User login
Don’t Miss Practice Management Tips for Novice and Seasoned Surgeons
Young vascular surgeons looking to make their marks and balance their lives, as well as more experienced surgeons seeking tips for more effective practice and career management, should consider the session led by Jeffrey Siracuse, MD, of Boston Medical Center, and Courtney Warner, MD, of Albany Medical College, Saratoga Springs, N.Y.
“This will be a great and informative session that, although geared toward young surgeons, will be highly useful for surgeons of all experience levels,” Dr. Siracuse said of the session.
“Topics include building a successful academic practice, building a successful private practice, how to work with other specialties, how to negotiate a contract, how to improve one’s current job, and more on the business side of medicine,” said Dr. Siracuse. The discussions will be followed by a Q&A panel.
The first job after a fellowship is often the first “real job” of any type that new surgeons have had, Dr. Siracuse said. Medical school offers many things, but young surgeons may be underprepared for how to negotiate for one’s first surgical position and how to set oneself up for success, he explained. That said, the keys for success are significantly different for surgeons entering an academic setting or private practice, he noted.
The session kicks off with presenters addressing both types of practice.
Faisal Aziz, MD, of Penn State Hershey College of Medicine, Hershey, addresses the “Top 10 Roadblocks to a Successful Academic Practice,” and Scott Berman, MD, of Carondelet Medical Group in Tucson, Ariz., takes on the “Top 10 Roadblocks to a Successful Private Practice.”
Some subjects likely to prompt lively discussion among seasoned veteran surgeons as well as novices include how to effectively negotiate and renegotiate contracts. The contracts presentation, “Top 10 Tips on Negotiating Contracts and Other Things I Learned in Business School,” is scheduled to be given by Bruce Perler, MD, of Johns Hopkins University in Baltimore, Md.
In addition, surgeons at all levels of experience can benefit from tips on how to work with individuals in other specialties, especially if one is competing with them for patients and cases, Dr. Siracuse said. Brandon Propper, MD, of San Antonio Military Medical Center, Texas, steps up to the plate with his “Top 10 Ways to Work With Other Specialties.”
The Practice Management Tips and Tricks for Young Vascular Surgeons session is recommended by the Community Practice Committee and the Young Surgeons Committee.
Friday, June 22
1:30 – 3:00 p.m.
HCC, Room 311
C5: Practice Management Tips and Tricks for Young Vascular Surgeons
Young vascular surgeons looking to make their marks and balance their lives, as well as more experienced surgeons seeking tips for more effective practice and career management, should consider the session led by Jeffrey Siracuse, MD, of Boston Medical Center, and Courtney Warner, MD, of Albany Medical College, Saratoga Springs, N.Y.
“This will be a great and informative session that, although geared toward young surgeons, will be highly useful for surgeons of all experience levels,” Dr. Siracuse said of the session.
“Topics include building a successful academic practice, building a successful private practice, how to work with other specialties, how to negotiate a contract, how to improve one’s current job, and more on the business side of medicine,” said Dr. Siracuse. The discussions will be followed by a Q&A panel.
The first job after a fellowship is often the first “real job” of any type that new surgeons have had, Dr. Siracuse said. Medical school offers many things, but young surgeons may be underprepared for how to negotiate for one’s first surgical position and how to set oneself up for success, he explained. That said, the keys for success are significantly different for surgeons entering an academic setting or private practice, he noted.
The session kicks off with presenters addressing both types of practice.
Faisal Aziz, MD, of Penn State Hershey College of Medicine, Hershey, addresses the “Top 10 Roadblocks to a Successful Academic Practice,” and Scott Berman, MD, of Carondelet Medical Group in Tucson, Ariz., takes on the “Top 10 Roadblocks to a Successful Private Practice.”
Some subjects likely to prompt lively discussion among seasoned veteran surgeons as well as novices include how to effectively negotiate and renegotiate contracts. The contracts presentation, “Top 10 Tips on Negotiating Contracts and Other Things I Learned in Business School,” is scheduled to be given by Bruce Perler, MD, of Johns Hopkins University in Baltimore, Md.
In addition, surgeons at all levels of experience can benefit from tips on how to work with individuals in other specialties, especially if one is competing with them for patients and cases, Dr. Siracuse said. Brandon Propper, MD, of San Antonio Military Medical Center, Texas, steps up to the plate with his “Top 10 Ways to Work With Other Specialties.”
The Practice Management Tips and Tricks for Young Vascular Surgeons session is recommended by the Community Practice Committee and the Young Surgeons Committee.
Friday, June 22
1:30 – 3:00 p.m.
HCC, Room 311
C5: Practice Management Tips and Tricks for Young Vascular Surgeons
Young vascular surgeons looking to make their marks and balance their lives, as well as more experienced surgeons seeking tips for more effective practice and career management, should consider the session led by Jeffrey Siracuse, MD, of Boston Medical Center, and Courtney Warner, MD, of Albany Medical College, Saratoga Springs, N.Y.
“This will be a great and informative session that, although geared toward young surgeons, will be highly useful for surgeons of all experience levels,” Dr. Siracuse said of the session.
“Topics include building a successful academic practice, building a successful private practice, how to work with other specialties, how to negotiate a contract, how to improve one’s current job, and more on the business side of medicine,” said Dr. Siracuse. The discussions will be followed by a Q&A panel.
The first job after a fellowship is often the first “real job” of any type that new surgeons have had, Dr. Siracuse said. Medical school offers many things, but young surgeons may be underprepared for how to negotiate for one’s first surgical position and how to set oneself up for success, he explained. That said, the keys for success are significantly different for surgeons entering an academic setting or private practice, he noted.
The session kicks off with presenters addressing both types of practice.
Faisal Aziz, MD, of Penn State Hershey College of Medicine, Hershey, addresses the “Top 10 Roadblocks to a Successful Academic Practice,” and Scott Berman, MD, of Carondelet Medical Group in Tucson, Ariz., takes on the “Top 10 Roadblocks to a Successful Private Practice.”
Some subjects likely to prompt lively discussion among seasoned veteran surgeons as well as novices include how to effectively negotiate and renegotiate contracts. The contracts presentation, “Top 10 Tips on Negotiating Contracts and Other Things I Learned in Business School,” is scheduled to be given by Bruce Perler, MD, of Johns Hopkins University in Baltimore, Md.
In addition, surgeons at all levels of experience can benefit from tips on how to work with individuals in other specialties, especially if one is competing with them for patients and cases, Dr. Siracuse said. Brandon Propper, MD, of San Antonio Military Medical Center, Texas, steps up to the plate with his “Top 10 Ways to Work With Other Specialties.”
The Practice Management Tips and Tricks for Young Vascular Surgeons session is recommended by the Community Practice Committee and the Young Surgeons Committee.
Friday, June 22
1:30 – 3:00 p.m.
HCC, Room 311
C5: Practice Management Tips and Tricks for Young Vascular Surgeons
Mix and Mingle at Friday’s Closing Reception
Mark the closing of the Exhibit Hall by attending the Closing Reception, set for 4:30 to 5:30 p.m. Friday in the Auditorium on Level 2 of the Hynes Convention Center.
VAM attendees have one more chance to visit with vendors and check out innovations in devices and medications. Guests have another to meet with friends old and new, to relax, and to enjoy cocktails and hors d’oeuvres.
Tickets are required and are available at Registration.
Mark the closing of the Exhibit Hall by attending the Closing Reception, set for 4:30 to 5:30 p.m. Friday in the Auditorium on Level 2 of the Hynes Convention Center.
VAM attendees have one more chance to visit with vendors and check out innovations in devices and medications. Guests have another to meet with friends old and new, to relax, and to enjoy cocktails and hors d’oeuvres.
Tickets are required and are available at Registration.
Mark the closing of the Exhibit Hall by attending the Closing Reception, set for 4:30 to 5:30 p.m. Friday in the Auditorium on Level 2 of the Hynes Convention Center.
VAM attendees have one more chance to visit with vendors and check out innovations in devices and medications. Guests have another to meet with friends old and new, to relax, and to enjoy cocktails and hors d’oeuvres.
Tickets are required and are available at Registration.
Advanced Practice Providers Vital to Vascular Team
The team approach has changed the entire field of medicine in the past 10-20 years and, in fact, is critical to optimal patient outcomes. That’s according to Anil Hingorani, MD, who will co-moderate a special forum Friday, “Improving Clinical Metrics With the Utilization of Advanced Practice Providers.”
It will be held from 1:30 to 3 p.m. in Ballroom A/B.
The team approach is front and center at this year’s Vascular Annual Meeting, which carries the theme: “Home of the Vascular Team – Partners in Patient Care.”
“Our vascular disease patients can be quite complex,” said Dr. Hingorani. “We will highlight that to take care of these complexities we need a team approach, and our team members can help tremendously.” This is true across the setting spectrum, be it rural, urban, suburban.
“Some NPs and PAs run our service. They help coordinate pre-op evaluations, post-op management, take care of research protocols, billing, and other office responsibilities,” he said.
He pointed out he is not a specialist in diabetes, but that his NP has a special interest and passion for the topic. The work she does for their diabetic patients “helps MY patients and helps MY procedures have better outcomes.”
PAs and NPs also help run research projects and are instrumental in working with fellows rotating through. With their work in what Dr. Hingorani referred to as the “three pillars” – clinical work, teaching, and research – they are tremendously important to the vascular team.
Advanced care providers also help improve outcomes, he said, when pay for performance and quantitating outcomes is becoming a standard part of health care. Admissions, discharges, surgical site infections, diabetes, follow-up all are important for patient care, and tracking all the details is vital to outcomes. It will be addressed in the forum, Dr. Hingorani said.
“Medicine is a specialty that hasn’t really caught on to MACRA, MIPS, and what pay for performance really entails,” he said. “Many are still figuring out, ‘What changes do I need to make to make this work for my patients and me? Where does my practice fit in?’ We’re going to have to keep working on that.”
Speakers will be primarily nurse practitioners and physician assistants. “We didn’t want surgeons telling PAs and NPs what they should be doing. It needs to be the PAs and NPs doing the speaking, focusing on issues important to them.”
Dr. Hingorani believes that the Vascular Annual Meeting is the first to stress the team approach theme. “I think it’s an important step and others will follow suit,” he said. “These ideas are resonating. They’re important and will be the way forward.
“I think we’ll be breaking new ground and will ripple across the societies.”
Besides a panel discussion at the end, forum topics include:
- Improving Metrics in Clinical Practice: The Value of APPs to a Vascular Practice
- There’s an APP for That: Workforce and Community Practice Experience
- National and International Trends in the Use of APPs, PAs in Surgery and Outcome Data
- Improving Metrics via Team-Based Care: The Wake Forest Baptist Health Experience
- Influence of APPs - MIPS and “Throughput” of Patients, Value/Quality/Financial Benefit and APPs
- Funny You Should Ask: What Advanced Practice Providers Bring to the Table
- How Advanced Practice Clinicians Can Add Value to Your Practice
- Driving Outcomes: University of Maryland Advanced Practice Providers Target Preventable Complications, Length of Stay, and Readmissions Univers LT Std
The team approach has changed the entire field of medicine in the past 10-20 years and, in fact, is critical to optimal patient outcomes. That’s according to Anil Hingorani, MD, who will co-moderate a special forum Friday, “Improving Clinical Metrics With the Utilization of Advanced Practice Providers.”
It will be held from 1:30 to 3 p.m. in Ballroom A/B.
The team approach is front and center at this year’s Vascular Annual Meeting, which carries the theme: “Home of the Vascular Team – Partners in Patient Care.”
“Our vascular disease patients can be quite complex,” said Dr. Hingorani. “We will highlight that to take care of these complexities we need a team approach, and our team members can help tremendously.” This is true across the setting spectrum, be it rural, urban, suburban.
“Some NPs and PAs run our service. They help coordinate pre-op evaluations, post-op management, take care of research protocols, billing, and other office responsibilities,” he said.
He pointed out he is not a specialist in diabetes, but that his NP has a special interest and passion for the topic. The work she does for their diabetic patients “helps MY patients and helps MY procedures have better outcomes.”
PAs and NPs also help run research projects and are instrumental in working with fellows rotating through. With their work in what Dr. Hingorani referred to as the “three pillars” – clinical work, teaching, and research – they are tremendously important to the vascular team.
Advanced care providers also help improve outcomes, he said, when pay for performance and quantitating outcomes is becoming a standard part of health care. Admissions, discharges, surgical site infections, diabetes, follow-up all are important for patient care, and tracking all the details is vital to outcomes. It will be addressed in the forum, Dr. Hingorani said.
“Medicine is a specialty that hasn’t really caught on to MACRA, MIPS, and what pay for performance really entails,” he said. “Many are still figuring out, ‘What changes do I need to make to make this work for my patients and me? Where does my practice fit in?’ We’re going to have to keep working on that.”
Speakers will be primarily nurse practitioners and physician assistants. “We didn’t want surgeons telling PAs and NPs what they should be doing. It needs to be the PAs and NPs doing the speaking, focusing on issues important to them.”
Dr. Hingorani believes that the Vascular Annual Meeting is the first to stress the team approach theme. “I think it’s an important step and others will follow suit,” he said. “These ideas are resonating. They’re important and will be the way forward.
“I think we’ll be breaking new ground and will ripple across the societies.”
Besides a panel discussion at the end, forum topics include:
- Improving Metrics in Clinical Practice: The Value of APPs to a Vascular Practice
- There’s an APP for That: Workforce and Community Practice Experience
- National and International Trends in the Use of APPs, PAs in Surgery and Outcome Data
- Improving Metrics via Team-Based Care: The Wake Forest Baptist Health Experience
- Influence of APPs - MIPS and “Throughput” of Patients, Value/Quality/Financial Benefit and APPs
- Funny You Should Ask: What Advanced Practice Providers Bring to the Table
- How Advanced Practice Clinicians Can Add Value to Your Practice
- Driving Outcomes: University of Maryland Advanced Practice Providers Target Preventable Complications, Length of Stay, and Readmissions Univers LT Std
The team approach has changed the entire field of medicine in the past 10-20 years and, in fact, is critical to optimal patient outcomes. That’s according to Anil Hingorani, MD, who will co-moderate a special forum Friday, “Improving Clinical Metrics With the Utilization of Advanced Practice Providers.”
It will be held from 1:30 to 3 p.m. in Ballroom A/B.
The team approach is front and center at this year’s Vascular Annual Meeting, which carries the theme: “Home of the Vascular Team – Partners in Patient Care.”
“Our vascular disease patients can be quite complex,” said Dr. Hingorani. “We will highlight that to take care of these complexities we need a team approach, and our team members can help tremendously.” This is true across the setting spectrum, be it rural, urban, suburban.
“Some NPs and PAs run our service. They help coordinate pre-op evaluations, post-op management, take care of research protocols, billing, and other office responsibilities,” he said.
He pointed out he is not a specialist in diabetes, but that his NP has a special interest and passion for the topic. The work she does for their diabetic patients “helps MY patients and helps MY procedures have better outcomes.”
PAs and NPs also help run research projects and are instrumental in working with fellows rotating through. With their work in what Dr. Hingorani referred to as the “three pillars” – clinical work, teaching, and research – they are tremendously important to the vascular team.
Advanced care providers also help improve outcomes, he said, when pay for performance and quantitating outcomes is becoming a standard part of health care. Admissions, discharges, surgical site infections, diabetes, follow-up all are important for patient care, and tracking all the details is vital to outcomes. It will be addressed in the forum, Dr. Hingorani said.
“Medicine is a specialty that hasn’t really caught on to MACRA, MIPS, and what pay for performance really entails,” he said. “Many are still figuring out, ‘What changes do I need to make to make this work for my patients and me? Where does my practice fit in?’ We’re going to have to keep working on that.”
Speakers will be primarily nurse practitioners and physician assistants. “We didn’t want surgeons telling PAs and NPs what they should be doing. It needs to be the PAs and NPs doing the speaking, focusing on issues important to them.”
Dr. Hingorani believes that the Vascular Annual Meeting is the first to stress the team approach theme. “I think it’s an important step and others will follow suit,” he said. “These ideas are resonating. They’re important and will be the way forward.
“I think we’ll be breaking new ground and will ripple across the societies.”
Besides a panel discussion at the end, forum topics include:
- Improving Metrics in Clinical Practice: The Value of APPs to a Vascular Practice
- There’s an APP for That: Workforce and Community Practice Experience
- National and International Trends in the Use of APPs, PAs in Surgery and Outcome Data
- Improving Metrics via Team-Based Care: The Wake Forest Baptist Health Experience
- Influence of APPs - MIPS and “Throughput” of Patients, Value/Quality/Financial Benefit and APPs
- Funny You Should Ask: What Advanced Practice Providers Bring to the Table
- How Advanced Practice Clinicians Can Add Value to Your Practice
- Driving Outcomes: University of Maryland Advanced Practice Providers Target Preventable Complications, Length of Stay, and Readmissions Univers LT Std
Dialectical behavior therapy reduces suicide attempts in adolescents
A form of behavioral therapy that focuses on enhancing emotion regulation, distress tolerance, and improving quality of life has shown promise in reducing self-harm and suicide attempts in adolescents, according to new research.
In a paper published in JAMA Psychiatry, researchers reported the outcomes of a randomized trial of dialectical behavior therapy (DBT) versus individual and group supportive therapy in 173 adolescents with a history of suicide attempts.
