cell-free) DNA in a blood sample.
The methylation turns genes on or off, explains Cleveland Clinic’s Dr. Klein in his post. “It’s like fingerprints and how fingerprints tell the difference between two people,” he wrote. “The methylation patterns are fingerprints that are characteristic of each kind of cancer. They look one way for lung cancer and different for colon cancer.”
The test returns one of two possible results: either “positive, cancer signal detected” or “negative, no cancer signal detected.”
According to the company, when a cancer signal is detected, the Galleri test predicts the cancer signal origin “with high accuracy, to help guide the next steps to diagnosis.”
However, one problem for clinical practice is all the follow-up tests an individual may undergo if their test comes back positive, said Sameek Roychowdhury, MD, PhD, an oncologist with Ohio State University Comprehensive Cancer Center, Columbus.
“Not everybody will have an actual cancer, but they may undergo many tests, with a lot of stress and cost and still not find anything. I can tell you every time someone undergoes a test looking for cancer, that is not an easy day,” Dr. Roychowdhury said in an interview.
In a large-scale validation study, the Galleri test had a specificity of 99.5% (false-positive rate of 0.5%), meaning in roughly 200 people tested without cancer, only one person received a false-positive result (that is, “cancer signal detected” when cancer is not present).
The overall sensitivity of the test for any stage of cancer was 51.5%, although it was higher for later-stage cancers (77% for stage III and 90.1% for stage IV) and lower for early-stage cancers (16.8% for stage I and 40.4% for stage II).
Exacerbate health disparities?
In Dr. Rebbeck’s view, the characteristics of the test are still “relatively poor for detecting very early cancers, so it will need additional tweaking before it really achieves the goal of multi-cancer EARLY detection,” he said.
Dr. Venstrom acknowledges that the test is “not perfect yet” and says the company will continue to update and improve its performance. “We have some new data coming out in September,” he said.
Clinical data are being accumulated in the United Kingdom, where the Galleri test is being investigated in a large trial run by the National Health Service (NHS). The company recently announced that the enrollment of 140,000 healthy cancer-free volunteers aged 50-77 into this trial has now been completed and claimed this the largest-ever study of a multi-cancer early detection test.
Dr. Roychowdhury said he would encourage anyone interested in the test to join a clinical trial.
Another expert approached for comment last year, when GRAIL first started marketing the test, was in agreement. This test should be viewed as one that is still under clinical investigation, commented William Grady, MD, a member of the clinical research division and public health sciences division at the Fred Hutchinson Cancer Research Center, Seattle.
“The Galleri test is still unproven in the clinical care setting and ... I am concerned that many of the results will be false-positives and will cause many unnecessary follow-up tests and imaging studies as well as anxiety in the people getting the test done,” Dr. Grady said.
Dr. Rebbeck said another issue that needs to be addressed is whether all populations will have access to and benefit from these types of blood tests to screen for cancer, given that they are expensive.
“There is a great danger – as we have seen with many other technological innovations – that the wealthy and connected benefit, but the majority of the population, and particularly those who are underserved, do not,” Dr. Rebbeck said.
“As a result, health disparities are created or exacerbated. This is something that needs to be addressed so that the future use of these tests will provide equitable benefits,” he added.
Dr. Rebbeck and Dr. Roychowdhury have reported no relevant financial relationships. Dr. Venstrom is an employee of GRAIL.
A version of this article first appeared on Medscape.com.
FDA acts against sales of unapproved mole and skin tag products on Amazon, other sites
press release issued on Aug. 9.
In addition to Amazon.com, the other two companies are Ariella Naturals, and Justified Laboratories.
Currently, no over-the-counter products are FDA-approved for the at-home removal of moles and skin tags, and use of unapproved products could be dangerous to consumers, according to the statement. These products may be sold as ointments, gels, sticks, or liquids, and may contain high concentrations of salicylic acid or other harmful ingredients. Introducing unapproved products in to interstate commerce violates the Federal Food, Drug, and Cosmetic Act.
Two products sold on Amazon are the “Deisana Skin Tag Remover, Mole Remover and Repair Gel Set” and “Skincell Mole Skin Tag Corrector Serum,” according to the letter sent to Amazon.
The warning letters alert the three companies that they have 15 days from receipt to address any violations. However, warning letters are not a final FDA action, according to the statement.
“The agency’s rigorous surveillance works to identify threats to public health and stop these products from reaching our communities,” Donald D. Ashley, JD, director of the Office of Compliance in the FDA’s Center for Drug Evaluation and Research, said in the press release. “This includes where online retailers like Amazon are involved in the interstate sale of unapproved drug products. We will continue to work diligently to ensure that online retailers do not sell products that violate federal law,” he added.
The statement emphasized that moles should be evaluated by a health care professional, as attempts at self-diagnosis and at-home treatment could lead to a delayed cancer diagnosis, and potentially to cancer progression.
Products marketed to consumers for at-home removal of moles, skin tags, and other skin lesions could cause injuries, infections, and scarring, according to a related consumer update first posted by the FDA in June, which was updated after the warning letters were sent out.
Consumers and health care professionals are encouraged to report any adverse events related to mole removal or skin tag removal products to the agency’s MedWatch Adverse Event Reporting program.
The FDA also offers an online guide, BeSafeRx, with advice for consumers about potential risks of using online pharmacies and how to do so safely.
press release issued on Aug. 9.
In addition to Amazon.com, the other two companies are Ariella Naturals, and Justified Laboratories.
Currently, no over-the-counter products are FDA-approved for the at-home removal of moles and skin tags, and use of unapproved products could be dangerous to consumers, according to the statement. These products may be sold as ointments, gels, sticks, or liquids, and may contain high concentrations of salicylic acid or other harmful ingredients. Introducing unapproved products in to interstate commerce violates the Federal Food, Drug, and Cosmetic Act.
Two products sold on Amazon are the “Deisana Skin Tag Remover, Mole Remover and Repair Gel Set” and “Skincell Mole Skin Tag Corrector Serum,” according to the letter sent to Amazon.
The warning letters alert the three companies that they have 15 days from receipt to address any violations. However, warning letters are not a final FDA action, according to the statement.
“The agency’s rigorous surveillance works to identify threats to public health and stop these products from reaching our communities,” Donald D. Ashley, JD, director of the Office of Compliance in the FDA’s Center for Drug Evaluation and Research, said in the press release. “This includes where online retailers like Amazon are involved in the interstate sale of unapproved drug products. We will continue to work diligently to ensure that online retailers do not sell products that violate federal law,” he added.
The statement emphasized that moles should be evaluated by a health care professional, as attempts at self-diagnosis and at-home treatment could lead to a delayed cancer diagnosis, and potentially to cancer progression.
Products marketed to consumers for at-home removal of moles, skin tags, and other skin lesions could cause injuries, infections, and scarring, according to a related consumer update first posted by the FDA in June, which was updated after the warning letters were sent out.
Consumers and health care professionals are encouraged to report any adverse events related to mole removal or skin tag removal products to the agency’s MedWatch Adverse Event Reporting program.
The FDA also offers an online guide, BeSafeRx, with advice for consumers about potential risks of using online pharmacies and how to do so safely.
press release issued on Aug. 9.
In addition to Amazon.com, the other two companies are Ariella Naturals, and Justified Laboratories.
Currently, no over-the-counter products are FDA-approved for the at-home removal of moles and skin tags, and use of unapproved products could be dangerous to consumers, according to the statement. These products may be sold as ointments, gels, sticks, or liquids, and may contain high concentrations of salicylic acid or other harmful ingredients. Introducing unapproved products in to interstate commerce violates the Federal Food, Drug, and Cosmetic Act.
Two products sold on Amazon are the “Deisana Skin Tag Remover, Mole Remover and Repair Gel Set” and “Skincell Mole Skin Tag Corrector Serum,” according to the letter sent to Amazon.
The warning letters alert the three companies that they have 15 days from receipt to address any violations. However, warning letters are not a final FDA action, according to the statement.
“The agency’s rigorous surveillance works to identify threats to public health and stop these products from reaching our communities,” Donald D. Ashley, JD, director of the Office of Compliance in the FDA’s Center for Drug Evaluation and Research, said in the press release. “This includes where online retailers like Amazon are involved in the interstate sale of unapproved drug products. We will continue to work diligently to ensure that online retailers do not sell products that violate federal law,” he added.
The statement emphasized that moles should be evaluated by a health care professional, as attempts at self-diagnosis and at-home treatment could lead to a delayed cancer diagnosis, and potentially to cancer progression.
Products marketed to consumers for at-home removal of moles, skin tags, and other skin lesions could cause injuries, infections, and scarring, according to a related consumer update first posted by the FDA in June, which was updated after the warning letters were sent out.
Consumers and health care professionals are encouraged to report any adverse events related to mole removal or skin tag removal products to the agency’s MedWatch Adverse Event Reporting program.
The FDA also offers an online guide, BeSafeRx, with advice for consumers about potential risks of using online pharmacies and how to do so safely.
Unique Treatment for Alopecia Areata Combining Epinephrine With an Intralesional Steroid
Alopecia areata (AA) is an autoimmune disorder characterized by transient hair loss with preservation of the hair follicle (HF). The lifetime incidence risk of AA is approximately 2%,1 with a mean age of onset of 25 to 36 years and with no clinically relevant significant differences between sex or ethnicity.2 Most commonly, it presents as round, well-demarcated patches of alopecia on the scalp and spontaneously resolves in nearly 30% of patients. However, severe disease is associated with younger age of presentation and can progress to a total loss of scalp or body hair—referred to as alopecia totalis and alopecia universalis, respectively—thus severely impacting quality of life.3,4
First-line treatment options for AA include potent topical steroids5,6 and intralesional (IL) steroids, most commonly IL triamcinolone acetonide (ILTA). Intralesional steroids have been found to be more effective than topicals in stimulating hair growth at the injection site.7,8 A recent systemic therapy—the Janus kinase inhibitor baricitinib—was approved by the US Food and Drug Administration for AA.9 Other systemic therapies such as oral corticosteroids have been studied in small trials with promising results.10 However, the risks of systemic therapies may outweigh the benefits.9,10
Another less common topical therapy is contact immunotherapy, which involves topical application of an unlicensed non–pharmaceutical-grade agent to areas affected with AA. It is reported to have a wide range of response rates (29%–87%).11
We report 2 cases of extensive AA that were treated with a novel combination regimen— 2.5 mg/mL of ILTA diluted with lidocaine 1% and epinephrine 1:100,000 in place of normal saline (NS)— which is a modification to an already widely used first-line treatment.
Case Reports
Patient 1—An 11-year-old girl presented with nonscarring alopecia of the vertex and occipital scalp. Three years prior she was treated with topical and IL corticosteroids by a different provider. Physical examination revealed almost complete alopecia involving the bottom two-thirds of the occipital scalp as well as the medial eyebrows (Figures 1A and 1B). Over the span of 1 year she was treated with betamethasone dipropionate cream 0.05% and several rounds of ILTA 2.5 mg/mL buffered with NS, with minimal improvement. A year after the initial presentation, the decision was made to initiate monthly injections of ILTA 2.5 mg/mL buffered with 1% lidocaine and epinephrine 1:100,000. Some hair regrowth of the occipital scalp was noted by 3 months, with near-complete regrowth of the scalp hair and eyebrows by 7 months and 5 months, respectively (Figures 1C and 1D). During this period, the patient continued to develop new areas of alopecia of the scalp and eyebrows, which also were injected with this combination. In total, the patient received 8 rounds of IL injections 4 to 6 weeks apart in the scalp and 6 rounds in the eyebrows. The treated areas showed resolution over a follow-up period of 14 months, though there was recurrence at the right medial eyebrow at 5 months. No localized skin atrophy or other adverse effects were noted.
Patient 2—A 34-year-old woman who was otherwise healthy presented with previously untreated AA involving the scalp of 2 months’ duration. Physical examination revealed the following areas of nonscarring alopecia: a 10×10-cm area of the right occipital scalp with some regrowth; a 10×14-cm area of the left parieto-occipital scalp; and a 1-cm area posterior to the vertex (Figure 2A). Given the extensive involvement, the decision was made to initiate ILTA 2.5 mg/mL buffered with 1% lidocaine and epinephrine 1:100,000 once monthly. Appreciable hair regrowth was noted within 1 month, mostly on the parietal scalp. Substantial improvement was noted after 3 months in all affected areas of the hair-bearing scalp, with near-complete regrowth on the left occipital scalp and greater than 50% regrowth on the right occipital scalp (Figure 2B). No adverse effects were noted. She currently has no alopecia.
Comment
Alopecia Pathogenesis—The most widely adopted theory of AA etiology implicates an aberrant immune response. The HF, which is a dynamic “mini-organ” with its own immune and hormonal microenvironment, is considered an “immune-privileged site”—meaning it is less exposed to immune responses than most other body areas. It is hypothesized that AA results from a breakdown in this immune privilege, with the subsequent attack on the peribulbar part of the follicle by CD8+ T lymphocytes. This lymphocytic infiltrate induces apoptosis in the HF keratinocytes, resulting in inhibition of hair shaft production.12 Other theories suggest a link to the sympathetic-adrenal-medullary system and hypothalamic-pituitary-adrenal axis.13
Therapies for Alopecia—Topical and IL corticosteroids are the first-line therapies for localized AA in patients with less than 50% scalp involvement. Triamcinolone acetonide generally is the IL steroid of choice because it is widely available and less atrophogenic than other steroids. Unlike topicals, ILTA bypasses the epidermis when injected, achieving direct access to the HF.14
High-quality controlled studies regarding the use of ILTA in AA are scarce. A meta-analysis concluded that 5 mg/mL and 10 mg/mL of ILTA diluted in NS were equally effective (80.9% [P<.05] vs 76.4% [P<.005], respectively). Concentrations of less than 5 mg/mL of ILTA resulted in lower rates of hair regrowth (62.3%; P=.04).15 The role of diluents other than NS has not been studied.
Benefits of Epinephrine in ILTA Therapy—The role of epinephrine 1:100,000 is to decrease the rate of clearance of triamcinolone acetonide from the HF, allowing for a better therapeutic effect. Laser Doppler blood flowmeter studies have shown that epinephrine 1:100,000 injected in the scalp causes vasoconstriction, thereby decreasing the blood flow rate of clearance of other substances in the same solution.16 Also, a more gradual systemic absorption is achieved, decreasing systemic side effects such as osteoporosis.17
Another potential benefit of epinephrine has been suggested in animal studies that demonstrate the important role of the sympathetic nervous system in HF growth. In a mouse study, chemical sympathectomy led to diminished norepinephrine levels in the skin, accompanied by a decreased keratinocyte proliferation and hair growth. Conversely, norepinephrine was found to promote HF growth in an organotypic skin culture model.18 Topically applied isoproterenol, a panadrenergic receptor agonist, accelerated HF growth in an organotypic skin culture. It also has been shown that external light and temperature changes stimulate hair growth via the sympathetic nervous system, promoting anagen HF growth in cultured skin explants, further linking HF activity with sympathetic nerve activity.19
In our experience, cases of AA that at first failed ILTA 5 mg/mL in NS have been successfully treated with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000. One such case was alopecia totalis, though we do not have high-quality photographs to present for this report. The 2 cases presented here are the ones with the best photographs to demonstrate our outcomes. Both were treated with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000 administered using a 0.5-in long 30-gauge needle, with 0.05 to 0.1 mL per injection approximately 0.51-cm apart. The treatment intervals were 4 weeks, with a maximal dose of 20 mg per session. In addition to the 2 cases reported here, the Table includes 2 other patients in our practice who were successfully treated with this novel regimen.
Prior to adopting this combination regimen, our standard therapy for AA was 5 mg/mL ILTA buffered with NS. Instead of NS, we now use the widely available 1% lidocaine with epinephrine 1:100,000 and dilute the ILTA to 2.5 mg/mL. We postulate that epinephrine 1:100,000 enhances therapeutic efficacy via local vasoconstriction, thus keeping the ILTA in situ longer than NS. This effect allows for a lower concentration of ILTA (2.5 mg/mL) to be effective. Furthermore, epinephrine 1:100,000 may have an independent effect, as suggested in mouse studies.18
Our first case demonstrated the ophiasis subtype of AA (symmetric bandlike hair loss), which has a poorer prognosis and is less responsive to therapy.20 In this patient, prior treatment with topical corticosteroids and ILTA in NS failed to induce a response. After a series of injections with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000, she entered remission. Our second case is one of alopecia subtotalis, which responded quickly, and the patient entered remission after just 3 months of treatment. These 2 cases are illustrative of the results that we regularly get and have come to expect with this treatment.
Conclusion
Our novel modified regimen of 2.5 mg/mL ILTA diluted with 1% lidocaine and epinephrine 1:100,000 has yielded a series of excellent outcomes in many of our most challenging AA cases without any untoward effects. Two cases are presented here. Higher-powered studies are needed to validate this new yet simple approach. A split-scalp or split-lesion study comparing ILTA with and without epinephrine 1:100,000 would be warranted for further investigation.
- Mirzoyev SA, Schrum AG, Davis MDP, et al. Lifetime incidence risk of alopecia areata estimated at 2.1 percent by Rochester Epidemiology Project, 1990-2009. J Invest Dermatol. 2014;134:1141-1142.
- Villasante Fricke AC, Miteva M. Epidemiology and burden of alopecia areata: a systematic review. Clin Cosmet Investig Dermatol. 2015;8:397-403.
- Tosti A, Bellavista S, Iorizzo M. Alopecia areata: a long term follow-up study of 191 patients. J Am Acad Dermatol. 2006;55:438-441.
- Walker SA, Rothman S. A statistical study and consideration of endocrine influences. J Invest Dermatol. 1950;14:403-413.
- Charuwichitratana S, Wattanakrai P, Tanrattanakorn S. Randomized double-blind placebo-controlled trial in the treatment of alopecia areata with 0.25% desoximetasone cream. Arch Dermatol. 2000;136:1276-1277.
- Tosti A, Iorizzo M, Botta GL, et al. Efficacy and safety of a new clobetasol propionate 0.05% foam in alopecia areata: a randomized, double-blind placebo-controlled trial. J Eur Acad Dermatol Venereol. 2006;20:1243-1247.
- Kubeyinje EP. Intralesional triamcinolone acetonide in alopecia areata amongst 62 Saudi Arabs. East Afr Med J. 1994;71:674-675.
- Porter D, Burton JL. A comparison of intra-lesional triamcinolonehexacetonide and triamcinolone acetonide in alopecia areata. Br J Dermatol. 1971;85:272-273.