DBT, developed by Marsha Linehan, PhD, as a team-based intervention for chronically suicidal patients with borderline personality disorder, is aimed at getting patients to focus on changing their behaviors so that they are able to meet their long-term goals. The use of DBT with adults has been tied to low dropout rates, and has been effective at reducing suicide attempts and self-harm.
In the study, the DBT consisted of weekly individual psychotherapy, multifamily group skills training, youth and parent telephone coaching, and a weekly therapist team consultation. The control group took part in individual sessions, group therapy, as-needed parent sessions, and a weekly therapist team consultation.
Researchers saw a 70% lower rate of suicide attempts, 68% lower rate of nonsuicidal self-injury, and 67% lower rate of self-harm in the DBT group, compared with the control group at the end of the 6-month treatment course. However, at 12 months, the differences between the two groups were no longer statistically significant.
“This is the first adolescent RCT [randomized, controlled trial] to our knowledge to demonstrate that DBT is effective at decreasing suicide attempts,” Elizabeth A. McCauley, PhD, of the Seattle Children’s Research Institute, and her coauthors.
At 12 months, those figures were 51.2% and 32.2% respectively.
Significantly, more participants in the DBT group completed the treatment, compared with those in individual and group supportive therapy (75.6% vs. 55.2%), although this did not appear to be responsible for the difference in outcomes.
“Although results of pattern-mixture models found no evidence of an informative attrition mechanism, we cannot rule out the possibility that differential treatment exposure is a mechanism that leads to the DBT outcomes,” the authors wrote. “Stronger DBT treatment retention is, however, an important finding given prior research that found difficulties with treatment engagement, and adherence among suicidal and self-harming youths.”
Parents were involved in both treatments, but “DBT included greater family involvement,” Dr. McCauley and her coauthors wrote. “This difference may have contributed to both greater retention and treatment effects, particularly because stronger family components are associated with treatment benefits for adolescent self-harm.”
The authors said the fact that both groups improved after 12 months provided support for the individual and group supportive therapy in these patients.
“Our findings add to data supporting other promising treatment approaches, including cognitive-behavioral therapy, mentalization-based therapy, and family-based treatments,” they concluded
The study was supported by the National Institutes of Mental Health. Eight authors declared grant support from NIMH, and two authors declared other funding unrelated to the study.
SOURCE: McCauley EA et al. JAMA Psychiatry. 2018 Jun 20. doi:10.1001/jamapsychiatry.2018.1109.
A form of behavioral therapy that focuses on enhancing emotion regulation, distress tolerance, and improving quality of life has shown promise in reducing self-harm and suicide attempts in adolescents, according to new research.
In a paper published in JAMA Psychiatry, researchers reported the outcomes of a randomized trial of dialectical behavior therapy (DBT) versus individual and group supportive therapy in 173 adolescents with a history of suicide attempts.
DBT, developed by Marsha Linehan, PhD, as a team-based intervention for chronically suicidal patients with borderline personality disorder, is aimed at getting patients to focus on changing their behaviors so that they are able to meet their long-term goals. The use of DBT with adults has been tied to low dropout rates, and has been effective at reducing suicide attempts and self-harm.
In the study, the DBT consisted of weekly individual psychotherapy, multifamily group skills training, youth and parent telephone coaching, and a weekly therapist team consultation. The control group took part in individual sessions, group therapy, as-needed parent sessions, and a weekly therapist team consultation.
Researchers saw a 70% lower rate of suicide attempts, 68% lower rate of nonsuicidal self-injury, and 67% lower rate of self-harm in the DBT group, compared with the control group at the end of the 6-month treatment course. However, at 12 months, the differences between the two groups were no longer statistically significant.
“This is the first adolescent RCT [randomized, controlled trial] to our knowledge to demonstrate that DBT is effective at decreasing suicide attempts,” Elizabeth A. McCauley, PhD, of the Seattle Children’s Research Institute, and her coauthors.
At 12 months, those figures were 51.2% and 32.2% respectively.
Significantly, more participants in the DBT group completed the treatment, compared with those in individual and group supportive therapy (75.6% vs. 55.2%), although this did not appear to be responsible for the difference in outcomes.
“Although results of pattern-mixture models found no evidence of an informative attrition mechanism, we cannot rule out the possibility that differential treatment exposure is a mechanism that leads to the DBT outcomes,” the authors wrote. “Stronger DBT treatment retention is, however, an important finding given prior research that found difficulties with treatment engagement, and adherence among suicidal and self-harming youths.”
Parents were involved in both treatments, but “DBT included greater family involvement,” Dr. McCauley and her coauthors wrote. “This difference may have contributed to both greater retention and treatment effects, particularly because stronger family components are associated with treatment benefits for adolescent self-harm.”
The authors said the fact that both groups improved after 12 months provided support for the individual and group supportive therapy in these patients.
“Our findings add to data supporting other promising treatment approaches, including cognitive-behavioral therapy, mentalization-based therapy, and family-based treatments,” they concluded
The study was supported by the National Institutes of Mental Health. Eight authors declared grant support from NIMH, and two authors declared other funding unrelated to the study.
SOURCE: McCauley EA et al. JAMA Psychiatry. 2018 Jun 20. doi:10.1001/jamapsychiatry.2018.1109.
A form of behavioral therapy that focuses on enhancing emotion regulation, distress tolerance, and improving quality of life has shown promise in reducing self-harm and suicide attempts in adolescents, according to new research.
In a paper published in JAMA Psychiatry, researchers reported the outcomes of a randomized trial of dialectical behavior therapy (DBT) versus individual and group supportive therapy in 173 adolescents with a history of suicide attempts.
DBT, developed by Marsha Linehan, PhD, as a team-based intervention for chronically suicidal patients with borderline personality disorder, is aimed at getting patients to focus on changing their behaviors so that they are able to meet their long-term goals. The use of DBT with adults has been tied to low dropout rates, and has been effective at reducing suicide attempts and self-harm.
In the study, the DBT consisted of weekly individual psychotherapy, multifamily group skills training, youth and parent telephone coaching, and a weekly therapist team consultation. The control group took part in individual sessions, group therapy, as-needed parent sessions, and a weekly therapist team consultation.
Researchers saw a 70% lower rate of suicide attempts, 68% lower rate of nonsuicidal self-injury, and 67% lower rate of self-harm in the DBT group, compared with the control group at the end of the 6-month treatment course. However, at 12 months, the differences between the two groups were no longer statistically significant.
“This is the first adolescent RCT [randomized, controlled trial] to our knowledge to demonstrate that DBT is effective at decreasing suicide attempts,” Elizabeth A. McCauley, PhD, of the Seattle Children’s Research Institute, and her coauthors.
At 12 months, those figures were 51.2% and 32.2% respectively.
Significantly, more participants in the DBT group completed the treatment, compared with those in individual and group supportive therapy (75.6% vs. 55.2%), although this did not appear to be responsible for the difference in outcomes.
“Although results of pattern-mixture models found no evidence of an informative attrition mechanism, we cannot rule out the possibility that differential treatment exposure is a mechanism that leads to the DBT outcomes,” the authors wrote. “Stronger DBT treatment retention is, however, an important finding given prior research that found difficulties with treatment engagement, and adherence among suicidal and self-harming youths.”
Parents were involved in both treatments, but “DBT included greater family involvement,” Dr. McCauley and her coauthors wrote. “This difference may have contributed to both greater retention and treatment effects, particularly because stronger family components are associated with treatment benefits for adolescent self-harm.”
The authors said the fact that both groups improved after 12 months provided support for the individual and group supportive therapy in these patients.
“Our findings add to data supporting other promising treatment approaches, including cognitive-behavioral therapy, mentalization-based therapy, and family-based treatments,” they concluded
The study was supported by the National Institutes of Mental Health. Eight authors declared grant support from NIMH, and two authors declared other funding unrelated to the study.
SOURCE: McCauley EA et al. JAMA Psychiatry. 2018 Jun 20. doi:10.1001/jamapsychiatry.2018.1109.
FROM JAMA PSYCHIATRY
Key clinical point: Dialectical behavior therapy reduces suicide attempts and self-harm in adolescents.
Major finding: DBT showed a 70% reduction in suicide attempts, compared with controls.
Study details: A randomized, controlled study of 173 adolescents with a history of suicide attempts.
Disclosures: The study was supported by the National Institutes of Mental Health. Eight authors declared grant support from NIMH, and two authors declared other funding unrelated to the study.
Source: McCauley EA et al. JAMA Psychiatry. 2018 Jun 20. doi: 10.1001/jamapsychiatry.2018.1109.
Recurrence of a small gastric gastrointestinal stromal tumor with high mitotic index
Gastrointestinal stromal tumor (GIST) is the most common soft tissue sarcoma of the gastrointestinal tract, usually arising from the interstitial cells of Cajal or similar cells in the outer wall of the gastrointestinal tract.1,2 Most GISTs have an activating mutation in KIT or platelet-derived growth factor receptor alpha (PDGFRA). Tumor size, mitotic rate, and anatomic site are the most common pathological features used to risk stratify GIST tumors.3-10 It is important to note when using such risk calculators that preoperative imatinib before determining tumor characteristics (such as mitoses per 50 high-power fields [hpf]) often changes the relevant parameters so that the same risk calculations may not apply. Tumors with a mitotic rate ≤5 mitoses per 50 hpf and a size ≤5 cm in greatest dimension have a lower recurrence rate after resection than tumors with a mitotic rate >5 mitoses per 50 hpf and a size >10 cm, and larger tumors can have a recurrence rate of up to 86%.11,12 Findings from a large observational study have suggested that the prognosis of gastric GIST in Korea and Japan may be more favorable compared with that in Western countries.13
The primary treatment of a localized primary GIST is surgical excision, but a cure is limited by recurrence.14,15 Imatinib is useful in the treatment of metastatic or recurrent GIST, and adjuvant treatment with imatinib after surgery has been shown to improve progression-free and overall survival in some cases.3,16-18 Responses to adjuvant imatinib depend on tumor sensitivity to the drug and the risk of recurrence. Drug sensitivity is largely dependent on the presence of mutations in KIT or PDGFRA.3,18 Recurrence risk is highly dependent on tumor size, tumor site, tumor rupture, and mitotic index.1,3,5,6,8,9,18,19 Findings on the use of gene expression patterns to predict recurrence risk have also been reported.20-27 However, recurrence risk is poorly understood for categories in which there are few cases with known outcomes, such as very small gastric GIST with a high mitotic index. For example, few cases of gastric GIST have been reported with a tumor size ≤2 cm, a mitotic rate >5 mitoses per 50 hpf, and adequate clinical follow-up. In such cases, it is difficult to assess the risk of recurrence.6 We report here the long-term outcome of a patient with a 1.8 cm gastric GIST with a mitotic index of 36 mitoses per 50 hpf and a KIT exon 11 mutation.
Case presentation and summary
A 69-year-old man presented with periumbilical and epigastric pain of 6-month duration. His medical history was notable for hyperlipidemia, hypertension, coronary angioplasty, and spinal surgery. He had a 40 pack-year smoking history and consumed 2 to 4 alcoholic drinks per day. The results of a physical examination were unremarkable. A computedtomographic (CT) scan showed no abnormalities. An esophagoduodenoscopy (EGD) revealed gastric ulcers. He was treated successfully with omeprazole 20 mg by mouth daily.
A month later, a follow-up EGD revealed a 1.8 × 1.5 cm submucosal mass 3 cm from the gastroesophageal junction. The patient underwent a fundus wedge resection, and a submucosal mass 1.8 cm in greatest dimension was removed. Pathologic examination revealed a GIST, spindle cell type, with a mitotic rate of 36 mitoses per 50 hpf with negative margins. Immunohistochemistry was positive for CD117. An exon 11 deletion (KVV558-560NV) was present in KIT. The patient’s risk of recurrence was unclear, and his follow-up included CT scans of the abdomen and pelvis every 3 to 4 months for the first 2 years, then every 6 months for the next 2.5 years.
A CT scan about 3.5 years after primary resection revealed small nonspecific liver hypodensities that became more prominent during the next year. About 5 years after primary resection, magnetic resonance imaging (MRI) revealed several liver lesions, the largest of which measured at 1.3 cm in greatest dimension. The patient’s liver metastases were readily identified by MRI (Figure 1) and CT imaging (Figure 2A).
Discussion
Small gastric GISTs are sometimes found by endoscopy performed for unrelated reasons. Recent data suggest that the incidence of gastric GIST may be higher than previously thought. In a Japanese study of patients with gastric cancer in which 100 stomachs were systematically examined pathologically, 50 microscopic GISTs were found in 35 patients.28 Most small gastric GISTs have a low mitotic index. Few cases have been described with a high mitotic index. In a study of 1765 cases of GIST of the stomach, 8 patients had a tumor size less than 2 cm and a mitotic index greater than 5. Of those, only 6 patients had long-term follow-up, and 3 were alive without disease at 2, 17, and 20 years of follow-up.7 These limited data make it impossible to predict outcomes in patients with small gastric GIST with a high mitotic index.
For patients who are at high risk of recurrence after surgery, 3 years of adjuvant imatinib treatment compared with 1 year has been shown to improve overall survival and is the current standard of care.10,17 A study comparing 5 and 3 years of imatinib is ongoing to establish whether a longer period of adjuvant treatment is warranted. In patients with metastatic GIST, lifelong imatinib until lack of benefit is considered optimal treatment.10 All patients should undergo KIT mutation analysis. Those with the PDGFRA D842V mutation, SDH (succinate dehydrogenase) deficiency, or neurofibromatosis-related GIST should not receive adjuvant imatinib.
This case has several unusual features. The small tumor size with a very high mitotic rate is rare. Such cases have not been reported in large numbers and have therefore not been reliably incorporated into risk prediction algorithms. In addition, despite a high mitotic index, the tumor was not FDG avid on PET imaging. The diagnosis of GIST is strongly supported by the KIT mutation and response to imatinib. This particular KIT mutation in larger GISTs is associated with aggressive disease. The present case adds to the data on the biology of small gastric GISTs with a high mitotic index and suggests the mitotic index in these tumors may be a more important predictor than size.
Acknowledgment
The authors thank Michael Franklin, MS, for editorial assistance, and Sabrina Porter for media edits.
1. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal tumours: origin and molecular oncology. Nat Rev Cancer. 2011;11(12):865-878.
2. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279(5350):577-580.
3. Corless CL, Ballman KV, Antonescu CR, et al. Pathologic and molecular features correlate with long-term outcome after adjuvant therapy of resected primary GI stromal tumor: the ACOSOG Z9001 trial. J Clin Oncol. 2014;32(15):1563-1570.
4. Huang J, Zheng DL, Qin FS, et al. Genetic and epigenetic silencing of SCARA5 may contribute to human hepatocellular carcinoma by activating FAK signaling. J Clin Invest. 2010;120(1):223-241.
5. Joensuu H, Vehtari A, Riihimaki J, et al. Risk of recurrence of gastrointestinal stromal tumour after surgery: an analysis of pooled population-based cohorts. Lancet Oncol. 2012;13(3):265-274.
6. Miettinen M, Lasota J. Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med. 2006;130(10):1466-1478.
7. Miettinen M, Sobin LH, Lasota J. Gastrointestinal stromal tumors of the stomach: a clinicopathologic, immunohistochemical, and molecular genetic study of 1765 cases with long-term follow-up. Am J Surg Pathol. 2005;29(1):52-68.
8. Patel S. Navigating risk stratification systems for the management of patients with GIST. Ann Surg Oncol. 2011;18(6):1698-1704.
9. Rossi S, Miceli R, Messerini L, et al. Natural history of imatinib-naive GISTs: a retrospective analysis of 929 cases with long-term follow-up and development of a survival nomogram based on mitotic index and size as continuous variables. Am J Surg Pathol. 2011;35(11):1646-1656.
10. National Comprehensive Cancer Network. Sarcoma. https://www.nccn.org. Accessed March 27, 2018.
11. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: a consensus approach. Int J Surg Pathol. 2002;10(2):81-89.
12. Huang HY, Li CF, Huang WW, et al. A modification of NIH consensus criteria to better distinguish the highly lethal subset of primary localized gastrointestinal stromal tumors: a subdivision of the original high-risk group on the basis of outcome. Surgery. 2007;141(6):748-756.
13. Kim MC, Yook JH, Yang HK, et al. Long-term surgical outcome of 1057 gastric GISTs according to 7th UICC/AJCC TNM system: multicenter observational study From Korea and Japan. Medicine (Baltimore). 2015;94(41):e1526.
14. Casali PG, Blay JY; ESMO/CONTICANET/EUROBONET Consensus Panel of experts. Soft tissue sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v198-v203.
15. Joensuu H, DeMatteo RP. The management of gastrointestinal stromal tumors: a model for targeted and multidisciplinary therapy of malignancy. Annu Rev Med. 2012;63:247-258.
16. Dematteo RP, Ballman KV, Antonescu CR, et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;373(9669):1097-1104.
17. Joensuu H, Eriksson M, Sundby Hall K, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: a randomized trial. JAMA. 2012;307(12):1265-1272.
18. Joensuu H, Rutkowski P, Nishida T, et al. KIT and PDGFRA mutations and the risk of GI stromal tumor recurrence. J Clin Oncol. 2015;33(6):634-642.
19. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: A consensus approach. Hum Pathol. 2002;33(5):459-465.