- King B, Ohyama M, Kwon O, et al. Two phase 3 trials of baricitinib for alopecia areata. N Engl J Med. 2022;386:1687-1699. doi:10.1056/NEJMoa2110343
- Lai VWY, Chen G, Gin D, et al. Systemic treatments for alopeciaareata: a systematic review. Australas J Dermatol. 2019;60:E1-E13. doi:10.1111/ajd.12913
- Rokhsar CK, Shupack JL, Vafai JJ, et al. Efficacy of topical sensitizers in the treatment of alopecia areata. J Am Acad Dermatol. 1998;39:751-761.
- Dainichi T, Kabashima K. Alopecia areata: what’s new in epidemiology, pathogenesis, diagnosis, and therapeutic options? J Dermatol Sci. 2017;86:3-12.
- Ito T. Recent advances in the pathogenesis of autoimmune hair loss disease alopecia areata. Clin Dev Immunol. 2013;2013:348546.
- Ramos PM, Anzai A, Duque-Estrada B, et al. Consensus on the treatment of alopecia areata—Brazilian Society of Dermatology. An Bras Dermatol. 2020;95(suppl 1):39-52.
- Yee BE, Tong Y, Goldenberg A, et al. Efficacy of different concentrations of intralesional triamcinolone acetonide for alopecia areata: a systematic review and meta-analysis. J Am Acad Dermatol. 2020;82:1018-1021.
- Na YC, Park R, Jeong HS, et al. Epinephrine vasoconstriction effect time in the scalp differs according to injection site and concentration. Dermatol Surg. 2016;42:1054-1060.
- Samrao A, Fu JM, Harris ST, et al. Bone mineral density in patients with alopecia areata treated with long-term intralesional corticosteroids. J Drugs Dermatol. 2013;12:E36-E40.
- Kong Y, Liu Y, Pan L, et al. Norepinephrine regulates keratinocyte proliferation to promote the growth of hair follicles. Cells Tissues Organs. 2015-2016;201:423-435.
- Fan SM, Chang YT, Chen CL, et al. External light activates hair follicle stem cells through eyes via an ipRGC-SCN-sympathetic neural pathway. Proc Natl Acad Sci U S A. 2018;115:E6880-E6889. Erratum appears in Proc Natl Acad Sci U S A. 2018;115:E12121.
- Spano F, Donovan JC. Alopecia areata: part 1: pathogenesis, diagnosis, and prognosis. Can Fam Physician. 2015;61:751-755.
Alopecia areata (AA) is an autoimmune disorder characterized by transient hair loss with preservation of the hair follicle (HF). The lifetime incidence risk of AA is approximately 2%,1 with a mean age of onset of 25 to 36 years and with no clinically relevant significant differences between sex or ethnicity.2 Most commonly, it presents as round, well-demarcated patches of alopecia on the scalp and spontaneously resolves in nearly 30% of patients. However, severe disease is associated with younger age of presentation and can progress to a total loss of scalp or body hair—referred to as alopecia totalis and alopecia universalis, respectively—thus severely impacting quality of life.3,4
First-line treatment options for AA include potent topical steroids5,6 and intralesional (IL) steroids, most commonly IL triamcinolone acetonide (ILTA). Intralesional steroids have been found to be more effective than topicals in stimulating hair growth at the injection site.7,8 A recent systemic therapy—the Janus kinase inhibitor baricitinib—was approved by the US Food and Drug Administration for AA.9 Other systemic therapies such as oral corticosteroids have been studied in small trials with promising results.10 However, the risks of systemic therapies may outweigh the benefits.9,10
Another less common topical therapy is contact immunotherapy, which involves topical application of an unlicensed non–pharmaceutical-grade agent to areas affected with AA. It is reported to have a wide range of response rates (29%–87%).11
We report 2 cases of extensive AA that were treated with a novel combination regimen— 2.5 mg/mL of ILTA diluted with lidocaine 1% and epinephrine 1:100,000 in place of normal saline (NS)— which is a modification to an already widely used first-line treatment.
Case Reports
Patient 1—An 11-year-old girl presented with nonscarring alopecia of the vertex and occipital scalp. Three years prior she was treated with topical and IL corticosteroids by a different provider. Physical examination revealed almost complete alopecia involving the bottom two-thirds of the occipital scalp as well as the medial eyebrows (Figures 1A and 1B). Over the span of 1 year she was treated with betamethasone dipropionate cream 0.05% and several rounds of ILTA 2.5 mg/mL buffered with NS, with minimal improvement. A year after the initial presentation, the decision was made to initiate monthly injections of ILTA 2.5 mg/mL buffered with 1% lidocaine and epinephrine 1:100,000. Some hair regrowth of the occipital scalp was noted by 3 months, with near-complete regrowth of the scalp hair and eyebrows by 7 months and 5 months, respectively (Figures 1C and 1D). During this period, the patient continued to develop new areas of alopecia of the scalp and eyebrows, which also were injected with this combination. In total, the patient received 8 rounds of IL injections 4 to 6 weeks apart in the scalp and 6 rounds in the eyebrows. The treated areas showed resolution over a follow-up period of 14 months, though there was recurrence at the right medial eyebrow at 5 months. No localized skin atrophy or other adverse effects were noted.
Patient 2—A 34-year-old woman who was otherwise healthy presented with previously untreated AA involving the scalp of 2 months’ duration. Physical examination revealed the following areas of nonscarring alopecia: a 10×10-cm area of the right occipital scalp with some regrowth; a 10×14-cm area of the left parieto-occipital scalp; and a 1-cm area posterior to the vertex (Figure 2A). Given the extensive involvement, the decision was made to initiate ILTA 2.5 mg/mL buffered with 1% lidocaine and epinephrine 1:100,000 once monthly. Appreciable hair regrowth was noted within 1 month, mostly on the parietal scalp. Substantial improvement was noted after 3 months in all affected areas of the hair-bearing scalp, with near-complete regrowth on the left occipital scalp and greater than 50% regrowth on the right occipital scalp (Figure 2B). No adverse effects were noted. She currently has no alopecia.
Comment
Alopecia Pathogenesis—The most widely adopted theory of AA etiology implicates an aberrant immune response. The HF, which is a dynamic “mini-organ” with its own immune and hormonal microenvironment, is considered an “immune-privileged site”—meaning it is less exposed to immune responses than most other body areas. It is hypothesized that AA results from a breakdown in this immune privilege, with the subsequent attack on the peribulbar part of the follicle by CD8+ T lymphocytes. This lymphocytic infiltrate induces apoptosis in the HF keratinocytes, resulting in inhibition of hair shaft production.12 Other theories suggest a link to the sympathetic-adrenal-medullary system and hypothalamic-pituitary-adrenal axis.13
Therapies for Alopecia—Topical and IL corticosteroids are the first-line therapies for localized AA in patients with less than 50% scalp involvement. Triamcinolone acetonide generally is the IL steroid of choice because it is widely available and less atrophogenic than other steroids. Unlike topicals, ILTA bypasses the epidermis when injected, achieving direct access to the HF.14
High-quality controlled studies regarding the use of ILTA in AA are scarce. A meta-analysis concluded that 5 mg/mL and 10 mg/mL of ILTA diluted in NS were equally effective (80.9% [P<.05] vs 76.4% [P<.005], respectively). Concentrations of less than 5 mg/mL of ILTA resulted in lower rates of hair regrowth (62.3%; P=.04).15 The role of diluents other than NS has not been studied.
Benefits of Epinephrine in ILTA Therapy—The role of epinephrine 1:100,000 is to decrease the rate of clearance of triamcinolone acetonide from the HF, allowing for a better therapeutic effect. Laser Doppler blood flowmeter studies have shown that epinephrine 1:100,000 injected in the scalp causes vasoconstriction, thereby decreasing the blood flow rate of clearance of other substances in the same solution.16 Also, a more gradual systemic absorption is achieved, decreasing systemic side effects such as osteoporosis.17
Another potential benefit of epinephrine has been suggested in animal studies that demonstrate the important role of the sympathetic nervous system in HF growth. In a mouse study, chemical sympathectomy led to diminished norepinephrine levels in the skin, accompanied by a decreased keratinocyte proliferation and hair growth. Conversely, norepinephrine was found to promote HF growth in an organotypic skin culture model.18 Topically applied isoproterenol, a panadrenergic receptor agonist, accelerated HF growth in an organotypic skin culture. It also has been shown that external light and temperature changes stimulate hair growth via the sympathetic nervous system, promoting anagen HF growth in cultured skin explants, further linking HF activity with sympathetic nerve activity.19
In our experience, cases of AA that at first failed ILTA 5 mg/mL in NS have been successfully treated with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000. One such case was alopecia totalis, though we do not have high-quality photographs to present for this report. The 2 cases presented here are the ones with the best photographs to demonstrate our outcomes. Both were treated with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000 administered using a 0.5-in long 30-gauge needle, with 0.05 to 0.1 mL per injection approximately 0.51-cm apart. The treatment intervals were 4 weeks, with a maximal dose of 20 mg per session. In addition to the 2 cases reported here, the Table includes 2 other patients in our practice who were successfully treated with this novel regimen.
Prior to adopting this combination regimen, our standard therapy for AA was 5 mg/mL ILTA buffered with NS. Instead of NS, we now use the widely available 1% lidocaine with epinephrine 1:100,000 and dilute the ILTA to 2.5 mg/mL. We postulate that epinephrine 1:100,000 enhances therapeutic efficacy via local vasoconstriction, thus keeping the ILTA in situ longer than NS. This effect allows for a lower concentration of ILTA (2.5 mg/mL) to be effective. Furthermore, epinephrine 1:100,000 may have an independent effect, as suggested in mouse studies.18
Our first case demonstrated the ophiasis subtype of AA (symmetric bandlike hair loss), which has a poorer prognosis and is less responsive to therapy.20 In this patient, prior treatment with topical corticosteroids and ILTA in NS failed to induce a response. After a series of injections with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000, she entered remission. Our second case is one of alopecia subtotalis, which responded quickly, and the patient entered remission after just 3 months of treatment. These 2 cases are illustrative of the results that we regularly get and have come to expect with this treatment.
Conclusion
Our novel modified regimen of 2.5 mg/mL ILTA diluted with 1% lidocaine and epinephrine 1:100,000 has yielded a series of excellent outcomes in many of our most challenging AA cases without any untoward effects. Two cases are presented here. Higher-powered studies are needed to validate this new yet simple approach. A split-scalp or split-lesion study comparing ILTA with and without epinephrine 1:100,000 would be warranted for further investigation.
Alopecia areata (AA) is an autoimmune disorder characterized by transient hair loss with preservation of the hair follicle (HF). The lifetime incidence risk of AA is approximately 2%,1 with a mean age of onset of 25 to 36 years and with no clinically relevant significant differences between sex or ethnicity.2 Most commonly, it presents as round, well-demarcated patches of alopecia on the scalp and spontaneously resolves in nearly 30% of patients. However, severe disease is associated with younger age of presentation and can progress to a total loss of scalp or body hair—referred to as alopecia totalis and alopecia universalis, respectively—thus severely impacting quality of life.3,4
First-line treatment options for AA include potent topical steroids5,6 and intralesional (IL) steroids, most commonly IL triamcinolone acetonide (ILTA). Intralesional steroids have been found to be more effective than topicals in stimulating hair growth at the injection site.7,8 A recent systemic therapy—the Janus kinase inhibitor baricitinib—was approved by the US Food and Drug Administration for AA.9 Other systemic therapies such as oral corticosteroids have been studied in small trials with promising results.10 However, the risks of systemic therapies may outweigh the benefits.9,10
Another less common topical therapy is contact immunotherapy, which involves topical application of an unlicensed non–pharmaceutical-grade agent to areas affected with AA. It is reported to have a wide range of response rates (29%–87%).11
We report 2 cases of extensive AA that were treated with a novel combination regimen— 2.5 mg/mL of ILTA diluted with lidocaine 1% and epinephrine 1:100,000 in place of normal saline (NS)— which is a modification to an already widely used first-line treatment.
Case Reports
Patient 1—An 11-year-old girl presented with nonscarring alopecia of the vertex and occipital scalp. Three years prior she was treated with topical and IL corticosteroids by a different provider. Physical examination revealed almost complete alopecia involving the bottom two-thirds of the occipital scalp as well as the medial eyebrows (Figures 1A and 1B). Over the span of 1 year she was treated with betamethasone dipropionate cream 0.05% and several rounds of ILTA 2.5 mg/mL buffered with NS, with minimal improvement. A year after the initial presentation, the decision was made to initiate monthly injections of ILTA 2.5 mg/mL buffered with 1% lidocaine and epinephrine 1:100,000. Some hair regrowth of the occipital scalp was noted by 3 months, with near-complete regrowth of the scalp hair and eyebrows by 7 months and 5 months, respectively (Figures 1C and 1D). During this period, the patient continued to develop new areas of alopecia of the scalp and eyebrows, which also were injected with this combination. In total, the patient received 8 rounds of IL injections 4 to 6 weeks apart in the scalp and 6 rounds in the eyebrows. The treated areas showed resolution over a follow-up period of 14 months, though there was recurrence at the right medial eyebrow at 5 months. No localized skin atrophy or other adverse effects were noted.
Patient 2—A 34-year-old woman who was otherwise healthy presented with previously untreated AA involving the scalp of 2 months’ duration. Physical examination revealed the following areas of nonscarring alopecia: a 10×10-cm area of the right occipital scalp with some regrowth; a 10×14-cm area of the left parieto-occipital scalp; and a 1-cm area posterior to the vertex (Figure 2A). Given the extensive involvement, the decision was made to initiate ILTA 2.5 mg/mL buffered with 1% lidocaine and epinephrine 1:100,000 once monthly. Appreciable hair regrowth was noted within 1 month, mostly on the parietal scalp. Substantial improvement was noted after 3 months in all affected areas of the hair-bearing scalp, with near-complete regrowth on the left occipital scalp and greater than 50% regrowth on the right occipital scalp (Figure 2B). No adverse effects were noted. She currently has no alopecia.
Comment
Alopecia Pathogenesis—The most widely adopted theory of AA etiology implicates an aberrant immune response. The HF, which is a dynamic “mini-organ” with its own immune and hormonal microenvironment, is considered an “immune-privileged site”—meaning it is less exposed to immune responses than most other body areas. It is hypothesized that AA results from a breakdown in this immune privilege, with the subsequent attack on the peribulbar part of the follicle by CD8+ T lymphocytes. This lymphocytic infiltrate induces apoptosis in the HF keratinocytes, resulting in inhibition of hair shaft production.12 Other theories suggest a link to the sympathetic-adrenal-medullary system and hypothalamic-pituitary-adrenal axis.13
Therapies for Alopecia—Topical and IL corticosteroids are the first-line therapies for localized AA in patients with less than 50% scalp involvement. Triamcinolone acetonide generally is the IL steroid of choice because it is widely available and less atrophogenic than other steroids. Unlike topicals, ILTA bypasses the epidermis when injected, achieving direct access to the HF.14
High-quality controlled studies regarding the use of ILTA in AA are scarce. A meta-analysis concluded that 5 mg/mL and 10 mg/mL of ILTA diluted in NS were equally effective (80.9% [P<.05] vs 76.4% [P<.005], respectively). Concentrations of less than 5 mg/mL of ILTA resulted in lower rates of hair regrowth (62.3%; P=.04).15 The role of diluents other than NS has not been studied.
Benefits of Epinephrine in ILTA Therapy—The role of epinephrine 1:100,000 is to decrease the rate of clearance of triamcinolone acetonide from the HF, allowing for a better therapeutic effect. Laser Doppler blood flowmeter studies have shown that epinephrine 1:100,000 injected in the scalp causes vasoconstriction, thereby decreasing the blood flow rate of clearance of other substances in the same solution.16 Also, a more gradual systemic absorption is achieved, decreasing systemic side effects such as osteoporosis.17
Another potential benefit of epinephrine has been suggested in animal studies that demonstrate the important role of the sympathetic nervous system in HF growth. In a mouse study, chemical sympathectomy led to diminished norepinephrine levels in the skin, accompanied by a decreased keratinocyte proliferation and hair growth. Conversely, norepinephrine was found to promote HF growth in an organotypic skin culture model.18 Topically applied isoproterenol, a panadrenergic receptor agonist, accelerated HF growth in an organotypic skin culture. It also has been shown that external light and temperature changes stimulate hair growth via the sympathetic nervous system, promoting anagen HF growth in cultured skin explants, further linking HF activity with sympathetic nerve activity.19
In our experience, cases of AA that at first failed ILTA 5 mg/mL in NS have been successfully treated with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000. One such case was alopecia totalis, though we do not have high-quality photographs to present for this report. The 2 cases presented here are the ones with the best photographs to demonstrate our outcomes. Both were treated with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000 administered using a 0.5-in long 30-gauge needle, with 0.05 to 0.1 mL per injection approximately 0.51-cm apart. The treatment intervals were 4 weeks, with a maximal dose of 20 mg per session. In addition to the 2 cases reported here, the Table includes 2 other patients in our practice who were successfully treated with this novel regimen.
Prior to adopting this combination regimen, our standard therapy for AA was 5 mg/mL ILTA buffered with NS. Instead of NS, we now use the widely available 1% lidocaine with epinephrine 1:100,000 and dilute the ILTA to 2.5 mg/mL. We postulate that epinephrine 1:100,000 enhances therapeutic efficacy via local vasoconstriction, thus keeping the ILTA in situ longer than NS. This effect allows for a lower concentration of ILTA (2.5 mg/mL) to be effective. Furthermore, epinephrine 1:100,000 may have an independent effect, as suggested in mouse studies.18
Our first case demonstrated the ophiasis subtype of AA (symmetric bandlike hair loss), which has a poorer prognosis and is less responsive to therapy.20 In this patient, prior treatment with topical corticosteroids and ILTA in NS failed to induce a response. After a series of injections with 2.5 mg/mL ILTA in 1% lidocaine and epinephrine 1:100,000, she entered remission. Our second case is one of alopecia subtotalis, which responded quickly, and the patient entered remission after just 3 months of treatment. These 2 cases are illustrative of the results that we regularly get and have come to expect with this treatment.
Conclusion
Our novel modified regimen of 2.5 mg/mL ILTA diluted with 1% lidocaine and epinephrine 1:100,000 has yielded a series of excellent outcomes in many of our most challenging AA cases without any untoward effects. Two cases are presented here. Higher-powered studies are needed to validate this new yet simple approach. A split-scalp or split-lesion study comparing ILTA with and without epinephrine 1:100,000 would be warranted for further investigation.