20. Antonescu CR, Viale A, Sarran L, et al. Gene expression in gastrointestinal stromal tumors is distinguished by KIT genotype and anatomic site. Clin Cancer Res. 2004;10(10):3282-3290.
21. Arne G, Kristiansson E, Nerman O, et al. Expression profiling of GIST: CD133 is associated with KIT exon 11 mutations, gastric location and poor prognosis. Int J Cancer. 2011;129(5):1149-1161.
22. Bertucci F, Finetti P, Ostrowski J, et al. Genomic Grade Index predicts postoperative clinical outcome of GIST. Br J Cancer. 2012;107(8):1433-1441.
23. Koon N, Schneider-Stock R, Sarlomo-Rikala M, et al. Molecular targets for tumour progression in gastrointestinal stromal tumours. Gut. 2004;53(2):235-240.
24. Lagarde P, Perot G, Kauffmann A, et al. Mitotic checkpoints and chromosome instability are strong predictors of clinical outcome in gastrointestinal stromal tumors. Clin Cancer Res. 2012;18(3):826-838.
25. Skubitz KM, Geschwind K, Xu WW, Koopmeiners JS, Skubitz AP. Gene expression identifies heterogeneity of metastatic behavior among gastrointestinal stromal tumors. J Transl Med. 2016;14:51.
26. Yamaguchi U, Nakayama R, Honda K, et al. Distinct gene expression-defined classes of gastrointestinal stromal tumor. J Clin Oncol. 2008;26(25):4100-4108.
27. Ylipaa A, Hunt KK, Yang J, et al. Integrative genomic characterization and a genomic staging system for gastrointestinal stromal tumors. Cancer. 2011;117(2):380-389.
28. Kawanowa K, Sakuma Y, Sakurai S, et al. High incidence of microscopic gastrointestinal stromal tumors in the stomach. Hum Pathol. 2006;37(12):1527-1535.
Gastrointestinal stromal tumor (GIST) is the most common soft tissue sarcoma of the gastrointestinal tract, usually arising from the interstitial cells of Cajal or similar cells in the outer wall of the gastrointestinal tract.1,2 Most GISTs have an activating mutation in KIT or platelet-derived growth factor receptor alpha (PDGFRA). Tumor size, mitotic rate, and anatomic site are the most common pathological features used to risk stratify GIST tumors.3-10 It is important to note when using such risk calculators that preoperative imatinib before determining tumor characteristics (such as mitoses per 50 high-power fields [hpf]) often changes the relevant parameters so that the same risk calculations may not apply. Tumors with a mitotic rate ≤5 mitoses per 50 hpf and a size ≤5 cm in greatest dimension have a lower recurrence rate after resection than tumors with a mitotic rate >5 mitoses per 50 hpf and a size >10 cm, and larger tumors can have a recurrence rate of up to 86%.11,12 Findings from a large observational study have suggested that the prognosis of gastric GIST in Korea and Japan may be more favorable compared with that in Western countries.13
The primary treatment of a localized primary GIST is surgical excision, but a cure is limited by recurrence.14,15 Imatinib is useful in the treatment of metastatic or recurrent GIST, and adjuvant treatment with imatinib after surgery has been shown to improve progression-free and overall survival in some cases.3,16-18 Responses to adjuvant imatinib depend on tumor sensitivity to the drug and the risk of recurrence. Drug sensitivity is largely dependent on the presence of mutations in KIT or PDGFRA.3,18 Recurrence risk is highly dependent on tumor size, tumor site, tumor rupture, and mitotic index.1,3,5,6,8,9,18,19 Findings on the use of gene expression patterns to predict recurrence risk have also been reported.20-27 However, recurrence risk is poorly understood for categories in which there are few cases with known outcomes, such as very small gastric GIST with a high mitotic index. For example, few cases of gastric GIST have been reported with a tumor size ≤2 cm, a mitotic rate >5 mitoses per 50 hpf, and adequate clinical follow-up. In such cases, it is difficult to assess the risk of recurrence.6 We report here the long-term outcome of a patient with a 1.8 cm gastric GIST with a mitotic index of 36 mitoses per 50 hpf and a KIT exon 11 mutation.
Case presentation and summary
A 69-year-old man presented with periumbilical and epigastric pain of 6-month duration. His medical history was notable for hyperlipidemia, hypertension, coronary angioplasty, and spinal surgery. He had a 40 pack-year smoking history and consumed 2 to 4 alcoholic drinks per day. The results of a physical examination were unremarkable. A computedtomographic (CT) scan showed no abnormalities. An esophagoduodenoscopy (EGD) revealed gastric ulcers. He was treated successfully with omeprazole 20 mg by mouth daily.
A month later, a follow-up EGD revealed a 1.8 × 1.5 cm submucosal mass 3 cm from the gastroesophageal junction. The patient underwent a fundus wedge resection, and a submucosal mass 1.8 cm in greatest dimension was removed. Pathologic examination revealed a GIST, spindle cell type, with a mitotic rate of 36 mitoses per 50 hpf with negative margins. Immunohistochemistry was positive for CD117. An exon 11 deletion (KVV558-560NV) was present in KIT. The patient’s risk of recurrence was unclear, and his follow-up included CT scans of the abdomen and pelvis every 3 to 4 months for the first 2 years, then every 6 months for the next 2.5 years.
A CT scan about 3.5 years after primary resection revealed small nonspecific liver hypodensities that became more prominent during the next year. About 5 years after primary resection, magnetic resonance imaging (MRI) revealed several liver lesions, the largest of which measured at 1.3 cm in greatest dimension. The patient’s liver metastases were readily identified by MRI (Figure 1) and CT imaging (Figure 2A).
Discussion
Small gastric GISTs are sometimes found by endoscopy performed for unrelated reasons. Recent data suggest that the incidence of gastric GIST may be higher than previously thought. In a Japanese study of patients with gastric cancer in which 100 stomachs were systematically examined pathologically, 50 microscopic GISTs were found in 35 patients.28 Most small gastric GISTs have a low mitotic index. Few cases have been described with a high mitotic index. In a study of 1765 cases of GIST of the stomach, 8 patients had a tumor size less than 2 cm and a mitotic index greater than 5. Of those, only 6 patients had long-term follow-up, and 3 were alive without disease at 2, 17, and 20 years of follow-up.7 These limited data make it impossible to predict outcomes in patients with small gastric GIST with a high mitotic index.
For patients who are at high risk of recurrence after surgery, 3 years of adjuvant imatinib treatment compared with 1 year has been shown to improve overall survival and is the current standard of care.10,17 A study comparing 5 and 3 years of imatinib is ongoing to establish whether a longer period of adjuvant treatment is warranted. In patients with metastatic GIST, lifelong imatinib until lack of benefit is considered optimal treatment.10 All patients should undergo KIT mutation analysis. Those with the PDGFRA D842V mutation, SDH (succinate dehydrogenase) deficiency, or neurofibromatosis-related GIST should not receive adjuvant imatinib.
This case has several unusual features. The small tumor size with a very high mitotic rate is rare. Such cases have not been reported in large numbers and have therefore not been reliably incorporated into risk prediction algorithms. In addition, despite a high mitotic index, the tumor was not FDG avid on PET imaging. The diagnosis of GIST is strongly supported by the KIT mutation and response to imatinib. This particular KIT mutation in larger GISTs is associated with aggressive disease. The present case adds to the data on the biology of small gastric GISTs with a high mitotic index and suggests the mitotic index in these tumors may be a more important predictor than size.
Acknowledgment
The authors thank Michael Franklin, MS, for editorial assistance, and Sabrina Porter for media edits.
Gastrointestinal stromal tumor (GIST) is the most common soft tissue sarcoma of the gastrointestinal tract, usually arising from the interstitial cells of Cajal or similar cells in the outer wall of the gastrointestinal tract.1,2 Most GISTs have an activating mutation in KIT or platelet-derived growth factor receptor alpha (PDGFRA). Tumor size, mitotic rate, and anatomic site are the most common pathological features used to risk stratify GIST tumors.3-10 It is important to note when using such risk calculators that preoperative imatinib before determining tumor characteristics (such as mitoses per 50 high-power fields [hpf]) often changes the relevant parameters so that the same risk calculations may not apply. Tumors with a mitotic rate ≤5 mitoses per 50 hpf and a size ≤5 cm in greatest dimension have a lower recurrence rate after resection than tumors with a mitotic rate >5 mitoses per 50 hpf and a size >10 cm, and larger tumors can have a recurrence rate of up to 86%.11,12 Findings from a large observational study have suggested that the prognosis of gastric GIST in Korea and Japan may be more favorable compared with that in Western countries.13
The primary treatment of a localized primary GIST is surgical excision, but a cure is limited by recurrence.14,15 Imatinib is useful in the treatment of metastatic or recurrent GIST, and adjuvant treatment with imatinib after surgery has been shown to improve progression-free and overall survival in some cases.3,16-18 Responses to adjuvant imatinib depend on tumor sensitivity to the drug and the risk of recurrence. Drug sensitivity is largely dependent on the presence of mutations in KIT or PDGFRA.3,18 Recurrence risk is highly dependent on tumor size, tumor site, tumor rupture, and mitotic index.1,3,5,6,8,9,18,19 Findings on the use of gene expression patterns to predict recurrence risk have also been reported.20-27 However, recurrence risk is poorly understood for categories in which there are few cases with known outcomes, such as very small gastric GIST with a high mitotic index. For example, few cases of gastric GIST have been reported with a tumor size ≤2 cm, a mitotic rate >5 mitoses per 50 hpf, and adequate clinical follow-up. In such cases, it is difficult to assess the risk of recurrence.6 We report here the long-term outcome of a patient with a 1.8 cm gastric GIST with a mitotic index of 36 mitoses per 50 hpf and a KIT exon 11 mutation.
Case presentation and summary
A 69-year-old man presented with periumbilical and epigastric pain of 6-month duration. His medical history was notable for hyperlipidemia, hypertension, coronary angioplasty, and spinal surgery. He had a 40 pack-year smoking history and consumed 2 to 4 alcoholic drinks per day. The results of a physical examination were unremarkable. A computedtomographic (CT) scan showed no abnormalities. An esophagoduodenoscopy (EGD) revealed gastric ulcers. He was treated successfully with omeprazole 20 mg by mouth daily.
A month later, a follow-up EGD revealed a 1.8 × 1.5 cm submucosal mass 3 cm from the gastroesophageal junction. The patient underwent a fundus wedge resection, and a submucosal mass 1.8 cm in greatest dimension was removed. Pathologic examination revealed a GIST, spindle cell type, with a mitotic rate of 36 mitoses per 50 hpf with negative margins. Immunohistochemistry was positive for CD117. An exon 11 deletion (KVV558-560NV) was present in KIT. The patient’s risk of recurrence was unclear, and his follow-up included CT scans of the abdomen and pelvis every 3 to 4 months for the first 2 years, then every 6 months for the next 2.5 years.
A CT scan about 3.5 years after primary resection revealed small nonspecific liver hypodensities that became more prominent during the next year. About 5 years after primary resection, magnetic resonance imaging (MRI) revealed several liver lesions, the largest of which measured at 1.3 cm in greatest dimension. The patient’s liver metastases were readily identified by MRI (Figure 1) and CT imaging (Figure 2A).
Discussion
Small gastric GISTs are sometimes found by endoscopy performed for unrelated reasons. Recent data suggest that the incidence of gastric GIST may be higher than previously thought. In a Japanese study of patients with gastric cancer in which 100 stomachs were systematically examined pathologically, 50 microscopic GISTs were found in 35 patients.28 Most small gastric GISTs have a low mitotic index. Few cases have been described with a high mitotic index. In a study of 1765 cases of GIST of the stomach, 8 patients had a tumor size less than 2 cm and a mitotic index greater than 5. Of those, only 6 patients had long-term follow-up, and 3 were alive without disease at 2, 17, and 20 years of follow-up.7 These limited data make it impossible to predict outcomes in patients with small gastric GIST with a high mitotic index.
For patients who are at high risk of recurrence after surgery, 3 years of adjuvant imatinib treatment compared with 1 year has been shown to improve overall survival and is the current standard of care.10,17 A study comparing 5 and 3 years of imatinib is ongoing to establish whether a longer period of adjuvant treatment is warranted. In patients with metastatic GIST, lifelong imatinib until lack of benefit is considered optimal treatment.10 All patients should undergo KIT mutation analysis. Those with the PDGFRA D842V mutation, SDH (succinate dehydrogenase) deficiency, or neurofibromatosis-related GIST should not receive adjuvant imatinib.
This case has several unusual features. The small tumor size with a very high mitotic rate is rare. Such cases have not been reported in large numbers and have therefore not been reliably incorporated into risk prediction algorithms. In addition, despite a high mitotic index, the tumor was not FDG avid on PET imaging. The diagnosis of GIST is strongly supported by the KIT mutation and response to imatinib. This particular KIT mutation in larger GISTs is associated with aggressive disease. The present case adds to the data on the biology of small gastric GISTs with a high mitotic index and suggests the mitotic index in these tumors may be a more important predictor than size.
Acknowledgment
The authors thank Michael Franklin, MS, for editorial assistance, and Sabrina Porter for media edits.
1. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal tumours: origin and molecular oncology. Nat Rev Cancer. 2011;11(12):865-878.
2. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279(5350):577-580.
3. Corless CL, Ballman KV, Antonescu CR, et al. Pathologic and molecular features correlate with long-term outcome after adjuvant therapy of resected primary GI stromal tumor: the ACOSOG Z9001 trial. J Clin Oncol. 2014;32(15):1563-1570.
4. Huang J, Zheng DL, Qin FS, et al. Genetic and epigenetic silencing of SCARA5 may contribute to human hepatocellular carcinoma by activating FAK signaling. J Clin Invest. 2010;120(1):223-241.
5. Joensuu H, Vehtari A, Riihimaki J, et al. Risk of recurrence of gastrointestinal stromal tumour after surgery: an analysis of pooled population-based cohorts. Lancet Oncol. 2012;13(3):265-274.
6. Miettinen M, Lasota J. Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med. 2006;130(10):1466-1478.
7. Miettinen M, Sobin LH, Lasota J. Gastrointestinal stromal tumors of the stomach: a clinicopathologic, immunohistochemical, and molecular genetic study of 1765 cases with long-term follow-up. Am J Surg Pathol. 2005;29(1):52-68.
8. Patel S. Navigating risk stratification systems for the management of patients with GIST. Ann Surg Oncol. 2011;18(6):1698-1704.
9. Rossi S, Miceli R, Messerini L, et al. Natural history of imatinib-naive GISTs: a retrospective analysis of 929 cases with long-term follow-up and development of a survival nomogram based on mitotic index and size as continuous variables. Am J Surg Pathol. 2011;35(11):1646-1656.
10. National Comprehensive Cancer Network. Sarcoma. https://www.nccn.org. Accessed March 27, 2018.
11. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: a consensus approach. Int J Surg Pathol. 2002;10(2):81-89.
12. Huang HY, Li CF, Huang WW, et al. A modification of NIH consensus criteria to better distinguish the highly lethal subset of primary localized gastrointestinal stromal tumors: a subdivision of the original high-risk group on the basis of outcome. Surgery. 2007;141(6):748-756.
13. Kim MC, Yook JH, Yang HK, et al. Long-term surgical outcome of 1057 gastric GISTs according to 7th UICC/AJCC TNM system: multicenter observational study From Korea and Japan. Medicine (Baltimore). 2015;94(41):e1526.
14. Casali PG, Blay JY; ESMO/CONTICANET/EUROBONET Consensus Panel of experts. Soft tissue sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v198-v203.
15. Joensuu H, DeMatteo RP. The management of gastrointestinal stromal tumors: a model for targeted and multidisciplinary therapy of malignancy. Annu Rev Med. 2012;63:247-258.
16. Dematteo RP, Ballman KV, Antonescu CR, et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;373(9669):1097-1104.
17. Joensuu H, Eriksson M, Sundby Hall K, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: a randomized trial. JAMA. 2012;307(12):1265-1272.
18. Joensuu H, Rutkowski P, Nishida T, et al. KIT and PDGFRA mutations and the risk of GI stromal tumor recurrence. J Clin Oncol. 2015;33(6):634-642.
19. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: A consensus approach. Hum Pathol. 2002;33(5):459-465.
20. Antonescu CR, Viale A, Sarran L, et al. Gene expression in gastrointestinal stromal tumors is distinguished by KIT genotype and anatomic site. Clin Cancer Res. 2004;10(10):3282-3290.
21. Arne G, Kristiansson E, Nerman O, et al. Expression profiling of GIST: CD133 is associated with KIT exon 11 mutations, gastric location and poor prognosis. Int J Cancer. 2011;129(5):1149-1161.
22. Bertucci F, Finetti P, Ostrowski J, et al. Genomic Grade Index predicts postoperative clinical outcome of GIST. Br J Cancer. 2012;107(8):1433-1441.
23. Koon N, Schneider-Stock R, Sarlomo-Rikala M, et al. Molecular targets for tumour progression in gastrointestinal stromal tumours. Gut. 2004;53(2):235-240.
24. Lagarde P, Perot G, Kauffmann A, et al. Mitotic checkpoints and chromosome instability are strong predictors of clinical outcome in gastrointestinal stromal tumors. Clin Cancer Res. 2012;18(3):826-838.