- Mirzoyev SA, Schrum AG, Davis MDP, et al. Lifetime incidence risk of alopecia areata estimated at 2.1 percent by Rochester Epidemiology Project, 1990-2009. J Invest Dermatol. 2014;134:1141-1142.
- Villasante Fricke AC, Miteva M. Epidemiology and burden of alopecia areata: a systematic review. Clin Cosmet Investig Dermatol. 2015;8:397-403.
- Tosti A, Bellavista S, Iorizzo M. Alopecia areata: a long term follow-up study of 191 patients. J Am Acad Dermatol. 2006;55:438-441.
- Walker SA, Rothman S. A statistical study and consideration of endocrine influences. J Invest Dermatol. 1950;14:403-413.
- Charuwichitratana S, Wattanakrai P, Tanrattanakorn S. Randomized double-blind placebo-controlled trial in the treatment of alopecia areata with 0.25% desoximetasone cream. Arch Dermatol. 2000;136:1276-1277.
- Tosti A, Iorizzo M, Botta GL, et al. Efficacy and safety of a new clobetasol propionate 0.05% foam in alopecia areata: a randomized, double-blind placebo-controlled trial. J Eur Acad Dermatol Venereol. 2006;20:1243-1247.
- Kubeyinje EP. Intralesional triamcinolone acetonide in alopecia areata amongst 62 Saudi Arabs. East Afr Med J. 1994;71:674-675.
- Porter D, Burton JL. A comparison of intra-lesional triamcinolonehexacetonide and triamcinolone acetonide in alopecia areata. Br J Dermatol. 1971;85:272-273.
- King B, Ohyama M, Kwon O, et al. Two phase 3 trials of baricitinib for alopecia areata. N Engl J Med. 2022;386:1687-1699. doi:10.1056/NEJMoa2110343
- Lai VWY, Chen G, Gin D, et al. Systemic treatments for alopeciaareata: a systematic review. Australas J Dermatol. 2019;60:E1-E13. doi:10.1111/ajd.12913
- Rokhsar CK, Shupack JL, Vafai JJ, et al. Efficacy of topical sensitizers in the treatment of alopecia areata. J Am Acad Dermatol. 1998;39:751-761.
- Dainichi T, Kabashima K. Alopecia areata: what’s new in epidemiology, pathogenesis, diagnosis, and therapeutic options? J Dermatol Sci. 2017;86:3-12.
- Ito T. Recent advances in the pathogenesis of autoimmune hair loss disease alopecia areata. Clin Dev Immunol. 2013;2013:348546.
- Ramos PM, Anzai A, Duque-Estrada B, et al. Consensus on the treatment of alopecia areata—Brazilian Society of Dermatology. An Bras Dermatol. 2020;95(suppl 1):39-52.
- Yee BE, Tong Y, Goldenberg A, et al. Efficacy of different concentrations of intralesional triamcinolone acetonide for alopecia areata: a systematic review and meta-analysis. J Am Acad Dermatol. 2020;82:1018-1021.
- Na YC, Park R, Jeong HS, et al. Epinephrine vasoconstriction effect time in the scalp differs according to injection site and concentration. Dermatol Surg. 2016;42:1054-1060.
- Samrao A, Fu JM, Harris ST, et al. Bone mineral density in patients with alopecia areata treated with long-term intralesional corticosteroids. J Drugs Dermatol. 2013;12:E36-E40.
- Kong Y, Liu Y, Pan L, et al. Norepinephrine regulates keratinocyte proliferation to promote the growth of hair follicles. Cells Tissues Organs. 2015-2016;201:423-435.
- Fan SM, Chang YT, Chen CL, et al. External light activates hair follicle stem cells through eyes via an ipRGC-SCN-sympathetic neural pathway. Proc Natl Acad Sci U S A. 2018;115:E6880-E6889. Erratum appears in Proc Natl Acad Sci U S A. 2018;115:E12121.
- Spano F, Donovan JC. Alopecia areata: part 1: pathogenesis, diagnosis, and prognosis. Can Fam Physician. 2015;61:751-755.
- Mirzoyev SA, Schrum AG, Davis MDP, et al. Lifetime incidence risk of alopecia areata estimated at 2.1 percent by Rochester Epidemiology Project, 1990-2009. J Invest Dermatol. 2014;134:1141-1142.
- Villasante Fricke AC, Miteva M. Epidemiology and burden of alopecia areata: a systematic review. Clin Cosmet Investig Dermatol. 2015;8:397-403.
- Tosti A, Bellavista S, Iorizzo M. Alopecia areata: a long term follow-up study of 191 patients. J Am Acad Dermatol. 2006;55:438-441.
- Walker SA, Rothman S. A statistical study and consideration of endocrine influences. J Invest Dermatol. 1950;14:403-413.
- Charuwichitratana S, Wattanakrai P, Tanrattanakorn S. Randomized double-blind placebo-controlled trial in the treatment of alopecia areata with 0.25% desoximetasone cream. Arch Dermatol. 2000;136:1276-1277.
- Tosti A, Iorizzo M, Botta GL, et al. Efficacy and safety of a new clobetasol propionate 0.05% foam in alopecia areata: a randomized, double-blind placebo-controlled trial. J Eur Acad Dermatol Venereol. 2006;20:1243-1247.
- Kubeyinje EP. Intralesional triamcinolone acetonide in alopecia areata amongst 62 Saudi Arabs. East Afr Med J. 1994;71:674-675.
- Porter D, Burton JL. A comparison of intra-lesional triamcinolonehexacetonide and triamcinolone acetonide in alopecia areata. Br J Dermatol. 1971;85:272-273.
- King B, Ohyama M, Kwon O, et al. Two phase 3 trials of baricitinib for alopecia areata. N Engl J Med. 2022;386:1687-1699. doi:10.1056/NEJMoa2110343
- Lai VWY, Chen G, Gin D, et al. Systemic treatments for alopeciaareata: a systematic review. Australas J Dermatol. 2019;60:E1-E13. doi:10.1111/ajd.12913
- Rokhsar CK, Shupack JL, Vafai JJ, et al. Efficacy of topical sensitizers in the treatment of alopecia areata. J Am Acad Dermatol. 1998;39:751-761.
- Dainichi T, Kabashima K. Alopecia areata: what’s new in epidemiology, pathogenesis, diagnosis, and therapeutic options? J Dermatol Sci. 2017;86:3-12.
- Ito T. Recent advances in the pathogenesis of autoimmune hair loss disease alopecia areata. Clin Dev Immunol. 2013;2013:348546.
- Ramos PM, Anzai A, Duque-Estrada B, et al. Consensus on the treatment of alopecia areata—Brazilian Society of Dermatology. An Bras Dermatol. 2020;95(suppl 1):39-52.
- Yee BE, Tong Y, Goldenberg A, et al. Efficacy of different concentrations of intralesional triamcinolone acetonide for alopecia areata: a systematic review and meta-analysis. J Am Acad Dermatol. 2020;82:1018-1021.
- Na YC, Park R, Jeong HS, et al. Epinephrine vasoconstriction effect time in the scalp differs according to injection site and concentration. Dermatol Surg. 2016;42:1054-1060.
- Samrao A, Fu JM, Harris ST, et al. Bone mineral density in patients with alopecia areata treated with long-term intralesional corticosteroids. J Drugs Dermatol. 2013;12:E36-E40.
- Kong Y, Liu Y, Pan L, et al. Norepinephrine regulates keratinocyte proliferation to promote the growth of hair follicles. Cells Tissues Organs. 2015-2016;201:423-435.
- Fan SM, Chang YT, Chen CL, et al. External light activates hair follicle stem cells through eyes via an ipRGC-SCN-sympathetic neural pathway. Proc Natl Acad Sci U S A. 2018;115:E6880-E6889. Erratum appears in Proc Natl Acad Sci U S A. 2018;115:E12121.
- Spano F, Donovan JC. Alopecia areata: part 1: pathogenesis, diagnosis, and prognosis. Can Fam Physician. 2015;61:751-755.
Practice Points
- Patients with alopecia areata that is refractory to first-line treatments may benefit from intralesional triamcinolone acetonide (ILTA) diluted to 2.5 mg/mL in 1% lidocaine and epinephrine 1:100,000 in place of normal saline.
- Local vasoconstriction due to epinephrine may potentiate ILTA effects and play an independent role.
Pancreatic cancer screening in new-onset diabetes appears cost effective
A risk-tailored early-detection strategy for pancreatic cancer that targets patients with new-onset diabetes could be cost effective, according to a recent study.
Screening for pancreatic ductal adenocarcinoma in asymptomatic adults is not recommended, but patients with new-onset diabetes have a risk that’s eight times higher than expected. Screening these patients could improve diagnosis and survival rates if the cancer can be identified at earlier stages, researchers led by Louise Wang, MD, a gastroenterology fellow at the University of Pennsylvania, Philadelphia, wrote in Clinical Gastroenterology and Hepatology.
“As we continue to improve therapies for early-stage pancreatic cancers, especially among the local/resectable stage, the case for the targeted early-detection strategy will be stronger,” they wrote. “Policy makers should take into consideration these novel findings when formulating [pancreatic ductal adenocarcinoma] screening policy and making coverage determinations.”
The research team compared early detection strategies for pancreatic ductal adenocarcinoma that target new-onset diabetes patients at age 50 years and older with standard of care, defined as no early detection strategy. They looked at various minimal predicted cancer risk thresholds versus current standard of care in a Markov state-transition decision model. The analysis assumed a health care sector perspective and a lifetime horizon, with two willingness-to-pay thresholds ($100,000 and $150,000) per quality-adjusted life-year gained.
The researchers used data from one of their previously published studies, which included 89,881 patients with new-onset diabetes diagnosed at age 50 or older. The cumulative incidence of pancreatic cancer was 0.42% during the 3 years after diagnosis.
In the early detection strategy, all patients 50 years and older who were newly diagnosed with diabetes mellitus were placed into low-risk and high-risk cohorts based on their predicted 3-year risk of pancreatic ductal adenocarcinoma under a range of assumed minimum-risk thresholds – 0.5%, 1%, 2%, 3%, 4%, and 5%; these thresholds were based on a previously established prediction model.
The research team found that the early detection strategy that targeted patients with a minimum predicted 3-year pancreatic ductal adenocarcinoma risk of 1% was cost effective, based on a willingness-to-pay threshold of $150,000 per quality-adjusted life-year. The incremental cost-effectiveness ratio was $116,911 per quality-adjusted life-year.
At a willingness-to-pay threshold of $100,000 per quality-adjusted life-year, the early detection strategy at the 2% risk threshold was cost effective. The incremental cost-effectiveness ratio was $63,045 per quality-adjusted life-year.
The most influential factors included the proportion of pancreatic ductal adenocarcinomas detected at the local stage, costs of treatment for metastatic cancer, utilities of local and regional cancers, and sensitivity of screening.
A probabilistic sensitivity analysis confirmed that, at a willingness-to-pay threshold of $150,000, early detection at the 1% risk threshold was favored at 30.6%, followed by the 0.5% risk threshold at 20.4%, compared with the standard of care at 1.7%. In addition, at a willingness-to-pay threshold of $100,000, early detection at the 1% risk threshold was favored at 27.3%, followed by the 2% risk threshold at 22.8%, as compared with the standard of care at 2%.
The two early detection strategies were cost effective, capturing 26%-45% of the pancreatic ductal adenocarcinoma cases in patients with new-onset diabetes.
The study authors noted several limitations, including the inability to incorporate out-of-pocket costs for patients, as well as focusing the analysis on the health care perspective.
“We acknowledge that, by incorporating the full consequences of decisions for all stakeholders, a societal perspective would have offered a more complete view on which to base public policy,” they wrote.
At the same time, “given the substantial prevalence of [new-onset diabetes] among [pancreatic ductal adenocarcinoma] cases, this strategy could improve the survival of a substantial proportion of sporadic PDAC cases in the general population,” they concluded.
The study authors reported various disclosures, including grants and research support from Takeda Pharmaceuticals USA, Janssen Pharmaceuticals, the National Institutes of Health, the Crohn’s and Colitis Foundation, Lilly Oncology, GSK, and Clovis Oncology.
Earlier detection of pancreatic ductal adenocarcinoma (PDAC) is essential to improving the survival for the group of patients diagnosed with PDAC each year. New-onset diabetes in adults 50 years or older is recognized as a risk factor for being diagnosed with PDAC within the following 3 years.
This study by Wang et al. uses previously described clinical prediction models to stratify the risk of PDAC in patients with new-onset diabetes. These models include age, body mass index, weight change, smoking, diabetic medications, and laboratory values (hemoglobin A1c, cholesterol, creatinine, alkaline phosphatase). They ran simulation models to determine the cost-effectiveness of screening for pancreatic cancer at various risk cut-offs. At the $150,000 willingness-to-pay threshold per quality-adjusted life-year, the 1% risk threshold was cost-effective. Stage shifting from a higher-stage cancer to a lower-stage cancer was the driving force behind the cost-effectiveness ratios.
Providers need to have a high index of suspicion when an adult over the age of 50 has had a new diagnosis of diabetes. Abnormalities detected in laboratory data, weight trends, symptoms, a history of underlying smoking or pancreatic disease may appropriately prompt an MRI/MRCP or endoscopic ultrasound. Better and more accessible risk progression calculators for these patients could be used in real time. The current study by Wang et al. will be a helpful tool as well for navigating disputes with payers about the utility of covering screening tests in the subgroup of patients that are higher risk.
Mark A. Gromski, MD, is assistant professor of medicine at Indiana University School of Medicine and a pancreatobiliary specialist and advanced endoscopist at IU Health. He reports having no relevant disclosures.
Earlier detection of pancreatic ductal adenocarcinoma (PDAC) is essential to improving the survival for the group of patients diagnosed with PDAC each year. New-onset diabetes in adults 50 years or older is recognized as a risk factor for being diagnosed with PDAC within the following 3 years.
This study by Wang et al. uses previously described clinical prediction models to stratify the risk of PDAC in patients with new-onset diabetes. These models include age, body mass index, weight change, smoking, diabetic medications, and laboratory values (hemoglobin A1c, cholesterol, creatinine, alkaline phosphatase). They ran simulation models to determine the cost-effectiveness of screening for pancreatic cancer at various risk cut-offs. At the $150,000 willingness-to-pay threshold per quality-adjusted life-year, the 1% risk threshold was cost-effective. Stage shifting from a higher-stage cancer to a lower-stage cancer was the driving force behind the cost-effectiveness ratios.
Providers need to have a high index of suspicion when an adult over the age of 50 has had a new diagnosis of diabetes. Abnormalities detected in laboratory data, weight trends, symptoms, a history of underlying smoking or pancreatic disease may appropriately prompt an MRI/MRCP or endoscopic ultrasound. Better and more accessible risk progression calculators for these patients could be used in real time. The current study by Wang et al. will be a helpful tool as well for navigating disputes with payers about the utility of covering screening tests in the subgroup of patients that are higher risk.
Mark A. Gromski, MD, is assistant professor of medicine at Indiana University School of Medicine and a pancreatobiliary specialist and advanced endoscopist at IU Health. He reports having no relevant disclosures.
Earlier detection of pancreatic ductal adenocarcinoma (PDAC) is essential to improving the survival for the group of patients diagnosed with PDAC each year. New-onset diabetes in adults 50 years or older is recognized as a risk factor for being diagnosed with PDAC within the following 3 years.
This study by Wang et al. uses previously described clinical prediction models to stratify the risk of PDAC in patients with new-onset diabetes. These models include age, body mass index, weight change, smoking, diabetic medications, and laboratory values (hemoglobin A1c, cholesterol, creatinine, alkaline phosphatase). They ran simulation models to determine the cost-effectiveness of screening for pancreatic cancer at various risk cut-offs. At the $150,000 willingness-to-pay threshold per quality-adjusted life-year, the 1% risk threshold was cost-effective. Stage shifting from a higher-stage cancer to a lower-stage cancer was the driving force behind the cost-effectiveness ratios.
Providers need to have a high index of suspicion when an adult over the age of 50 has had a new diagnosis of diabetes. Abnormalities detected in laboratory data, weight trends, symptoms, a history of underlying smoking or pancreatic disease may appropriately prompt an MRI/MRCP or endoscopic ultrasound. Better and more accessible risk progression calculators for these patients could be used in real time. The current study by Wang et al. will be a helpful tool as well for navigating disputes with payers about the utility of covering screening tests in the subgroup of patients that are higher risk.
Mark A. Gromski, MD, is assistant professor of medicine at Indiana University School of Medicine and a pancreatobiliary specialist and advanced endoscopist at IU Health. He reports having no relevant disclosures.
A risk-tailored early-detection strategy for pancreatic cancer that targets patients with new-onset diabetes could be cost effective, according to a recent study.
Screening for pancreatic ductal adenocarcinoma in asymptomatic adults is not recommended, but patients with new-onset diabetes have a risk that’s eight times higher than expected. Screening these patients could improve diagnosis and survival rates if the cancer can be identified at earlier stages, researchers led by Louise Wang, MD, a gastroenterology fellow at the University of Pennsylvania, Philadelphia, wrote in Clinical Gastroenterology and Hepatology.
“As we continue to improve therapies for early-stage pancreatic cancers, especially among the local/resectable stage, the case for the targeted early-detection strategy will be stronger,” they wrote. “Policy makers should take into consideration these novel findings when formulating [pancreatic ductal adenocarcinoma] screening policy and making coverage determinations.”
The research team compared early detection strategies for pancreatic ductal adenocarcinoma that target new-onset diabetes patients at age 50 years and older with standard of care, defined as no early detection strategy. They looked at various minimal predicted cancer risk thresholds versus current standard of care in a Markov state-transition decision model. The analysis assumed a health care sector perspective and a lifetime horizon, with two willingness-to-pay thresholds ($100,000 and $150,000) per quality-adjusted life-year gained.
The researchers used data from one of their previously published studies, which included 89,881 patients with new-onset diabetes diagnosed at age 50 or older. The cumulative incidence of pancreatic cancer was 0.42% during the 3 years after diagnosis.
In the early detection strategy, all patients 50 years and older who were newly diagnosed with diabetes mellitus were placed into low-risk and high-risk cohorts based on their predicted 3-year risk of pancreatic ductal adenocarcinoma under a range of assumed minimum-risk thresholds – 0.5%, 1%, 2%, 3%, 4%, and 5%; these thresholds were based on a previously established prediction model.