25. Skubitz KM, Geschwind K, Xu WW, Koopmeiners JS, Skubitz AP. Gene expression identifies heterogeneity of metastatic behavior among gastrointestinal stromal tumors. J Transl Med. 2016;14:51.
26. Yamaguchi U, Nakayama R, Honda K, et al. Distinct gene expression-defined classes of gastrointestinal stromal tumor. J Clin Oncol. 2008;26(25):4100-4108.
27. Ylipaa A, Hunt KK, Yang J, et al. Integrative genomic characterization and a genomic staging system for gastrointestinal stromal tumors. Cancer. 2011;117(2):380-389.
28. Kawanowa K, Sakuma Y, Sakurai S, et al. High incidence of microscopic gastrointestinal stromal tumors in the stomach. Hum Pathol. 2006;37(12):1527-1535.
1. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal tumours: origin and molecular oncology. Nat Rev Cancer. 2011;11(12):865-878.
2. Hirota S, Isozaki K, Moriyama Y, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science. 1998;279(5350):577-580.
3. Corless CL, Ballman KV, Antonescu CR, et al. Pathologic and molecular features correlate with long-term outcome after adjuvant therapy of resected primary GI stromal tumor: the ACOSOG Z9001 trial. J Clin Oncol. 2014;32(15):1563-1570.
4. Huang J, Zheng DL, Qin FS, et al. Genetic and epigenetic silencing of SCARA5 may contribute to human hepatocellular carcinoma by activating FAK signaling. J Clin Invest. 2010;120(1):223-241.
5. Joensuu H, Vehtari A, Riihimaki J, et al. Risk of recurrence of gastrointestinal stromal tumour after surgery: an analysis of pooled population-based cohorts. Lancet Oncol. 2012;13(3):265-274.
6. Miettinen M, Lasota J. Gastrointestinal stromal tumors: review on morphology, molecular pathology, prognosis, and differential diagnosis. Arch Pathol Lab Med. 2006;130(10):1466-1478.
7. Miettinen M, Sobin LH, Lasota J. Gastrointestinal stromal tumors of the stomach: a clinicopathologic, immunohistochemical, and molecular genetic study of 1765 cases with long-term follow-up. Am J Surg Pathol. 2005;29(1):52-68.
8. Patel S. Navigating risk stratification systems for the management of patients with GIST. Ann Surg Oncol. 2011;18(6):1698-1704.
9. Rossi S, Miceli R, Messerini L, et al. Natural history of imatinib-naive GISTs: a retrospective analysis of 929 cases with long-term follow-up and development of a survival nomogram based on mitotic index and size as continuous variables. Am J Surg Pathol. 2011;35(11):1646-1656.
10. National Comprehensive Cancer Network. Sarcoma. https://www.nccn.org. Accessed March 27, 2018.
11. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: a consensus approach. Int J Surg Pathol. 2002;10(2):81-89.
12. Huang HY, Li CF, Huang WW, et al. A modification of NIH consensus criteria to better distinguish the highly lethal subset of primary localized gastrointestinal stromal tumors: a subdivision of the original high-risk group on the basis of outcome. Surgery. 2007;141(6):748-756.
13. Kim MC, Yook JH, Yang HK, et al. Long-term surgical outcome of 1057 gastric GISTs according to 7th UICC/AJCC TNM system: multicenter observational study From Korea and Japan. Medicine (Baltimore). 2015;94(41):e1526.
14. Casali PG, Blay JY; ESMO/CONTICANET/EUROBONET Consensus Panel of experts. Soft tissue sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2010;21(Suppl 5):v198-v203.
15. Joensuu H, DeMatteo RP. The management of gastrointestinal stromal tumors: a model for targeted and multidisciplinary therapy of malignancy. Annu Rev Med. 2012;63:247-258.
16. Dematteo RP, Ballman KV, Antonescu CR, et al. Adjuvant imatinib mesylate after resection of localised, primary gastrointestinal stromal tumour: a randomised, double-blind, placebo-controlled trial. Lancet. 2009;373(9669):1097-1104.
17. Joensuu H, Eriksson M, Sundby Hall K, et al. One vs three years of adjuvant imatinib for operable gastrointestinal stromal tumor: a randomized trial. JAMA. 2012;307(12):1265-1272.
18. Joensuu H, Rutkowski P, Nishida T, et al. KIT and PDGFRA mutations and the risk of GI stromal tumor recurrence. J Clin Oncol. 2015;33(6):634-642.
19. Fletcher CD, Berman JJ, Corless C, et al. Diagnosis of gastrointestinal stromal tumors: A consensus approach. Hum Pathol. 2002;33(5):459-465.
20. Antonescu CR, Viale A, Sarran L, et al. Gene expression in gastrointestinal stromal tumors is distinguished by KIT genotype and anatomic site. Clin Cancer Res. 2004;10(10):3282-3290.
21. Arne G, Kristiansson E, Nerman O, et al. Expression profiling of GIST: CD133 is associated with KIT exon 11 mutations, gastric location and poor prognosis. Int J Cancer. 2011;129(5):1149-1161.
22. Bertucci F, Finetti P, Ostrowski J, et al. Genomic Grade Index predicts postoperative clinical outcome of GIST. Br J Cancer. 2012;107(8):1433-1441.
23. Koon N, Schneider-Stock R, Sarlomo-Rikala M, et al. Molecular targets for tumour progression in gastrointestinal stromal tumours. Gut. 2004;53(2):235-240.
24. Lagarde P, Perot G, Kauffmann A, et al. Mitotic checkpoints and chromosome instability are strong predictors of clinical outcome in gastrointestinal stromal tumors. Clin Cancer Res. 2012;18(3):826-838.
25. Skubitz KM, Geschwind K, Xu WW, Koopmeiners JS, Skubitz AP. Gene expression identifies heterogeneity of metastatic behavior among gastrointestinal stromal tumors. J Transl Med. 2016;14:51.
26. Yamaguchi U, Nakayama R, Honda K, et al. Distinct gene expression-defined classes of gastrointestinal stromal tumor. J Clin Oncol. 2008;26(25):4100-4108.
27. Ylipaa A, Hunt KK, Yang J, et al. Integrative genomic characterization and a genomic staging system for gastrointestinal stromal tumors. Cancer. 2011;117(2):380-389.
28. Kawanowa K, Sakuma Y, Sakurai S, et al. High incidence of microscopic gastrointestinal stromal tumors in the stomach. Hum Pathol. 2006;37(12):1527-1535.
Striking rash in a patient with lung cancer on a checkpoint inhibitor
Lung cancer remains the most common cause of cancer death in the United States and worldwide.1 Despite advances in the treatment of the disease and development of targeted therapy, the 5-year overall survival in stage IV non–small-cell lung cancer remains poor, ranging from 6% to 10%.2 More recently, checkpoint inhibitors have had a major impact on the treatment of lung cancer. Nivolumab was the first program cell death protein-1 (PD-1) inhibitor approved for malignant melanoma.3 In July 2015, it was approved as a second-line treatment of squamous cell carcinoma of the lung.4 Since then, the use of nivolumab has extended to other malignancies such as head and neck cancer, renal cell carcinoma, and the list continues to expand. In lung cancer, it demonstrated superior overall survival of 9 months, compared with 6 months with docetaxel.4 Other checkpoint inhibitors such as pembrolizumab5 and atezolizumab6 were subsequently developed, and are also used in the treatment of lung cancer.
Serious potential autoimmune complications arise in up to 30% of patients treated with PD-1 inhibitors. Dermatologic toxicity is the most common immune-related adverse event in these patients. In addition to vitiligo, most common is a reticular maculopapular rash on the trunk and extremities. Other adverse events, such as photosensitivity, alopecia, xerosis, and hair color changes, are reported less frequently.7 We report here a case of rash at an unusual location (auricular and periauricular) with skin exfoliation mimicking other common skin conditions such as eczema and psoriasis.
Case presentation and summary
A 57-year-old woman with a history of cerebrovascular accident with residual left lower-leg paresis presented for acute onset expressive aphasia in the absence of other constitutional or neurological findings. Magnetic resonance imaging of the brain showed a posterior, left parietal lobe lesion of 1.6 cm with intralesional hemorrhage and surrounding edema suggestive of brain metastasis. The patient had a 35 pack-year history of smoking. A staging work-up with computed-tomographic (CT) scans showed a spiculated enhancing nodule in the superior segment of the right lower lobe plus mediastinal adenopathy.
The patient underwent a CT-guided core biopsy of the spiculated nodule, which was found to be consistent with adenocarcinoma of the lung. It was negative for EGFR mutation or ALK rearrangement. She received stereotactic radiosurgery to the left posterior parietal lesion, and after completion of radiation, was started on systemic chemotherapy with cisplatin plus pemetrexed for adenocarcinoma of the lung. She received 4 cycles of chemotherapy. Repeat imaging with a PET-CT showed interval increase of the mediastinal hypermetabolic lymphadenopathy with new hypermetabolic pretracheal lymph nodes and interval development of multiple liver metastases in the right and left lobes of the liver (Figure 1). She was started on second-line therapy with nivolumab at a dose of 240 mg every 2 weeks. The treatment was complicated initially by new onset grade 2 papular pruritic rash after cycle 2 of therapy. The rash involved the upper and lower extremities, sparing the palms, soles, trunk, abdomen, and the back. It resolved with treatment delay and topical steroids.
The patient resumed treatment with nivolumab after complete resolution of the rash. However, she developed grade 2 nephritis after cycle 5 with a creatinine level of 1.98 mg/dL (reference range, 0.6-1.2 mg/ dL). This was resolved after treatment with oral prednisone, at a starting dose of 1 mg/kg and tapered over 4 weeks. PET CT scans obtained after cycles 5 and 11 showed no metabolic activity in the mediastinum or the liver and markedly decreased uptake in the right lower lobe nodule, down to an SUV of 1.7 with no new nodules. An MRI of the brain was stable (Figure 2).
After cycle 16 of nivolumab, the patient developed a severe eczematous rash with excoriations at the base of both ears involving the periauricular and auricular areas bilaterally (Figure 3).
She completed 4 weeks of steroid therapy on a tapering schedule. Treatment with nivolumab was resumed afterward with no adverse autoimmune complications. At her last visit (25 months after initiating a PD-1 inhibitor), there was no clinical or radiologic evidence of lung cancer nor any of autoimmune adverse effects.
Discussion
Among multiple autoimmune complications, dermatologic toxicity is the most common immune-related adverse event, occuring in about 30% to 40% of patients7,8 and with an average onset of 3-4 weeks after initiating treatment with checkpoint inhibitors.9 In addition to vitiligo, the most common type of rash described is a reticular maculopapular rash on the trunk and extremities.10 Other findings, such as photosensitivity, alopecia, xerosis, and hair color changes, have been reported in smaller numbers. Skin exfoliation, as seen in the present case, has been reported in fewer than 1% of the cases.4 Perivascular lymphocytic infiltrates extending deep into the dermis are most likely to be seen if the lesions are biopsied. Both the location of the rash in our patient and its relapsing nature are rare and make it more interesting as it presents a diagnostic dilemma for treating physicians. Ear, nose, and throat surgeons are more likely to encounter such a complication with the expanded use of PD-1 and PD-ligand 1 inhibitors in advanced head and neck cancers. The differential diagnosis includes localized eczema, psoriatic rash, skin infection, or an autoimmune phenomenon.
The location of the rash was also of concern because there have been reports of autoimmune inner-ear disease related to immunotherapy.11 After the failure of treatment with empiric antibiotics and topical steroids, in addition to the development of a new rash on her abdomen, we concluded that this case might represent an unusual autoimmune skin complication. The resolution of the skin lesions in both locations (the ears and the abdomen) with the oral steroid therapy, supported our suspected diagnosis of autoimmune dermatitis.
It is essential that these complications are detected early and misdiagnosis is avoided because timely treatment with steroids will prevent progression to more severe problems such as Steven-Johnson syndrome, toxic epidermal necrolysis,12 or extension into the inner ear.11This case is part of a growing spectrum of other unusual cases seen with immunotherapy treatment, such as erythema nodosum-like reactions,13 bullous dermatitis,14 and psoriasiform eruptions.15 It highlights the need for an awareness of expanding dermatologic complications from immunotherapy beyond the reported common manifestations. Established guidelines and algorithms for the management of immune-related dermatologic toxicity are available to assist the physician in treatment (Table 1).16 Skin biopsy should be considered if the diagnosis remains uncertain, although starting empiric treatment with steroids is a widely acceptable approach. Reassessing the skin rash in 48 hours to 1 week after treatment initiation is crucial because steroid-refractory cases will need additional immunosuppression. Early termination of steroids is associated with higher recurrence rate, therefore tapering steroids over 4 weeks is highly recommended before resuming treatment with checkpoint inhibitors.
In summary, increased awareness among health care professionals of the common and unusual complications of immunotherapy agents is important and essential in patient care. In addition to oncologists, head and neck surgeons, pulmonologists, urologists, dermatologists, and general internists will encounter patients with immunotherapy-related complications. Patient education should be emphasized to ensure prompt investigation and treatment of complications. Finally, it is not yet clear whether the development of autoimmune reactions predicts disease response to treatment. In a series of 134 patients with lung cancer, the occurrence of autoimmune adverse events correlated with improved survival.17 More research is needed to identify prognostic and predictive biomarkers for response to immunotherapy.
Conclusion
This pattern of autoimmune dermatitis localizing to the ears is rare (<1% of cases of dermatitis). Nevertheless, it raises the awareness for dermatologic complications of immunotherapy beyond the classical reported manifestations. Prompt diagnosis and treatment is essential to avoid serious complications such as Steven-Johnson syndrome, toxic epidermal necrolysis, and potentially damage to the inner ear.
1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87-108.
2. Goldstraw P, Chansky K, Crowley J, et al. The IASLC Lung Cancer Staging Project: proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11:39-51.
3. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
4. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123-135.
5. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemo-therapy for PD- L1- positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823- 1833.
6. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255-265.
7. Collins LK, Chapman MS, Carter JB, Samie FH. Cutaneous adverse events of the immune checkpoint inhibitors. Curr Prob Cancer. 2017;41:125-128.
8. Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26(12):2375.
9. Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691-2697.
10. Belum VR, Benhuri B, Postow MA, et al. Characterisation and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer. 2016;60:12-25.
11. Zibelman M, Pollak N, Olszanski AJ. Autoimmune inner ear disease in a melanoma patient treated with pembrolizumab. J Immunother Cancer. 2016;4:8.
12. Nayar N, Briscoe K, Penas PF. Toxic epidermal necrolysis-like reaction with severe satellite cell necrosis associated with nivolumab in a patient with ipilimumab refractory metastatic melanoma. J Immunother. 2016;39(3):149-152.
13. Tetzlaff MT, Jazaeri AA, Torres-Cabala CA, et al. Erythema nodosum-like panniculitis mimicking disease recurrence: a novel toxicity from immune checkpoint blockade therapy - report of 2 patients. J Cutan Pathol. 2017;44(12):1080-1086.
14. Naidoo J, Schindler K, Querfeld C, et al. Autoimmune bullous skin disorders with immune checkpoint inhibitors targeting PD-1 and PD-L1. Cancer Immunol Res. 2016;4(5):383-389.
15. Ohtsuka M, Miura T, Mori T, Ishikawa M, Yamamoto T. Occurrence of psoriasiform eruption during nivolumab therapy for primary oral mucosal melanoma. JAMA Dermatol. 2015;151(7):797-799.
16. Haanen JBAG, Carbonnel F, Robert C, et al; ESMO Guidelines Committee. Management of toxicities from immunotherapy: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(suppl 4):iv119-iv142.
17. Haratani K, Hayashi H, Chiba Y, et al. Association of immune-related adverse events with nivolumab efficacy in non-small-cell lung cancer. JAMA Oncol. 2018;4(3):374-378.
Lung cancer remains the most common cause of cancer death in the United States and worldwide.1 Despite advances in the treatment of the disease and development of targeted therapy, the 5-year overall survival in stage IV non–small-cell lung cancer remains poor, ranging from 6% to 10%.2 More recently, checkpoint inhibitors have had a major impact on the treatment of lung cancer. Nivolumab was the first program cell death protein-1 (PD-1) inhibitor approved for malignant melanoma.3 In July 2015, it was approved as a second-line treatment of squamous cell carcinoma of the lung.4 Since then, the use of nivolumab has extended to other malignancies such as head and neck cancer, renal cell carcinoma, and the list continues to expand. In lung cancer, it demonstrated superior overall survival of 9 months, compared with 6 months with docetaxel.4 Other checkpoint inhibitors such as pembrolizumab5 and atezolizumab6 were subsequently developed, and are also used in the treatment of lung cancer.
Serious potential autoimmune complications arise in up to 30% of patients treated with PD-1 inhibitors. Dermatologic toxicity is the most common immune-related adverse event in these patients. In addition to vitiligo, most common is a reticular maculopapular rash on the trunk and extremities. Other adverse events, such as photosensitivity, alopecia, xerosis, and hair color changes, are reported less frequently.7 We report here a case of rash at an unusual location (auricular and periauricular) with skin exfoliation mimicking other common skin conditions such as eczema and psoriasis.
Case presentation and summary
A 57-year-old woman with a history of cerebrovascular accident with residual left lower-leg paresis presented for acute onset expressive aphasia in the absence of other constitutional or neurological findings. Magnetic resonance imaging of the brain showed a posterior, left parietal lobe lesion of 1.6 cm with intralesional hemorrhage and surrounding edema suggestive of brain metastasis. The patient had a 35 pack-year history of smoking. A staging work-up with computed-tomographic (CT) scans showed a spiculated enhancing nodule in the superior segment of the right lower lobe plus mediastinal adenopathy.