The research team found that the early detection strategy that targeted patients with a minimum predicted 3-year pancreatic ductal adenocarcinoma risk of 1% was cost effective, based on a willingness-to-pay threshold of $150,000 per quality-adjusted life-year. The incremental cost-effectiveness ratio was $116,911 per quality-adjusted life-year.
At a willingness-to-pay threshold of $100,000 per quality-adjusted life-year, the early detection strategy at the 2% risk threshold was cost effective. The incremental cost-effectiveness ratio was $63,045 per quality-adjusted life-year.
The most influential factors included the proportion of pancreatic ductal adenocarcinomas detected at the local stage, costs of treatment for metastatic cancer, utilities of local and regional cancers, and sensitivity of screening.
A probabilistic sensitivity analysis confirmed that, at a willingness-to-pay threshold of $150,000, early detection at the 1% risk threshold was favored at 30.6%, followed by the 0.5% risk threshold at 20.4%, compared with the standard of care at 1.7%. In addition, at a willingness-to-pay threshold of $100,000, early detection at the 1% risk threshold was favored at 27.3%, followed by the 2% risk threshold at 22.8%, as compared with the standard of care at 2%.
The two early detection strategies were cost effective, capturing 26%-45% of the pancreatic ductal adenocarcinoma cases in patients with new-onset diabetes.
The study authors noted several limitations, including the inability to incorporate out-of-pocket costs for patients, as well as focusing the analysis on the health care perspective.
“We acknowledge that, by incorporating the full consequences of decisions for all stakeholders, a societal perspective would have offered a more complete view on which to base public policy,” they wrote.
At the same time, “given the substantial prevalence of [new-onset diabetes] among [pancreatic ductal adenocarcinoma] cases, this strategy could improve the survival of a substantial proportion of sporadic PDAC cases in the general population,” they concluded.
The study authors reported various disclosures, including grants and research support from Takeda Pharmaceuticals USA, Janssen Pharmaceuticals, the National Institutes of Health, the Crohn’s and Colitis Foundation, Lilly Oncology, GSK, and Clovis Oncology.
A risk-tailored early-detection strategy for pancreatic cancer that targets patients with new-onset diabetes could be cost effective, according to a recent study.
Screening for pancreatic ductal adenocarcinoma in asymptomatic adults is not recommended, but patients with new-onset diabetes have a risk that’s eight times higher than expected. Screening these patients could improve diagnosis and survival rates if the cancer can be identified at earlier stages, researchers led by Louise Wang, MD, a gastroenterology fellow at the University of Pennsylvania, Philadelphia, wrote in Clinical Gastroenterology and Hepatology.
“As we continue to improve therapies for early-stage pancreatic cancers, especially among the local/resectable stage, the case for the targeted early-detection strategy will be stronger,” they wrote. “Policy makers should take into consideration these novel findings when formulating [pancreatic ductal adenocarcinoma] screening policy and making coverage determinations.”
The research team compared early detection strategies for pancreatic ductal adenocarcinoma that target new-onset diabetes patients at age 50 years and older with standard of care, defined as no early detection strategy. They looked at various minimal predicted cancer risk thresholds versus current standard of care in a Markov state-transition decision model. The analysis assumed a health care sector perspective and a lifetime horizon, with two willingness-to-pay thresholds ($100,000 and $150,000) per quality-adjusted life-year gained.
The researchers used data from one of their previously published studies, which included 89,881 patients with new-onset diabetes diagnosed at age 50 or older. The cumulative incidence of pancreatic cancer was 0.42% during the 3 years after diagnosis.
In the early detection strategy, all patients 50 years and older who were newly diagnosed with diabetes mellitus were placed into low-risk and high-risk cohorts based on their predicted 3-year risk of pancreatic ductal adenocarcinoma under a range of assumed minimum-risk thresholds – 0.5%, 1%, 2%, 3%, 4%, and 5%; these thresholds were based on a previously established prediction model.
The research team found that the early detection strategy that targeted patients with a minimum predicted 3-year pancreatic ductal adenocarcinoma risk of 1% was cost effective, based on a willingness-to-pay threshold of $150,000 per quality-adjusted life-year. The incremental cost-effectiveness ratio was $116,911 per quality-adjusted life-year.
At a willingness-to-pay threshold of $100,000 per quality-adjusted life-year, the early detection strategy at the 2% risk threshold was cost effective. The incremental cost-effectiveness ratio was $63,045 per quality-adjusted life-year.
The most influential factors included the proportion of pancreatic ductal adenocarcinomas detected at the local stage, costs of treatment for metastatic cancer, utilities of local and regional cancers, and sensitivity of screening.
A probabilistic sensitivity analysis confirmed that, at a willingness-to-pay threshold of $150,000, early detection at the 1% risk threshold was favored at 30.6%, followed by the 0.5% risk threshold at 20.4%, compared with the standard of care at 1.7%. In addition, at a willingness-to-pay threshold of $100,000, early detection at the 1% risk threshold was favored at 27.3%, followed by the 2% risk threshold at 22.8%, as compared with the standard of care at 2%.
The two early detection strategies were cost effective, capturing 26%-45% of the pancreatic ductal adenocarcinoma cases in patients with new-onset diabetes.
The study authors noted several limitations, including the inability to incorporate out-of-pocket costs for patients, as well as focusing the analysis on the health care perspective.
“We acknowledge that, by incorporating the full consequences of decisions for all stakeholders, a societal perspective would have offered a more complete view on which to base public policy,” they wrote.
At the same time, “given the substantial prevalence of [new-onset diabetes] among [pancreatic ductal adenocarcinoma] cases, this strategy could improve the survival of a substantial proportion of sporadic PDAC cases in the general population,” they concluded.
The study authors reported various disclosures, including grants and research support from Takeda Pharmaceuticals USA, Janssen Pharmaceuticals, the National Institutes of Health, the Crohn’s and Colitis Foundation, Lilly Oncology, GSK, and Clovis Oncology.
FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY
Mysterious cases of illness with an unusual cause
So begins the search for evidence.
No relations or common journeys
Between March and July 2021, cases of the bacterial infectious disease sprang up in Georgia, Kansas, Minnesota, and Texas, with the disease being fatal for two of those affected. Usually, cases of melioidosis occur in the United States after traveling to regions where the pathogen is prevalent. However, none of the patients had undertaken any previous international travel.
When the genomes of the bacterial strains (Burkholderia pseudomallei) were sequenced, they showed a high level of concordance, suggesting a common source of infection. The bacterial strain is similar to those that are found in Southeast Asia above all. An imported product from there was taken into consideration as the trigger.
The Centers for Disease Control and Prevention examined blood samples from the patients, as well as samples from the soil, water, food, and household items around their homes.
Aroma spray as a trigger
In October, the cause of the melioidosis was finally identified in the house of the patient from Georgia: an aromatherapy spray. The genetic fingerprint of the bacterial strain matched with that from the other patients. The common trigger was thus discovered.
The contaminated spray, with a lavender-chamomile scent for room fragrancing, was sold between February and October in some branches of Walmart, as well as in their online store. The product was therefore recalled and it was checked whether the ingredients were also being used in other products.
The CDC requested physicians to also take melioidosis into account if they were presented with acute bacterial infections that did not respond to normal antibiotics and to inquire whether the affected room spray had been used.
More information about melioidosis
Melioidosis is an infectious disease affecting humans and animals. The trigger is the bacteria B pseudomallei. The disease appears predominantly in tropical regions, especially in Southeast Asia and northern Australia.
Transmission
The bacteria can be found in contaminated water and soil. It is disseminated between humans and animals through direct contact with the infectious source, such as through inhaling dust particles or water droplets, or through consuming contaminated water or food. Human-to-human transmission is extremely rare. Recently however, tropical saltwater fish were identified as potential carriers.
Symptoms
Melioidosis has a wide range of symptoms, which can lead to its being confused with other diseases such as tuberculosis or other forms of pneumonia. There are different forms of the disease, each with different symptoms.
- Localized infection: localized pain and swelling, fever, ulceration, and abscess.
- Pulmonary infection: cough, chest pain, high fever, headaches, and loss of appetite
- Bacteremia: fever, headaches, breathing problems, stomach discomfort, joint pain, and disorientation.
- Disseminated infection: fever, weight loss, stomach or chest pain, muscle or joint pain, headaches, central nervous system infections, and epileptic seizures.
The incubation time is not clearly defined and can be from 1 day to several years; however, the symptoms mostly emerge 2-4 weeks after exposure. The risk factors include diabetes, high alcohol consumption, chronic pulmonary or kidney disease, and immunodeficiencies.
Diagnosis based on the symptoms is often difficult since the clinical picture is similar to other, more common conditions.
Therapy
If the melioidosis is identified as such, it can be treated with only mildly effective antibiotics, since it has a natural resistance to many commonly used antibiotics. The type of infection and the course of treatment also affects the long-term outcome. Without treatment, 90% of the infections have a fatal outcome. With appropriate treatment, the mortality rate still lies at 40%.
Therapy generally begins with intravenous antibiotic therapy for at least 2-8 weeks (ceftazidime or meropenem). Oral antibiotic therapy then follows for 3-6 months (trimethoprim-sulfamethoxazole or amoxicillin/clavulanic acid). If the patient is allergic to penicillin, alternative antibiotics can be used.