The patient underwent a CT-guided core biopsy of the spiculated nodule, which was found to be consistent with adenocarcinoma of the lung. It was negative for EGFR mutation or ALK rearrangement. She received stereotactic radiosurgery to the left posterior parietal lesion, and after completion of radiation, was started on systemic chemotherapy with cisplatin plus pemetrexed for adenocarcinoma of the lung. She received 4 cycles of chemotherapy. Repeat imaging with a PET-CT showed interval increase of the mediastinal hypermetabolic lymphadenopathy with new hypermetabolic pretracheal lymph nodes and interval development of multiple liver metastases in the right and left lobes of the liver (Figure 1). She was started on second-line therapy with nivolumab at a dose of 240 mg every 2 weeks. The treatment was complicated initially by new onset grade 2 papular pruritic rash after cycle 2 of therapy. The rash involved the upper and lower extremities, sparing the palms, soles, trunk, abdomen, and the back. It resolved with treatment delay and topical steroids.
The patient resumed treatment with nivolumab after complete resolution of the rash. However, she developed grade 2 nephritis after cycle 5 with a creatinine level of 1.98 mg/dL (reference range, 0.6-1.2 mg/ dL). This was resolved after treatment with oral prednisone, at a starting dose of 1 mg/kg and tapered over 4 weeks. PET CT scans obtained after cycles 5 and 11 showed no metabolic activity in the mediastinum or the liver and markedly decreased uptake in the right lower lobe nodule, down to an SUV of 1.7 with no new nodules. An MRI of the brain was stable (Figure 2).
After cycle 16 of nivolumab, the patient developed a severe eczematous rash with excoriations at the base of both ears involving the periauricular and auricular areas bilaterally (Figure 3).
She completed 4 weeks of steroid therapy on a tapering schedule. Treatment with nivolumab was resumed afterward with no adverse autoimmune complications. At her last visit (25 months after initiating a PD-1 inhibitor), there was no clinical or radiologic evidence of lung cancer nor any of autoimmune adverse effects.
Discussion
Among multiple autoimmune complications, dermatologic toxicity is the most common immune-related adverse event, occuring in about 30% to 40% of patients7,8 and with an average onset of 3-4 weeks after initiating treatment with checkpoint inhibitors.9 In addition to vitiligo, the most common type of rash described is a reticular maculopapular rash on the trunk and extremities.10 Other findings, such as photosensitivity, alopecia, xerosis, and hair color changes, have been reported in smaller numbers. Skin exfoliation, as seen in the present case, has been reported in fewer than 1% of the cases.4 Perivascular lymphocytic infiltrates extending deep into the dermis are most likely to be seen if the lesions are biopsied. Both the location of the rash in our patient and its relapsing nature are rare and make it more interesting as it presents a diagnostic dilemma for treating physicians. Ear, nose, and throat surgeons are more likely to encounter such a complication with the expanded use of PD-1 and PD-ligand 1 inhibitors in advanced head and neck cancers. The differential diagnosis includes localized eczema, psoriatic rash, skin infection, or an autoimmune phenomenon.
The location of the rash was also of concern because there have been reports of autoimmune inner-ear disease related to immunotherapy.11 After the failure of treatment with empiric antibiotics and topical steroids, in addition to the development of a new rash on her abdomen, we concluded that this case might represent an unusual autoimmune skin complication. The resolution of the skin lesions in both locations (the ears and the abdomen) with the oral steroid therapy, supported our suspected diagnosis of autoimmune dermatitis.
It is essential that these complications are detected early and misdiagnosis is avoided because timely treatment with steroids will prevent progression to more severe problems such as Steven-Johnson syndrome, toxic epidermal necrolysis,12 or extension into the inner ear.11This case is part of a growing spectrum of other unusual cases seen with immunotherapy treatment, such as erythema nodosum-like reactions,13 bullous dermatitis,14 and psoriasiform eruptions.15 It highlights the need for an awareness of expanding dermatologic complications from immunotherapy beyond the reported common manifestations. Established guidelines and algorithms for the management of immune-related dermatologic toxicity are available to assist the physician in treatment (Table 1).16 Skin biopsy should be considered if the diagnosis remains uncertain, although starting empiric treatment with steroids is a widely acceptable approach. Reassessing the skin rash in 48 hours to 1 week after treatment initiation is crucial because steroid-refractory cases will need additional immunosuppression. Early termination of steroids is associated with higher recurrence rate, therefore tapering steroids over 4 weeks is highly recommended before resuming treatment with checkpoint inhibitors.
In summary, increased awareness among health care professionals of the common and unusual complications of immunotherapy agents is important and essential in patient care. In addition to oncologists, head and neck surgeons, pulmonologists, urologists, dermatologists, and general internists will encounter patients with immunotherapy-related complications. Patient education should be emphasized to ensure prompt investigation and treatment of complications. Finally, it is not yet clear whether the development of autoimmune reactions predicts disease response to treatment. In a series of 134 patients with lung cancer, the occurrence of autoimmune adverse events correlated with improved survival.17 More research is needed to identify prognostic and predictive biomarkers for response to immunotherapy.
Conclusion
This pattern of autoimmune dermatitis localizing to the ears is rare (<1% of cases of dermatitis). Nevertheless, it raises the awareness for dermatologic complications of immunotherapy beyond the classical reported manifestations. Prompt diagnosis and treatment is essential to avoid serious complications such as Steven-Johnson syndrome, toxic epidermal necrolysis, and potentially damage to the inner ear.
Lung cancer remains the most common cause of cancer death in the United States and worldwide.1 Despite advances in the treatment of the disease and development of targeted therapy, the 5-year overall survival in stage IV non–small-cell lung cancer remains poor, ranging from 6% to 10%.2 More recently, checkpoint inhibitors have had a major impact on the treatment of lung cancer. Nivolumab was the first program cell death protein-1 (PD-1) inhibitor approved for malignant melanoma.3 In July 2015, it was approved as a second-line treatment of squamous cell carcinoma of the lung.4 Since then, the use of nivolumab has extended to other malignancies such as head and neck cancer, renal cell carcinoma, and the list continues to expand. In lung cancer, it demonstrated superior overall survival of 9 months, compared with 6 months with docetaxel.4 Other checkpoint inhibitors such as pembrolizumab5 and atezolizumab6 were subsequently developed, and are also used in the treatment of lung cancer.
Serious potential autoimmune complications arise in up to 30% of patients treated with PD-1 inhibitors. Dermatologic toxicity is the most common immune-related adverse event in these patients. In addition to vitiligo, most common is a reticular maculopapular rash on the trunk and extremities. Other adverse events, such as photosensitivity, alopecia, xerosis, and hair color changes, are reported less frequently.7 We report here a case of rash at an unusual location (auricular and periauricular) with skin exfoliation mimicking other common skin conditions such as eczema and psoriasis.
Case presentation and summary
A 57-year-old woman with a history of cerebrovascular accident with residual left lower-leg paresis presented for acute onset expressive aphasia in the absence of other constitutional or neurological findings. Magnetic resonance imaging of the brain showed a posterior, left parietal lobe lesion of 1.6 cm with intralesional hemorrhage and surrounding edema suggestive of brain metastasis. The patient had a 35 pack-year history of smoking. A staging work-up with computed-tomographic (CT) scans showed a spiculated enhancing nodule in the superior segment of the right lower lobe plus mediastinal adenopathy.
The patient underwent a CT-guided core biopsy of the spiculated nodule, which was found to be consistent with adenocarcinoma of the lung. It was negative for EGFR mutation or ALK rearrangement. She received stereotactic radiosurgery to the left posterior parietal lesion, and after completion of radiation, was started on systemic chemotherapy with cisplatin plus pemetrexed for adenocarcinoma of the lung. She received 4 cycles of chemotherapy. Repeat imaging with a PET-CT showed interval increase of the mediastinal hypermetabolic lymphadenopathy with new hypermetabolic pretracheal lymph nodes and interval development of multiple liver metastases in the right and left lobes of the liver (Figure 1). She was started on second-line therapy with nivolumab at a dose of 240 mg every 2 weeks. The treatment was complicated initially by new onset grade 2 papular pruritic rash after cycle 2 of therapy. The rash involved the upper and lower extremities, sparing the palms, soles, trunk, abdomen, and the back. It resolved with treatment delay and topical steroids.
The patient resumed treatment with nivolumab after complete resolution of the rash. However, she developed grade 2 nephritis after cycle 5 with a creatinine level of 1.98 mg/dL (reference range, 0.6-1.2 mg/ dL). This was resolved after treatment with oral prednisone, at a starting dose of 1 mg/kg and tapered over 4 weeks. PET CT scans obtained after cycles 5 and 11 showed no metabolic activity in the mediastinum or the liver and markedly decreased uptake in the right lower lobe nodule, down to an SUV of 1.7 with no new nodules. An MRI of the brain was stable (Figure 2).
After cycle 16 of nivolumab, the patient developed a severe eczematous rash with excoriations at the base of both ears involving the periauricular and auricular areas bilaterally (Figure 3).
She completed 4 weeks of steroid therapy on a tapering schedule. Treatment with nivolumab was resumed afterward with no adverse autoimmune complications. At her last visit (25 months after initiating a PD-1 inhibitor), there was no clinical or radiologic evidence of lung cancer nor any of autoimmune adverse effects.
Discussion
Among multiple autoimmune complications, dermatologic toxicity is the most common immune-related adverse event, occuring in about 30% to 40% of patients7,8 and with an average onset of 3-4 weeks after initiating treatment with checkpoint inhibitors.9 In addition to vitiligo, the most common type of rash described is a reticular maculopapular rash on the trunk and extremities.10 Other findings, such as photosensitivity, alopecia, xerosis, and hair color changes, have been reported in smaller numbers. Skin exfoliation, as seen in the present case, has been reported in fewer than 1% of the cases.4 Perivascular lymphocytic infiltrates extending deep into the dermis are most likely to be seen if the lesions are biopsied. Both the location of the rash in our patient and its relapsing nature are rare and make it more interesting as it presents a diagnostic dilemma for treating physicians. Ear, nose, and throat surgeons are more likely to encounter such a complication with the expanded use of PD-1 and PD-ligand 1 inhibitors in advanced head and neck cancers. The differential diagnosis includes localized eczema, psoriatic rash, skin infection, or an autoimmune phenomenon.
The location of the rash was also of concern because there have been reports of autoimmune inner-ear disease related to immunotherapy.11 After the failure of treatment with empiric antibiotics and topical steroids, in addition to the development of a new rash on her abdomen, we concluded that this case might represent an unusual autoimmune skin complication. The resolution of the skin lesions in both locations (the ears and the abdomen) with the oral steroid therapy, supported our suspected diagnosis of autoimmune dermatitis.
It is essential that these complications are detected early and misdiagnosis is avoided because timely treatment with steroids will prevent progression to more severe problems such as Steven-Johnson syndrome, toxic epidermal necrolysis,12 or extension into the inner ear.11This case is part of a growing spectrum of other unusual cases seen with immunotherapy treatment, such as erythema nodosum-like reactions,13 bullous dermatitis,14 and psoriasiform eruptions.15 It highlights the need for an awareness of expanding dermatologic complications from immunotherapy beyond the reported common manifestations. Established guidelines and algorithms for the management of immune-related dermatologic toxicity are available to assist the physician in treatment (Table 1).16 Skin biopsy should be considered if the diagnosis remains uncertain, although starting empiric treatment with steroids is a widely acceptable approach. Reassessing the skin rash in 48 hours to 1 week after treatment initiation is crucial because steroid-refractory cases will need additional immunosuppression. Early termination of steroids is associated with higher recurrence rate, therefore tapering steroids over 4 weeks is highly recommended before resuming treatment with checkpoint inhibitors.
In summary, increased awareness among health care professionals of the common and unusual complications of immunotherapy agents is important and essential in patient care. In addition to oncologists, head and neck surgeons, pulmonologists, urologists, dermatologists, and general internists will encounter patients with immunotherapy-related complications. Patient education should be emphasized to ensure prompt investigation and treatment of complications. Finally, it is not yet clear whether the development of autoimmune reactions predicts disease response to treatment. In a series of 134 patients with lung cancer, the occurrence of autoimmune adverse events correlated with improved survival.17 More research is needed to identify prognostic and predictive biomarkers for response to immunotherapy.
Conclusion
This pattern of autoimmune dermatitis localizing to the ears is rare (<1% of cases of dermatitis). Nevertheless, it raises the awareness for dermatologic complications of immunotherapy beyond the classical reported manifestations. Prompt diagnosis and treatment is essential to avoid serious complications such as Steven-Johnson syndrome, toxic epidermal necrolysis, and potentially damage to the inner ear.
1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87-108.
2. Goldstraw P, Chansky K, Crowley J, et al. The IASLC Lung Cancer Staging Project: proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11:39-51.
3. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
4. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123-135.
5. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemo-therapy for PD- L1- positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823- 1833.
6. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255-265.
7. Collins LK, Chapman MS, Carter JB, Samie FH. Cutaneous adverse events of the immune checkpoint inhibitors. Curr Prob Cancer. 2017;41:125-128.
8. Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26(12):2375.
9. Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691-2697.
10. Belum VR, Benhuri B, Postow MA, et al. Characterisation and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer. 2016;60:12-25.
11. Zibelman M, Pollak N, Olszanski AJ. Autoimmune inner ear disease in a melanoma patient treated with pembrolizumab. J Immunother Cancer. 2016;4:8.
12. Nayar N, Briscoe K, Penas PF. Toxic epidermal necrolysis-like reaction with severe satellite cell necrosis associated with nivolumab in a patient with ipilimumab refractory metastatic melanoma. J Immunother. 2016;39(3):149-152.
13. Tetzlaff MT, Jazaeri AA, Torres-Cabala CA, et al. Erythema nodosum-like panniculitis mimicking disease recurrence: a novel toxicity from immune checkpoint blockade therapy - report of 2 patients. J Cutan Pathol. 2017;44(12):1080-1086.
14. Naidoo J, Schindler K, Querfeld C, et al. Autoimmune bullous skin disorders with immune checkpoint inhibitors targeting PD-1 and PD-L1. Cancer Immunol Res. 2016;4(5):383-389.
15. Ohtsuka M, Miura T, Mori T, Ishikawa M, Yamamoto T. Occurrence of psoriasiform eruption during nivolumab therapy for primary oral mucosal melanoma. JAMA Dermatol. 2015;151(7):797-799.
16. Haanen JBAG, Carbonnel F, Robert C, et al; ESMO Guidelines Committee. Management of toxicities from immunotherapy: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(suppl 4):iv119-iv142.
17. Haratani K, Hayashi H, Chiba Y, et al. Association of immune-related adverse events with nivolumab efficacy in non-small-cell lung cancer. JAMA Oncol. 2018;4(3):374-378.
1.Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin. 2015;65(2):87-108.
2. Goldstraw P, Chansky K, Crowley J, et al. The IASLC Lung Cancer Staging Project: proposals for revision of the TNM stage groupings in the forthcoming (Eighth) edition of the TNM classification for lung cancer. J Thorac Oncol. 2016;11:39-51.
3. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
4. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;373:123-135.
5. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemo-therapy for PD- L1- positive non-small-cell lung cancer. N Engl J Med. 2016;375:1823- 1833.
6. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in patients with previously treated non-small-cell lung cancer (OAK): a phase 3, open-label, multicentre randomised controlled trial. Lancet. 2017;389:255-265.
7. Collins LK, Chapman MS, Carter JB, Samie FH. Cutaneous adverse events of the immune checkpoint inhibitors. Curr Prob Cancer. 2017;41:125-128.
8. Naidoo J, Page DB, Li BT, et al. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol. 2015;26(12):2375.
9. Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691-2697.
10. Belum VR, Benhuri B, Postow MA, et al. Characterisation and management of dermatologic adverse events to agents targeting the PD-1 receptor. Eur J Cancer. 2016;60:12-25.
11. Zibelman M, Pollak N, Olszanski AJ. Autoimmune inner ear disease in a melanoma patient treated with pembrolizumab. J Immunother Cancer. 2016;4:8.
12. Nayar N, Briscoe K, Penas PF. Toxic epidermal necrolysis-like reaction with severe satellite cell necrosis associated with nivolumab in a patient with ipilimumab refractory metastatic melanoma. J Immunother. 2016;39(3):149-152.
13. Tetzlaff MT, Jazaeri AA, Torres-Cabala CA, et al. Erythema nodosum-like panniculitis mimicking disease recurrence: a novel toxicity from immune checkpoint blockade therapy - report of 2 patients. J Cutan Pathol. 2017;44(12):1080-1086.
14. Naidoo J, Schindler K, Querfeld C, et al. Autoimmune bullous skin disorders with immune checkpoint inhibitors targeting PD-1 and PD-L1. Cancer Immunol Res. 2016;4(5):383-389.
15. Ohtsuka M, Miura T, Mori T, Ishikawa M, Yamamoto T. Occurrence of psoriasiform eruption during nivolumab therapy for primary oral mucosal melanoma. JAMA Dermatol. 2015;151(7):797-799.
16. Haanen JBAG, Carbonnel F, Robert C, et al; ESMO Guidelines Committee. Management of toxicities from immunotherapy: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(suppl 4):iv119-iv142.
17. Haratani K, Hayashi H, Chiba Y, et al. Association of immune-related adverse events with nivolumab efficacy in non-small-cell lung cancer. JAMA Oncol. 2018;4(3):374-378.
Effective management of severe radiation dermatitis after head and neck radiotherapy
Head and neck cancer is among the most prevalent cancers in developing countries.1 Most of the patients in developing countries present in locally advanced stages, and radical radiation therapy with concurrent chemotherapy is the standard treatment.1 Radiation therapy is associated with radiation dermatitis, which causes severe symptoms in the patient and can lead to disruption of treatment, diminished rates of disease control rates, and impaired patient quality of life.2 The management of advanced radiation dermatitis is difficult and can cause consequential late morbidity to patients.2 We report here the rare case of a patient with locally advanced tonsil carcinoma who developed grade 3 radiation dermatitis while receiving radical chemoradiation. The patient’s radiation dermatitis was effectively managed with the use of a silver-containing antimicrobial dressing that yielded remarkable results, so the patient was able to resume and complete radiation therapy.
Case presentation and summary
A 48-year-old man was diagnosed with squamous cell carcinoma of the right tonsil, with bilateral neck nodes (Stage T4a N2c M0; The American Joint Committee on Cancer staging manual, 7th edition). In view of the locally advanced status of his disease, the patient was scheduled for radical radiation therapy at 70 Gy in 35 fractions over 7 weeks along with weekly chemotherapy (cisplatin 40 mg/m2). During the course of radiation therapy, the patient was monitored twice a week, and symptomatic care was done for radiation-therapy–induced toxicities.
The patient presented with grade 3 radiation dermatitis after receiving 58 Gy in 29 fractions over 5 weeks (grade 0, no change; grades 3 and 4, severe change). The radiation dermatitis involved the anterior and bilateral neck with moist desquamation of the skin (Figure 1).
It was associated with severe pain, difficulty in swallowing, and oral mucositis. The patient was subsequently admitted to the hospital; radiation therapy was stopped, and treatment was initiated to ease the effects of the radiation dermatitis. Analgesics were administered for the pain, and adequate hydration and nutritional support was administered through a nasogastric tube. The patient’s score on the Bates-Jensen Wound Assessment Tool (BWAT) for monitoring wound status was 44, which falls in extreme severity status.
In view of the extreme severity status of the radiation dermatitis, after cleaning the wound with sterile water, we covered it with an antimicrobial dressing that contained silver salt (Mepilex AG; Mölnlycke Health Care, Norcross, GA). The dressing was changed regularly every 4 days. There was a gradual improvement in the radiation dermatitis (Figure 2).
Discussion
Head and neck cancer is one of the most common cancers in developing countries.1 Most patients present with locally advanced disease, so chemoradiation is the standard treatment in these patents. Radiation therapy is associated with acute and chronic toxicities. The common radiation therapy toxicities are directed at skin and mucosa, which leads to radiation dermatitis and radiation mucositis, respectively.2 These toxicities are graded as per the Radiation Therapy Oncology Group (RTOG) criteria (Table 2).3
Acute radiation dermatitis is radiation therapy dose-dependent and manifests within a few days to weeks after starting external beam radiation therapy. Its presentation varies in severity and gradually manifests as erythema, dry or moist desquamation, and ulceration when severe. These can cause severe symptoms in the patient, leading to frequent breaks in treatment, decreased rates of disease control, and impaired patient quality of life.2 Apart from RTOG grading, radiation dermatitis can also be scored using the BWAT. This tool has been validated across many studies to score initial wound status and monitor the subsequent status numerically.4 The radiation dermatitis of the index case was scored and monitored with both RTOG and BWAT scores.The management of advanced radiation dermatitis is difficult, and it causes consequential late morbidity in patients. A range of topical agents and dressings are used to treat radiation dermatitis, but there is minimal evidence to support their use.5 The Multinational Association for Supportive Care in Cancer treatment guidelines for prevention and treatment of radiation dermatitis have also concluded that there is a lack of sufficient evidence in the literature to support the superiority for any specific intervention.6 Management of radiation dermatitis varies among practitioners because of the inconclusive evidence for available treatment options.
The use of silver-based antimicrobial dressings has been reported in the literature in the prevention and treatment of radiation dermatitis, but with mixed results.7 Such dressings absorb exudate, maintain a moist environment that promotes wound healing, fight infection, and minimize the risk for maceration, according to the product information sheet.8 Clinical study findings have shown silver to be effective in fighting many different types of pathogens, including Methicillin-resistant Staphylococcus aureus and other drug-resistant bacteria.
Aquino-Parsons and colleagues studied 196 patients with breast cancer who were undergoing whole-breast radiation therapy.9 They showed that there was no benefit of silver-containing foam dressings for the prevention of acute grade 3 radiation dermatitis compared with patients who received standard skin care (with moisturizing cream, topical steroids, saline compress, and silver sulfadiazine cream). However, the incidence of itching in the last week of radiation and 1 week after treatment completion was lower among the patients who used the dressings.
Diggelmann and colleagues studied 24 patients with breast cancer who were undergoing radiation therapy.10 Each of the erythematous areas (n = 34) was randomly divided into 2 groups; 1 group was treated with Mepilex Lite dressing and the other with standard aqueous cream. There was a significant reduction in the severity of acute radiation dermatitis in the areas on which Mepilex Lite dressings were used compared with the areas on which standard aqueous cream was used.
The patient in the present case had severe grade 3 acute radiation dermatitis with a BWAT score indicative of extreme severity. After cleaning the wound with sterile water, instead of using the standard aqueous cream on the wounds, we used Mepilex AG, an antimicrobial dressing that contains silver salt. The results were remarkable (Figure 2 and Table 2). The patient was able to restart radiation therapy, and he completed his scheduled doses.
This case highlights the effectiveness of a silver-based antimicrobial dressing in the management of advanced and severe radiation dermatitis. Further large and randomized studies are needed to test the routine use of the dressing in the management of radiation dermatitis.
1. Simard EP, Torre LA, Jemal A. International trends in head and neck cancer incidence rates: differences by country, sex and anatomic site. Oral Oncol. 2014;50(5):387-403.
2. Hymes SR, Strom EA, Fife C. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 2006;54(1):28-46.
3. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31(5):1341-1346.
4. Harris C, Bates-Jensen B, Parslow N, Raizman R, Singh M, Ketchen R. Bates‐Jensen wound assessment tool: pictorial guide validation project. J Wound Ostomy Continence Nurs. 2010;37(3):253-259.
5. Lucey P, Zouzias C, Franco L, Chennupati SK, Kalnicki S, McLellan BN. Practice patterns for the prophylaxis and treatment of acute radiation dermatitis in the United States. Support Care Cancer. 2017;25(9):2857-2862.
6. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013;21(10):2933-2948.
7. Vavassis P, Gelinas M, Chabot Tr J, Nguyen-Tân PF. Phase 2 study of silver leaf dressing for treatment of radiation-induced dermatitis in patients receiving radiotherapy to the head and neck. J Otolaryngology Head Neck Surg. 2008;37(1):124-129.
8. Mepilex Ag product information. Mölnlycke Health Care website. http://www.molnlycke.us/advanced-wound-care-products/antimicrobial-products/mepilex-ag/#confirm. Accessed May 3, 2018.
9. Aquino-Parsons C, Lomas S, Smith K, et al. Phase III study of silver leaf nylon dressing vs standard care for reduction of inframammary moist desquamation in patients undergoing adjuvant whole breast radiation therapy. J Med Imaging Radiat Sci. 2010;41(4):215-221.
10. Diggelmann KV, Zytkovicz AE, Tuaine JM, Bennett NC, Kelly LE, Herst PM. Mepilex Lite dressings for the management of radiation-induced erythema: a systematic inpatient controlled clinical trial. Br J Radiol. 2010;83(995):971-978.
Head and neck cancer is among the most prevalent cancers in developing countries.1 Most of the patients in developing countries present in locally advanced stages, and radical radiation therapy with concurrent chemotherapy is the standard treatment.1 Radiation therapy is associated with radiation dermatitis, which causes severe symptoms in the patient and can lead to disruption of treatment, diminished rates of disease control rates, and impaired patient quality of life.2 The management of advanced radiation dermatitis is difficult and can cause consequential late morbidity to patients.2 We report here the rare case of a patient with locally advanced tonsil carcinoma who developed grade 3 radiation dermatitis while receiving radical chemoradiation. The patient’s radiation dermatitis was effectively managed with the use of a silver-containing antimicrobial dressing that yielded remarkable results, so the patient was able to resume and complete radiation therapy.
Case presentation and summary
A 48-year-old man was diagnosed with squamous cell carcinoma of the right tonsil, with bilateral neck nodes (Stage T4a N2c M0; The American Joint Committee on Cancer staging manual, 7th edition). In view of the locally advanced status of his disease, the patient was scheduled for radical radiation therapy at 70 Gy in 35 fractions over 7 weeks along with weekly chemotherapy (cisplatin 40 mg/m2). During the course of radiation therapy, the patient was monitored twice a week, and symptomatic care was done for radiation-therapy–induced toxicities.
The patient presented with grade 3 radiation dermatitis after receiving 58 Gy in 29 fractions over 5 weeks (grade 0, no change; grades 3 and 4, severe change). The radiation dermatitis involved the anterior and bilateral neck with moist desquamation of the skin (Figure 1).
It was associated with severe pain, difficulty in swallowing, and oral mucositis. The patient was subsequently admitted to the hospital; radiation therapy was stopped, and treatment was initiated to ease the effects of the radiation dermatitis. Analgesics were administered for the pain, and adequate hydration and nutritional support was administered through a nasogastric tube. The patient’s score on the Bates-Jensen Wound Assessment Tool (BWAT) for monitoring wound status was 44, which falls in extreme severity status.
In view of the extreme severity status of the radiation dermatitis, after cleaning the wound with sterile water, we covered it with an antimicrobial dressing that contained silver salt (Mepilex AG; Mölnlycke Health Care, Norcross, GA). The dressing was changed regularly every 4 days. There was a gradual improvement in the radiation dermatitis (Figure 2).
Discussion
Head and neck cancer is one of the most common cancers in developing countries.1 Most patients present with locally advanced disease, so chemoradiation is the standard treatment in these patents. Radiation therapy is associated with acute and chronic toxicities. The common radiation therapy toxicities are directed at skin and mucosa, which leads to radiation dermatitis and radiation mucositis, respectively.2 These toxicities are graded as per the Radiation Therapy Oncology Group (RTOG) criteria (Table 2).3
Acute radiation dermatitis is radiation therapy dose-dependent and manifests within a few days to weeks after starting external beam radiation therapy. Its presentation varies in severity and gradually manifests as erythema, dry or moist desquamation, and ulceration when severe. These can cause severe symptoms in the patient, leading to frequent breaks in treatment, decreased rates of disease control, and impaired patient quality of life.2 Apart from RTOG grading, radiation dermatitis can also be scored using the BWAT. This tool has been validated across many studies to score initial wound status and monitor the subsequent status numerically.4 The radiation dermatitis of the index case was scored and monitored with both RTOG and BWAT scores.The management of advanced radiation dermatitis is difficult, and it causes consequential late morbidity in patients. A range of topical agents and dressings are used to treat radiation dermatitis, but there is minimal evidence to support their use.5 The Multinational Association for Supportive Care in Cancer treatment guidelines for prevention and treatment of radiation dermatitis have also concluded that there is a lack of sufficient evidence in the literature to support the superiority for any specific intervention.6 Management of radiation dermatitis varies among practitioners because of the inconclusive evidence for available treatment options.
The use of silver-based antimicrobial dressings has been reported in the literature in the prevention and treatment of radiation dermatitis, but with mixed results.7 Such dressings absorb exudate, maintain a moist environment that promotes wound healing, fight infection, and minimize the risk for maceration, according to the product information sheet.8 Clinical study findings have shown silver to be effective in fighting many different types of pathogens, including Methicillin-resistant Staphylococcus aureus and other drug-resistant bacteria.
Aquino-Parsons and colleagues studied 196 patients with breast cancer who were undergoing whole-breast radiation therapy.9 They showed that there was no benefit of silver-containing foam dressings for the prevention of acute grade 3 radiation dermatitis compared with patients who received standard skin care (with moisturizing cream, topical steroids, saline compress, and silver sulfadiazine cream). However, the incidence of itching in the last week of radiation and 1 week after treatment completion was lower among the patients who used the dressings.
Diggelmann and colleagues studied 24 patients with breast cancer who were undergoing radiation therapy.10 Each of the erythematous areas (n = 34) was randomly divided into 2 groups; 1 group was treated with Mepilex Lite dressing and the other with standard aqueous cream. There was a significant reduction in the severity of acute radiation dermatitis in the areas on which Mepilex Lite dressings were used compared with the areas on which standard aqueous cream was used.
The patient in the present case had severe grade 3 acute radiation dermatitis with a BWAT score indicative of extreme severity. After cleaning the wound with sterile water, instead of using the standard aqueous cream on the wounds, we used Mepilex AG, an antimicrobial dressing that contains silver salt. The results were remarkable (Figure 2 and Table 2). The patient was able to restart radiation therapy, and he completed his scheduled doses.
This case highlights the effectiveness of a silver-based antimicrobial dressing in the management of advanced and severe radiation dermatitis. Further large and randomized studies are needed to test the routine use of the dressing in the management of radiation dermatitis.
Head and neck cancer is among the most prevalent cancers in developing countries.1 Most of the patients in developing countries present in locally advanced stages, and radical radiation therapy with concurrent chemotherapy is the standard treatment.1 Radiation therapy is associated with radiation dermatitis, which causes severe symptoms in the patient and can lead to disruption of treatment, diminished rates of disease control rates, and impaired patient quality of life.2 The management of advanced radiation dermatitis is difficult and can cause consequential late morbidity to patients.2 We report here the rare case of a patient with locally advanced tonsil carcinoma who developed grade 3 radiation dermatitis while receiving radical chemoradiation. The patient’s radiation dermatitis was effectively managed with the use of a silver-containing antimicrobial dressing that yielded remarkable results, so the patient was able to resume and complete radiation therapy.
Case presentation and summary
A 48-year-old man was diagnosed with squamous cell carcinoma of the right tonsil, with bilateral neck nodes (Stage T4a N2c M0; The American Joint Committee on Cancer staging manual, 7th edition). In view of the locally advanced status of his disease, the patient was scheduled for radical radiation therapy at 70 Gy in 35 fractions over 7 weeks along with weekly chemotherapy (cisplatin 40 mg/m2). During the course of radiation therapy, the patient was monitored twice a week, and symptomatic care was done for radiation-therapy–induced toxicities.
The patient presented with grade 3 radiation dermatitis after receiving 58 Gy in 29 fractions over 5 weeks (grade 0, no change; grades 3 and 4, severe change). The radiation dermatitis involved the anterior and bilateral neck with moist desquamation of the skin (Figure 1).
It was associated with severe pain, difficulty in swallowing, and oral mucositis. The patient was subsequently admitted to the hospital; radiation therapy was stopped, and treatment was initiated to ease the effects of the radiation dermatitis. Analgesics were administered for the pain, and adequate hydration and nutritional support was administered through a nasogastric tube. The patient’s score on the Bates-Jensen Wound Assessment Tool (BWAT) for monitoring wound status was 44, which falls in extreme severity status.
In view of the extreme severity status of the radiation dermatitis, after cleaning the wound with sterile water, we covered it with an antimicrobial dressing that contained silver salt (Mepilex AG; Mölnlycke Health Care, Norcross, GA). The dressing was changed regularly every 4 days. There was a gradual improvement in the radiation dermatitis (Figure 2).
Discussion
Head and neck cancer is one of the most common cancers in developing countries.1 Most patients present with locally advanced disease, so chemoradiation is the standard treatment in these patents. Radiation therapy is associated with acute and chronic toxicities. The common radiation therapy toxicities are directed at skin and mucosa, which leads to radiation dermatitis and radiation mucositis, respectively.2 These toxicities are graded as per the Radiation Therapy Oncology Group (RTOG) criteria (Table 2).3
Acute radiation dermatitis is radiation therapy dose-dependent and manifests within a few days to weeks after starting external beam radiation therapy. Its presentation varies in severity and gradually manifests as erythema, dry or moist desquamation, and ulceration when severe. These can cause severe symptoms in the patient, leading to frequent breaks in treatment, decreased rates of disease control, and impaired patient quality of life.2 Apart from RTOG grading, radiation dermatitis can also be scored using the BWAT. This tool has been validated across many studies to score initial wound status and monitor the subsequent status numerically.4 The radiation dermatitis of the index case was scored and monitored with both RTOG and BWAT scores.The management of advanced radiation dermatitis is difficult, and it causes consequential late morbidity in patients. A range of topical agents and dressings are used to treat radiation dermatitis, but there is minimal evidence to support their use.5 The Multinational Association for Supportive Care in Cancer treatment guidelines for prevention and treatment of radiation dermatitis have also concluded that there is a lack of sufficient evidence in the literature to support the superiority for any specific intervention.6 Management of radiation dermatitis varies among practitioners because of the inconclusive evidence for available treatment options.