Use as a bioweapon
The CDC classifies B. pseudomallei as a potential pathogen for biological attack (class-B candidate). The agency lists the potential reasons for use as a bioweapon as:
- The pathogen can be found naturally in certain regions.
- The triggered disease can take a serious course and ultimately be fatal without appropriate therapy.
- In the past, the United States has used similar pathogens in wars as bioweapons.
In a potential attack, the pathogen could be spread through air, water, or food, and by doing so, many people would be exposed. Any contact with the bacteria can result in melioidosis. As the bacteria cannot be seen, smelled, or tasted, the biological attack would not be recognized for some time. A certain amount of time can also pass until the pathogen is identified, once fever and respiratory diseases have developed.
In such an emergency, the CDC would collaborate with other federal and local authorities to supply specialized testing laboratories and provide the public with information.
This content was translated from Coliquio. A version appeared on Medscape.com.
So begins the search for evidence.
No relations or common journeys
Between March and July 2021, cases of the bacterial infectious disease sprang up in Georgia, Kansas, Minnesota, and Texas, with the disease being fatal for two of those affected. Usually, cases of melioidosis occur in the United States after traveling to regions where the pathogen is prevalent. However, none of the patients had undertaken any previous international travel.
When the genomes of the bacterial strains (Burkholderia pseudomallei) were sequenced, they showed a high level of concordance, suggesting a common source of infection. The bacterial strain is similar to those that are found in Southeast Asia above all. An imported product from there was taken into consideration as the trigger.
The Centers for Disease Control and Prevention examined blood samples from the patients, as well as samples from the soil, water, food, and household items around their homes.
Aroma spray as a trigger
In October, the cause of the melioidosis was finally identified in the house of the patient from Georgia: an aromatherapy spray. The genetic fingerprint of the bacterial strain matched with that from the other patients. The common trigger was thus discovered.
The contaminated spray, with a lavender-chamomile scent for room fragrancing, was sold between February and October in some branches of Walmart, as well as in their online store. The product was therefore recalled and it was checked whether the ingredients were also being used in other products.
The CDC requested physicians to also take melioidosis into account if they were presented with acute bacterial infections that did not respond to normal antibiotics and to inquire whether the affected room spray had been used.
More information about melioidosis
Melioidosis is an infectious disease affecting humans and animals. The trigger is the bacteria B pseudomallei. The disease appears predominantly in tropical regions, especially in Southeast Asia and northern Australia.
Transmission
The bacteria can be found in contaminated water and soil. It is disseminated between humans and animals through direct contact with the infectious source, such as through inhaling dust particles or water droplets, or through consuming contaminated water or food. Human-to-human transmission is extremely rare. Recently however, tropical saltwater fish were identified as potential carriers.
Symptoms
Melioidosis has a wide range of symptoms, which can lead to its being confused with other diseases such as tuberculosis or other forms of pneumonia. There are different forms of the disease, each with different symptoms.
- Localized infection: localized pain and swelling, fever, ulceration, and abscess.
- Pulmonary infection: cough, chest pain, high fever, headaches, and loss of appetite
- Bacteremia: fever, headaches, breathing problems, stomach discomfort, joint pain, and disorientation.
- Disseminated infection: fever, weight loss, stomach or chest pain, muscle or joint pain, headaches, central nervous system infections, and epileptic seizures.
The incubation time is not clearly defined and can be from 1 day to several years; however, the symptoms mostly emerge 2-4 weeks after exposure. The risk factors include diabetes, high alcohol consumption, chronic pulmonary or kidney disease, and immunodeficiencies.
Diagnosis based on the symptoms is often difficult since the clinical picture is similar to other, more common conditions.
Therapy
If the melioidosis is identified as such, it can be treated with only mildly effective antibiotics, since it has a natural resistance to many commonly used antibiotics. The type of infection and the course of treatment also affects the long-term outcome. Without treatment, 90% of the infections have a fatal outcome. With appropriate treatment, the mortality rate still lies at 40%.
Therapy generally begins with intravenous antibiotic therapy for at least 2-8 weeks (ceftazidime or meropenem). Oral antibiotic therapy then follows for 3-6 months (trimethoprim-sulfamethoxazole or amoxicillin/clavulanic acid). If the patient is allergic to penicillin, alternative antibiotics can be used.
Use as a bioweapon
The CDC classifies B. pseudomallei as a potential pathogen for biological attack (class-B candidate). The agency lists the potential reasons for use as a bioweapon as:
- The pathogen can be found naturally in certain regions.
- The triggered disease can take a serious course and ultimately be fatal without appropriate therapy.
- In the past, the United States has used similar pathogens in wars as bioweapons.
In a potential attack, the pathogen could be spread through air, water, or food, and by doing so, many people would be exposed. Any contact with the bacteria can result in melioidosis. As the bacteria cannot be seen, smelled, or tasted, the biological attack would not be recognized for some time. A certain amount of time can also pass until the pathogen is identified, once fever and respiratory diseases have developed.
In such an emergency, the CDC would collaborate with other federal and local authorities to supply specialized testing laboratories and provide the public with information.
This content was translated from Coliquio. A version appeared on Medscape.com.
So begins the search for evidence.
No relations or common journeys
Between March and July 2021, cases of the bacterial infectious disease sprang up in Georgia, Kansas, Minnesota, and Texas, with the disease being fatal for two of those affected. Usually, cases of melioidosis occur in the United States after traveling to regions where the pathogen is prevalent. However, none of the patients had undertaken any previous international travel.
When the genomes of the bacterial strains (Burkholderia pseudomallei) were sequenced, they showed a high level of concordance, suggesting a common source of infection. The bacterial strain is similar to those that are found in Southeast Asia above all. An imported product from there was taken into consideration as the trigger.
The Centers for Disease Control and Prevention examined blood samples from the patients, as well as samples from the soil, water, food, and household items around their homes.
Aroma spray as a trigger
In October, the cause of the melioidosis was finally identified in the house of the patient from Georgia: an aromatherapy spray. The genetic fingerprint of the bacterial strain matched with that from the other patients. The common trigger was thus discovered.
The contaminated spray, with a lavender-chamomile scent for room fragrancing, was sold between February and October in some branches of Walmart, as well as in their online store. The product was therefore recalled and it was checked whether the ingredients were also being used in other products.
The CDC requested physicians to also take melioidosis into account if they were presented with acute bacterial infections that did not respond to normal antibiotics and to inquire whether the affected room spray had been used.
More information about melioidosis
Melioidosis is an infectious disease affecting humans and animals. The trigger is the bacteria B pseudomallei. The disease appears predominantly in tropical regions, especially in Southeast Asia and northern Australia.
Transmission
The bacteria can be found in contaminated water and soil. It is disseminated between humans and animals through direct contact with the infectious source, such as through inhaling dust particles or water droplets, or through consuming contaminated water or food. Human-to-human transmission is extremely rare. Recently however, tropical saltwater fish were identified as potential carriers.
Symptoms
Melioidosis has a wide range of symptoms, which can lead to its being confused with other diseases such as tuberculosis or other forms of pneumonia. There are different forms of the disease, each with different symptoms.
- Localized infection: localized pain and swelling, fever, ulceration, and abscess.
- Pulmonary infection: cough, chest pain, high fever, headaches, and loss of appetite
- Bacteremia: fever, headaches, breathing problems, stomach discomfort, joint pain, and disorientation.
- Disseminated infection: fever, weight loss, stomach or chest pain, muscle or joint pain, headaches, central nervous system infections, and epileptic seizures.
The incubation time is not clearly defined and can be from 1 day to several years; however, the symptoms mostly emerge 2-4 weeks after exposure. The risk factors include diabetes, high alcohol consumption, chronic pulmonary or kidney disease, and immunodeficiencies.
Diagnosis based on the symptoms is often difficult since the clinical picture is similar to other, more common conditions.
Therapy
If the melioidosis is identified as such, it can be treated with only mildly effective antibiotics, since it has a natural resistance to many commonly used antibiotics. The type of infection and the course of treatment also affects the long-term outcome. Without treatment, 90% of the infections have a fatal outcome. With appropriate treatment, the mortality rate still lies at 40%.
Therapy generally begins with intravenous antibiotic therapy for at least 2-8 weeks (ceftazidime or meropenem). Oral antibiotic therapy then follows for 3-6 months (trimethoprim-sulfamethoxazole or amoxicillin/clavulanic acid). If the patient is allergic to penicillin, alternative antibiotics can be used.
Use as a bioweapon
The CDC classifies B. pseudomallei as a potential pathogen for biological attack (class-B candidate). The agency lists the potential reasons for use as a bioweapon as:
- The pathogen can be found naturally in certain regions.
- The triggered disease can take a serious course and ultimately be fatal without appropriate therapy.
- In the past, the United States has used similar pathogens in wars as bioweapons.
In a potential attack, the pathogen could be spread through air, water, or food, and by doing so, many people would be exposed. Any contact with the bacteria can result in melioidosis. As the bacteria cannot be seen, smelled, or tasted, the biological attack would not be recognized for some time. A certain amount of time can also pass until the pathogen is identified, once fever and respiratory diseases have developed.
In such an emergency, the CDC would collaborate with other federal and local authorities to supply specialized testing laboratories and provide the public with information.
This content was translated from Coliquio. A version appeared on Medscape.com.