The use of silver-based antimicrobial dressings has been reported in the literature in the prevention and treatment of radiation dermatitis, but with mixed results.7 Such dressings absorb exudate, maintain a moist environment that promotes wound healing, fight infection, and minimize the risk for maceration, according to the product information sheet.8 Clinical study findings have shown silver to be effective in fighting many different types of pathogens, including Methicillin-resistant Staphylococcus aureus and other drug-resistant bacteria.
Aquino-Parsons and colleagues studied 196 patients with breast cancer who were undergoing whole-breast radiation therapy.9 They showed that there was no benefit of silver-containing foam dressings for the prevention of acute grade 3 radiation dermatitis compared with patients who received standard skin care (with moisturizing cream, topical steroids, saline compress, and silver sulfadiazine cream). However, the incidence of itching in the last week of radiation and 1 week after treatment completion was lower among the patients who used the dressings.
Diggelmann and colleagues studied 24 patients with breast cancer who were undergoing radiation therapy.10 Each of the erythematous areas (n = 34) was randomly divided into 2 groups; 1 group was treated with Mepilex Lite dressing and the other with standard aqueous cream. There was a significant reduction in the severity of acute radiation dermatitis in the areas on which Mepilex Lite dressings were used compared with the areas on which standard aqueous cream was used.
The patient in the present case had severe grade 3 acute radiation dermatitis with a BWAT score indicative of extreme severity. After cleaning the wound with sterile water, instead of using the standard aqueous cream on the wounds, we used Mepilex AG, an antimicrobial dressing that contains silver salt. The results were remarkable (Figure 2 and Table 2). The patient was able to restart radiation therapy, and he completed his scheduled doses.
This case highlights the effectiveness of a silver-based antimicrobial dressing in the management of advanced and severe radiation dermatitis. Further large and randomized studies are needed to test the routine use of the dressing in the management of radiation dermatitis.
1. Simard EP, Torre LA, Jemal A. International trends in head and neck cancer incidence rates: differences by country, sex and anatomic site. Oral Oncol. 2014;50(5):387-403.
2. Hymes SR, Strom EA, Fife C. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 2006;54(1):28-46.
3. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31(5):1341-1346.
4. Harris C, Bates-Jensen B, Parslow N, Raizman R, Singh M, Ketchen R. Bates‐Jensen wound assessment tool: pictorial guide validation project. J Wound Ostomy Continence Nurs. 2010;37(3):253-259.
5. Lucey P, Zouzias C, Franco L, Chennupati SK, Kalnicki S, McLellan BN. Practice patterns for the prophylaxis and treatment of acute radiation dermatitis in the United States. Support Care Cancer. 2017;25(9):2857-2862.
6. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013;21(10):2933-2948.
7. Vavassis P, Gelinas M, Chabot Tr J, Nguyen-Tân PF. Phase 2 study of silver leaf dressing for treatment of radiation-induced dermatitis in patients receiving radiotherapy to the head and neck. J Otolaryngology Head Neck Surg. 2008;37(1):124-129.
8. Mepilex Ag product information. Mölnlycke Health Care website. http://www.molnlycke.us/advanced-wound-care-products/antimicrobial-products/mepilex-ag/#confirm. Accessed May 3, 2018.
9. Aquino-Parsons C, Lomas S, Smith K, et al. Phase III study of silver leaf nylon dressing vs standard care for reduction of inframammary moist desquamation in patients undergoing adjuvant whole breast radiation therapy. J Med Imaging Radiat Sci. 2010;41(4):215-221.
10. Diggelmann KV, Zytkovicz AE, Tuaine JM, Bennett NC, Kelly LE, Herst PM. Mepilex Lite dressings for the management of radiation-induced erythema: a systematic inpatient controlled clinical trial. Br J Radiol. 2010;83(995):971-978.
1. Simard EP, Torre LA, Jemal A. International trends in head and neck cancer incidence rates: differences by country, sex and anatomic site. Oral Oncol. 2014;50(5):387-403.
2. Hymes SR, Strom EA, Fife C. Radiation dermatitis: clinical presentation, pathophysiology, and treatment 2006. J Am Acad Dermatol. 2006;54(1):28-46.
3. Cox JD, Stetz J, Pajak TF. Toxicity criteria of the Radiation Therapy Oncology Group (RTOG) and the European Organization for Research and Treatment of Cancer (EORTC). Int J Radiat Oncol Biol Phys. 1995;31(5):1341-1346.
4. Harris C, Bates-Jensen B, Parslow N, Raizman R, Singh M, Ketchen R. Bates‐Jensen wound assessment tool: pictorial guide validation project. J Wound Ostomy Continence Nurs. 2010;37(3):253-259.
5. Lucey P, Zouzias C, Franco L, Chennupati SK, Kalnicki S, McLellan BN. Practice patterns for the prophylaxis and treatment of acute radiation dermatitis in the United States. Support Care Cancer. 2017;25(9):2857-2862.
6. Wong RK, Bensadoun RJ, Boers-Doets CB, et al. Clinical practice guidelines for the prevention and treatment of acute and late radiation reactions from the MASCC Skin Toxicity Study Group. Support Care Cancer. 2013;21(10):2933-2948.
7. Vavassis P, Gelinas M, Chabot Tr J, Nguyen-Tân PF. Phase 2 study of silver leaf dressing for treatment of radiation-induced dermatitis in patients receiving radiotherapy to the head and neck. J Otolaryngology Head Neck Surg. 2008;37(1):124-129.
8. Mepilex Ag product information. Mölnlycke Health Care website. http://www.molnlycke.us/advanced-wound-care-products/antimicrobial-products/mepilex-ag/#confirm. Accessed May 3, 2018.
9. Aquino-Parsons C, Lomas S, Smith K, et al. Phase III study of silver leaf nylon dressing vs standard care for reduction of inframammary moist desquamation in patients undergoing adjuvant whole breast radiation therapy. J Med Imaging Radiat Sci. 2010;41(4):215-221.
10. Diggelmann KV, Zytkovicz AE, Tuaine JM, Bennett NC, Kelly LE, Herst PM. Mepilex Lite dressings for the management of radiation-induced erythema: a systematic inpatient controlled clinical trial. Br J Radiol. 2010;83(995):971-978.
The long-term effects of posttreatment exercise on pain in young women with breast cancer
Breast cancer is one of the most prevalent cancers in women worldwide, with more than 1 million new cases diagnosed annually.1 Prognosis for the disease has improved significantly, but 25% to 60% of women living with breast cancer experience some level of pain ranging from mild to severe, the nature of which can evolve from acute to chronic.2 Pre-, intra-, and post-treatment risk factors have been found to correlate with the development of acute and chronic pain and include young age, type of breast surgery (lumpectomy or total mastectomy), axillary node dissection, radiation therapy, and hormonal therapy.3-5 Chemotherapy, particularly anthracycline- and taxane-based regimens, has also been shown to induce pain, arthralgia, myalgia, and peripheral neuropathy during treatment.6 In particular, postradiation pain may result from subcutaneous fibrosis with fixation to underlying musculature and the development of fibrous flaps in the internal axilla.7 These tissue changes are commonly subclinical, occurring 4 to 12 months postradiation,8 and can progress undetected until pain and upper-limb disability develop.
The presence of persistent pain has a considerable impact on the quality of life in survivors of breast cancer: psychological distress is prevalent (anxiety, depression, worry, fear), the performance of daily activities is diminished (eg, bathing, dressing, preparing meals, shopping), and economic independence is compromised by the inability to work or reduced employment and income. These factors directly and indirectly contribute to an increase in the use of health care services.9,10
The management of pain is often characterized by pharmacologic-related treatment, such as the use of opioids and nonsteroidal anti-inflammatory medications, and nonpharmacologic-related treatment, such as exercise. Empirical evidence has shown that rehabilitative exercise programs, which commonly include a combination of resistance training and aerobic exercises, can effectively reduce pain in breast cancer survivors.10-12 Women living with breast cancer who are directed to rehabilitative exercise programs experience an improvement not only in pain levels but also in their ability to engage in activities of daily living, in their psychological health, and in their overall quality of life.13-15 However, despite evidence to support exercise programs to reduce pain related to breast cancer treatment, residual pain and upper-limb discomfort are common complaints in breast cancer survivors, and there is little focus on the duration of effectiveness of such programs for reducing pain after treatment for breast cancer. The objective of this study was to determine if an exercise program initiated postradiation would improve long-term pain levels in a carefully selected population of young women who were living with breast cancer and had no history of shoulder pathology or significant treatment complications.
Methods
Design
We used a pilot randomized control trial to compare the long-term effectiveness of a 12-week postradiation exercise program versus standard care on residual pain levels in young women (aged 18-45 years) living with breast cancer. The program was initiated 3 to 4 weeks postradiation to allow for acute inflammatory reactions to subside. Pain severity and interference were assessed using the Brief Pain Inventory-Short Form (BPI-SF), a tool for assessing cancer pain.16,17 Pain levels for isolated shoulder movements were also recorded on examination by a physical therapist. All measures were collected at 6 time points (T1-T6): postsurgery and preradiation (T1, baseline), postradiation and preintervention (T2), and 4 points during an 18-month period postradiation (T3-T6 at 3, 6, 12, and 18 months postradiation).
Sample
Young women living with breast cancer who met our eligibility criteria were identified from 2 clinics at the Jewish General Hospital – the Segal Cancer Center and the Department of Radiation Oncology in Montréal, Québec, Canada. Inclusion criteria included women with a diagnosis of stage I to stage III breast cancer, who were 18 to 45 years old, were scheduled for postoperative adjuvant radiation therapy, had an Eastern Cooperative Oncology Group Performance Status of 0 or 1 (normal ambulatory function, minimal symptoms), and who consented to participate in the study. Exclusion criteria included women with a metastatic (stage IV) diagnosis; significant musculoskeletal, cardiac, pulmonary, or metabolic comorbidities that would not allow for participation in physical activity; a previous breast cancer diagnosis with treatment to the ipsilateral or contralateral sides; postsurgical lymphedema; postsurgical capsulitis, tendonitis, or other shoulder inflammatory complications; and any contraindication to exercise. The recruitment goal was outlined as 50 patients per group; however, a protracted accrual time because of the stringent study criteria yielded a sample of 29 and 30 patients for the intervention and control groups, respectively, which was sufficient for significant testing of differences between the 2 study groups.18
Variables and measures
Clinical characteristics. We used standardized questions and chart review to document the participants’ clinical characteristics and to capture information on the following: the stage and subtype of breast cancer, hormonal and human epidermal growth factor receptors (HER2) (estrogen receptor, progesterone receptor, and HER2 status), extent of surgery (lumpectomy or total mastectomy), and other modalities of treatment (eg, chemotherapy, radiation therapy).
Pain assessment. The BPI-SF was used to assess participants’ cancer-related pain. Pain severity ranged from 0 (no pain), 1 to 4 (mild pain), 5 to 6 (moderate pain), to 7 to 10 (severe pain).18,19 The questionnaire also identifies the pain interference in daily activities using a Likert scale ranging from 0 (Does not interfere) to 10 (Completely interferes) in the following 7 domains or subscales: General Activity, Walking, Mood, Sleep, Work, Relations with Others, and Enjoyment of Life.16 For the purpose of this study, mean scores were tabulated using both pain intensity and interference scales.
Another important component of the BPI-SF instructs participants to localize pain by means of a body diagram. For purpose of analysis, 3 pain regions were established: shoulder girdle/chest wall on the affected side; neck and other upper extremity, including hand(s), forearm(s), wrist(s), and finger(s); and other regions, including abdominal discomfort, leg(s), hip(s), knee(s), ankle(s), lower back, and feet. In addition, pain levels on movement (Yes/No) were recorded for isolated shoulder flexion, abduction, and horizontal abduction (sitting and standing). The measurements were completed by a single physical therapist throughout the course of the study to minimize variance.
Procedure
The study protocol was approved by the Research Ethics Board at the Jewish General Hospital. Recruitment occurred from 2011 through 2015. The research was in accordance with the ethical standards of the responsible committee on human experimentation. Eligible women were recruited by the research coordinator who described the purpose, risks, and benefits of the study; advised on confidentiality, data collection, and intervention allocation procedures; and highlighted voluntary participation. The research coordinator addressed any concerns on the part of the participants before obtaining their written informed consent. Random allocation to the intervention and control groups was established using a web-based randomization plan generator (www.randomization.com). A single individual was responsible for the randomization process, and treatment assignments were revealed after each participant’s name had been entered. A physical therapist performed 6 sequential evaluations (T1-T6) at the time of participants’ medical follow-up appointments.
Intervention
The 12-week exercise intervention started 3 weeks postradiation and was composed of an initial 6-week program of low-level cardiovascular and resistance exercises that progressed to a set of more advanced exercises for the remaining 6 weeks. Participants were instructed to warm up for at least 10 minutes with a cardiovascular exercise of their choice (eg, a recumbent cross trainer, walking, or stairs) before doing a combined strength, endurance, and stretching exercise program for the upper body.20 The final portion of the exercise intervention included a period of light cool-down. Weight training resistance levels were based on a maximum 8 to 10 repetitions for strength and a maximum of 20 repetitions for endurance training exercises, which progressed gradually over the course of the 12-week exercise program to ensure participant safety.21,22 Participants in the intervention group were supervised at least once a week by an exercise physiologist at a center for oncology patients (Hope & Cope Wellness Centre), and patients were encouraged to perform the program at home 2 to 3 times a week. Those who were not able to exercise consistently at the center were provided with equipment and instructed on how to do the program safely at home.
By comparison, the control group received standard care, which included advice on the benefits of an active lifestyle, including exercise, but without a specific intervention. Participants were not restricted in their physical activity and/or sport participation levels, and their weekly activity levels were calculated using the Metabolic Equivalent of Task and recorded at each of the 6 time points.
Statistical analysis
Descriptive statistics were used to examine participant characteristics. The quantitative data collected through the BPI-SF measures were analyzed with JMP software (version 11.2; SAS Institute, Cary, NC). Continuous variables were tested for statistical significance (P ≤ .05) through the chi-square (categorical), analysis of variance, and nonparametric Wilcoxon tests. The analyses did not include missing data.
Results
A total of 59 young women were randomized into the intervention (n = 29) and control (n = 30) groups. Of those, 2 participants dropped out of the study because of family and time constraints, and 3 participants died, 2 from the control and 1 from the intervention group, after subsequently developing metastatic disease. Baseline data including comparative tumor characteristics, surgical interventions, and treatment interventions have been published in relation to other elements of this study.23,24 The participants had a mean age of 39.2 years (standard deviation [SD], 5.0). More than half of them had an invasive ductal carcinoma (69.5%) and were estrogen positive (78.0%), progesterone positive (74.6%), or HER2 positive (20.3%), whereas 10.2% were triple negative. Most of the participants had undergone breast-sparing procedures (86.4% lumpectomy), and 18.6% had a total mastectomy. By random chance, the intervention group had higher rates of total mastectomy (24.4% and 13.3%, respectively) and surgical reconstruction (12.2% and 6.7%, respectively) compared with the control group. Most of the women (71.2%) received chemotherapy, and all received radiation therapy. In the intervention group, 37.2% received radiation therapy localized to the axilla, and 88% received a boost of radiation to the surgical bed. Self-reported exercise diaries were returned by 15 of the 29 intervention participants, and training frequencies among them varied significantly (1-6 times a week).
The findings showed that there was little variance between the intervention and control groups in BPI-SF severity scores from T1 to T6, so the means and SDs of the BPI-SF scores were grouped at 6 time points (Table 1). There was no statistically significant difference between baseline measures at T1 (1.68; SD, 1.17) and measures at 18 months postintervention (T6: 1.46; SD, 1.37). At baseline, 87.7% of the women reported no pain (31.5%) or mild levels of pain (55.6%), and 13% reported moderate or severe pain. Over the duration of the study from T1 to T6, these primarily low levels of pain (BPI-SF, 0-4) remained consistent with a favorable shift toward having no pain (T1: 31.5%; T6: 24.4%). By 18 months postintervention, 95.7% of women reported no or mild pain, with 4.9% reporting moderate pain.
Similarly, there was little variance over time (T1-T6) and no statistically significant differences between the 2 groups in BPI-SF–measured levels of pain interference in daily activities (Table 2). Moreover, a domain analysis showed that there were no statistically significant differences in pain interference scores when comparing the type and extent of surgery (total mastectomy: 0.59 [1.17]; lumpectomy: 0.94 [1.96]). By chance – and not related directly to the objectives of this study – there was a statistically significant difference between the intervention and control groups in the interference of pain on the Enjoyment of Life domain in favor of the control group.
The sites of pain captured by the BPI-SF shed light on the preceding findings (Figure 1). At baseline (T1, postsurgery and preradiation), 37.0% of participants reported pain in the shoulder girdle–chest wall region, whereas 20.4% reported pain in the general neck–upper extremity region and 50% in other regions. Postradiation, shoulder girdle–chest wall pain was identified as the highest reported site of pain (49.1%; T2, postradiation and preintervention) and remained elevated at 3 months (T3) and 6 months (T4) postradiation (46.9% and 45.5%, respectively). At 12 and 18 months postradiation (T5 and T6), the principal focus of pain shifted once again to “other” regions at 30% and 32.5%, respectively, and the neck–upper extremity region at 10% and 15%, respectively. Shoulder girdle–chest wall pain concomitantly improved at those time points (15% and 25% respectively) but was not eliminated.
Pain levels recorded on physical examination for isolated shoulder range of movements were recently published,24 and they have been abbreviated and reproduced in this paper (Figure 2) to allow for a comparison of findings between the exercise intervention group and the control group to help determine the sensitivity of these tools for use in breast cancer patients. At baseline, pain levels with active movement were noted to be slightly greater in the intervention group for flexion and abduction.
Following the intervention, at 3 and 6 months postradiation (T3 and T4), the intervention group showed a steady decrease in pain levels in flexion and abduction, whereas the control group showed a 5-fold increase in pain with horizontal abduction. Furthermore, participants in the intervention group reported having no pain on movement 12 months postradiation (T5); however, recurrence of pain was apparent with all shoulder movements by 18 months postradiation (T6) in both the intervention and control groups.
Discussion
Previous studies have hypothesized that younger age (18-39 years), adjuvant radiotherapy, and axillary node dissection are risk factors for chronic pain in breast cancer survivors.22,25 Persistent pain is prevalent in 12% to 51% of breast cancer survivors, with up to one-third experiencing some pain more than 5 years after treatment,26,27 and our study outcomes concur with those findings. In our study, pain, as measured by the BPI-SF, was found to persist for most participants (75.6%) after the 18-month follow-up. The results of our trial showed that a 12-week exercise intervention administered postsurgery and postradiation had no statistically significant effect on long-term (18 months) pain severity and its interference in daily life. It is worth noting that body regions that had not been directly related to either surgical or radiation treatment for breast cancer were commonly identified as areas of pain but were not specifically targeted by our intervention. However, focusing on pain severity (BPI-SF), our findings suggest that the benefits of targeted upper-extremity exercise on pain in the intermediate time course of follow-up (T3, T4, and T5) was notable compared with the control group, which received standard care. The apparent recurrence of pain at 18 months in both groups was not anticipated and needs to be further investigated.
More specific objective assessments of pain on active shoulder movement identified distinct patterns of pain that could not be isolated using the BPI-SF alone. The incidence and localization of pain on movement differed between the population of women who received a specific exercise intervention and those who received standard care (Figure 2). Patterns of pain over time fluctuated in the control group, whereas the intervention group reported a linear decrease in pain. Residual pain on shoulder movement remained apparent in both groups at 18-months postradiation, but that finding was not reflected in the BPI-SF results. The literature supports our findings on persistent pain among breast cancer survivors,3,7,8,28-30 and in our study of young women carefully screened and excluded for pre-existent shoulder conditions or comorbid medical conditions, recurrent articular pain was nonetheless prevalent. It seems that unidentified or multiple factors may be part of the etiology of pain in this young adult cohort.
Although the BPI-SF is a generic measurement tool commonly used to assess and measure cancer patients’ pain levels, the lack of variance in our BPI-SF severity and interference outcomes over time (T1-T6) (Table 1, Table 2), the variety of “other” unrelated regions (Figure 1) identified by the BPI-SF, and the contrast in our findings on specific physical examination emphasize the potential limitations of this clinical tool.
Moreover, the BPI-SF has not been validated specifically for breast cancer. Harrington and colleagues have recommended using the BPI-SF to assess pain in women with breast cancer,31 but the use of a more multidimensional measurement tool that evaluates axillary, chest, trunk, and upper-limb pain may prove to be more valuable in this population.
Limitations
Recruitment of young adult women was difficult because of our stringent inclusion criteria, the long-term follow-up, and the relatively small population of breast cancer patients in this age demographic. Therefore, the duration of the recruitment phase, despite our having access to a specialized young adult and adolescent clinic in our institute, greatly surpassed the expectations we had when we designed the study. In addition, there remains an inherent bias in participants who accept participation in a study that includes exercise interventions. Potential participants who exercise regularly or have a positive inclination toward doing exercise are more likely to participate. Despite the prescription of a targeted 12-week upper-limb intervention in this study, the general activity levels of both groups may have had an impact on the significance of this study. In addition, the low adherence to the use of self-reported logs failed to capture the true compliance rates of our participants because their lack of tracking does not indicate failure to comply with the program. The use of weekly or biweekly telephone calls to monitor compliance rates of activity more vigilantly may be used in future studies.
Conclusions
Advances in clinical management of breast cancer have improved survival outcomes, and morbidity over recent years, yet symptoms such as pain remain prevalent in this population. The results of this study showed that a targeted, 12-week upper-limb exercise intervention postradiation transiently improved levels of shoulder pain without a concomitant impact on chronic pain or any positive influence on activities of daily living 18 months posttreatment. Furthermore, future studies should use a variety of measurement tools to evaluate trunk and upper-limb pain in women with breast cancer and investigate the optimal timing of postradiation exercise interventions.
Acknowledgments
The authors thank Hope & Cope, the CURE foundation, and the Jewish General Hospital Foundation/Weekend to End Breast Cancer for providing the financial resources needed to sustain this research study. They also thank the McGill Adolescent and Young Adult program for its continued support. Previous oral presentations of research Muanza TM, et al. Randomized clinical trial of a progressive exercise program for young women with breast cancer undergoing radiation therapy. Int J Radiat Oncol Biol Phys. 2015;93(3):s35-s36.
1. World Health Organization. Breast cancer: prevention and control. www.who.int/cancer/detection/breastcancer/en/. Updated 2017. Accessed September 16, 2016.
2. Andersen KG, Kehlet H. Persistent pain after breast cancer treatment: a critical review of risk factors and strategies for prevention. J Pain. 2011;12(7):725-746.
3. Ernst MF, Voogd AC, Balder W, Klinkenbijl JH, Roukema JA. Early and late morbidity associated with axillary levels I-III dissection in breast cancer. J Surg Oncol. 2002;79(3):151-155; discussion 156.
4. Gulluoglu BM, Cingi A, Cakir T, Gercek A, Barlas A, Eti Z. Factors related to post-treatment chronic pain in breast cancer survivors: the interference of pain with life functions. Int J Fertil Womens Med. 2006;51(2):75-82.
5. Jung BF, Ahrendt GM, Oaklander AL, Dworkin RH. Neuropathic pain following breast cancer surgery: proposed classification and research update. Pain. 2003;104(1-2):1-13.
6. Saibil S, Fitzgerald B, Freedman OC, et al. Incidence of taxane-induced pain and distress in patients receiving chemotherapy for early-stage breast cancer: a retrospective, outcomes-based survey. Curr Oncol. 2010;17(4):42-47.
7. Tengrup I, Tennvall-Nittby L, Christiansson I, Laurin M. Arm morbidity after breast-conserving therapy for breast cancer. Acta Oncol. 2000;39(3):393-397.
8. Johansen J, Overgaard J, Blichert-Toft M, Overgaard M. Treatment of morbidity associated with the management of the axilla in breast-conserving therapy. Acta Oncol. 2000;39(3):349-354.
9. Mittmann N, Porter JM, Rangrej J, et al. Health system costs for stage-specific breast cancer: a population-based approach. Curr Oncol. 2014;21(6):281-293.
10. Page A. Keeping patients safe: transforming the work environment of nurses. Washington, DC: National Academies Press; 2004.
11. McNeely ML, Campbell K, Ospina M, et al. Exercise interventions for upper-limb dysfunction due to breast cancer treatment. Cochrane Database Syst Rev. 2010;(6):CD005211. doi:10.1002/14651858.CD005211.pub2
12. Wong P, Muanza T, Hijal T, et al. Effect of exercise in reducing breast and chest-wall pain in patients with breast cancer: a pilot study. Curr Oncol. 2012;19(3):e129-e135.
13. Fernández-Lao C, Cantarero-Villanueva I, Fernández-de-Las-Peñas C, del Moral-Ávila R, Castro-Sánchez AM, Arroyo-Morales M. Effectiveness of a multidimensional physical therapy program on pain, pressure hypersensitivity, and trigger points in breast cancer survivors: a randomized controlled clinical trial. Clin J Pain. 2012;28(2):113-121.
14. Courneya KS, Mackey JR, Bell GJ, Jones LW, Field CJ, Fairey AS. Randomized controlled trial of exercise training in postmenopausal breast cancer survivors: cardiopulmonary and quality of life outcomes. J Clin Oncol. 2003;21(9):1660-1668.
15. Segal R, Evans W, Johnson D, et al. Structured exercise improves physical functioning in women with stages I and II breast cancer: results of a randomized controlled trial. J Clin Oncol. 2001;19(3):657-665.
16. Cleeland CS, Ryan KM. Pain assessment: global use of the Brief Pain Inventory. Ann Acad Med Singapore. 1994;23(2):129-138.
17. Kumar SP. Utilization of Brief Pain Inventory as an assessment tool for pain in patients with cancer: a focused review. Indian J Palliat Care. 2011;17(2):108-115.
18. Van Voorhis CRW, Morgan BL. Understanding power and rules of thumb for determining sample sizes. Tutor Quant Methods Psychol. 2007;3(2):43-50.
19. Serlin RC, Mendoza TR, Nakamura Y, Edwards KR, Cleeland CS. When is cancer pain mild, moderate or severe? Grading pain severity by its interference with function. Pain. 1995;61(2):277-284.
20. Lee TS, Kilbreath SL, Refshauge KM, Pendlebury SC, Beith JM, Lee MJ. Pectoral stretching program for women undergoing radiotherapy for breast cancer. Breast Cancer Res Treat. 2007;102(3):313-321.
21. Schmitz KH, Courneya KS, Matthews C, et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42(7):1409-1426.
22. Pollock ML, Gaesser GA, Butcher JD, et al. ACSM position stand: the recommended quantity and quality of exercise for developing and maintaining cardiorespiratory and muscular fitness, and flexibility in healthy adults. Med Sci Sports Exerc. 1998;30(6):975-991.
23. Ibrahim M, Muanza T, Smirnow N, et al. Time course of upper limb function and return-to-work post-radiotherapy in young adults with breast cancer: a pilot randomized control trial on effects of targeted exercise program. J Cancer Surviv. 2017;11(6):791-799.
24. Ibrahim M, Muanza T, Smirnow N, et al. A pilot randomized controlled trial on the effects of a progressive exercise program on the range of motion and upper extremity grip strength in young adults with breast cancer. Clin Breast Cancer. 2018;18(1):e55-e64.
25. Gärtner R, Jensen MB, Nielsen J, Ewertz M, Kroman N, Kehlet H. Prevalence of and factors associated with persistent pain following breast cancer surgery. JAMA. 2009;302(18):1985-1992.
26. Hayes SC, Johansson K, Stout NL, et al. Upper-body morbidity after breast cancer: incidence and evidence for evaluation, prevention, and management within a prospective surveillance model of care. Cancer. 2012;118(suppl 8):2237-2249.
27. Kärki A, Simonen R, Mälkiä E, Selfe J. Impairments, activity limitations and participation restrictions 6 and 12 months after breast cancer operation. J Rehabil Med. 2005;37(3):180-188.
28. Katz J, Poleshuck EL, Andrus CH, et al. Risk factors for acute pain and its persistence following breast cancer surgery. Pain. 2005;119(1-3):16-25.
29. Tasmuth T, von Smitten K, Hietanen P, Kataja M, Kalso E. Pain and other symptoms after different treatment modalities of breast cancer. Ann Oncol. 1995;6(5):453-459.
30. Whelan TJ, Levine M, Julian J, Kirkbride P, Skingley P. The effects of radiation therapy on quality of life of women with breast carcinoma: results of a randomized trial. Ontario Clinical Oncology Group. Cancer. 2000;88(10):2260-2266.
31. Harrington S, Gilchrist L, Sander A. Breast cancer EDGE task force outcomes: clinical measures of pain. Rehabil Oncol. 2014;32(1):13-21.
Breast cancer is one of the most prevalent cancers in women worldwide, with more than 1 million new cases diagnosed annually.1 Prognosis for the disease has improved significantly, but 25% to 60% of women living with breast cancer experience some level of pain ranging from mild to severe, the nature of which can evolve from acute to chronic.2 Pre-, intra-, and post-treatment risk factors have been found to correlate with the development of acute and chronic pain and include young age, type of breast surgery (lumpectomy or total mastectomy), axillary node dissection, radiation therapy, and hormonal therapy.3-5 Chemotherapy, particularly anthracycline- and taxane-based regimens, has also been shown to induce pain, arthralgia, myalgia, and peripheral neuropathy during treatment.6 In particular, postradiation pain may result from subcutaneous fibrosis with fixation to underlying musculature and the development of fibrous flaps in the internal axilla.7 These tissue changes are commonly subclinical, occurring 4 to 12 months postradiation,8 and can progress undetected until pain and upper-limb disability develop.
The presence of persistent pain has a considerable impact on the quality of life in survivors of breast cancer: psychological distress is prevalent (anxiety, depression, worry, fear), the performance of daily activities is diminished (eg, bathing, dressing, preparing meals, shopping), and economic independence is compromised by the inability to work or reduced employment and income. These factors directly and indirectly contribute to an increase in the use of health care services.9,10
The management of pain is often characterized by pharmacologic-related treatment, such as the use of opioids and nonsteroidal anti-inflammatory medications, and nonpharmacologic-related treatment, such as exercise. Empirical evidence has shown that rehabilitative exercise programs, which commonly include a combination of resistance training and aerobic exercises, can effectively reduce pain in breast cancer survivors.10-12 Women living with breast cancer who are directed to rehabilitative exercise programs experience an improvement not only in pain levels but also in their ability to engage in activities of daily living, in their psychological health, and in their overall quality of life.13-15 However, despite evidence to support exercise programs to reduce pain related to breast cancer treatment, residual pain and upper-limb discomfort are common complaints in breast cancer survivors, and there is little focus on the duration of effectiveness of such programs for reducing pain after treatment for breast cancer. The objective of this study was to determine if an exercise program initiated postradiation would improve long-term pain levels in a carefully selected population of young women who were living with breast cancer and had no history of shoulder pathology or significant treatment complications.
Methods
Design
We used a pilot randomized control trial to compare the long-term effectiveness of a 12-week postradiation exercise program versus standard care on residual pain levels in young women (aged 18-45 years) living with breast cancer. The program was initiated 3 to 4 weeks postradiation to allow for acute inflammatory reactions to subside. Pain severity and interference were assessed using the Brief Pain Inventory-Short Form (BPI-SF), a tool for assessing cancer pain.16,17 Pain levels for isolated shoulder movements were also recorded on examination by a physical therapist. All measures were collected at 6 time points (T1-T6): postsurgery and preradiation (T1, baseline), postradiation and preintervention (T2), and 4 points during an 18-month period postradiation (T3-T6 at 3, 6, 12, and 18 months postradiation).
Sample
Young women living with breast cancer who met our eligibility criteria were identified from 2 clinics at the Jewish General Hospital – the Segal Cancer Center and the Department of Radiation Oncology in Montréal, Québec, Canada. Inclusion criteria included women with a diagnosis of stage I to stage III breast cancer, who were 18 to 45 years old, were scheduled for postoperative adjuvant radiation therapy, had an Eastern Cooperative Oncology Group Performance Status of 0 or 1 (normal ambulatory function, minimal symptoms), and who consented to participate in the study. Exclusion criteria included women with a metastatic (stage IV) diagnosis; significant musculoskeletal, cardiac, pulmonary, or metabolic comorbidities that would not allow for participation in physical activity; a previous breast cancer diagnosis with treatment to the ipsilateral or contralateral sides; postsurgical lymphedema; postsurgical capsulitis, tendonitis, or other shoulder inflammatory complications; and any contraindication to exercise. The recruitment goal was outlined as 50 patients per group; however, a protracted accrual time because of the stringent study criteria yielded a sample of 29 and 30 patients for the intervention and control groups, respectively, which was sufficient for significant testing of differences between the 2 study groups.18
Variables and measures
Clinical characteristics. We used standardized questions and chart review to document the participants’ clinical characteristics and to capture information on the following: the stage and subtype of breast cancer, hormonal and human epidermal growth factor receptors (HER2) (estrogen receptor, progesterone receptor, and HER2 status), extent of surgery (lumpectomy or total mastectomy), and other modalities of treatment (eg, chemotherapy, radiation therapy).
Pain assessment. The BPI-SF was used to assess participants’ cancer-related pain. Pain severity ranged from 0 (no pain), 1 to 4 (mild pain), 5 to 6 (moderate pain), to 7 to 10 (severe pain).18,19 The questionnaire also identifies the pain interference in daily activities using a Likert scale ranging from 0 (Does not interfere) to 10 (Completely interferes) in the following 7 domains or subscales: General Activity, Walking, Mood, Sleep, Work, Relations with Others, and Enjoyment of Life.16 For the purpose of this study, mean scores were tabulated using both pain intensity and interference scales.
Another important component of the BPI-SF instructs participants to localize pain by means of a body diagram. For purpose of analysis, 3 pain regions were established: shoulder girdle/chest wall on the affected side; neck and other upper extremity, including hand(s), forearm(s), wrist(s), and finger(s); and other regions, including abdominal discomfort, leg(s), hip(s), knee(s), ankle(s), lower back, and feet. In addition, pain levels on movement (Yes/No) were recorded for isolated shoulder flexion, abduction, and horizontal abduction (sitting and standing). The measurements were completed by a single physical therapist throughout the course of the study to minimize variance.
Procedure
The study protocol was approved by the Research Ethics Board at the Jewish General Hospital. Recruitment occurred from 2011 through 2015. The research was in accordance with the ethical standards of the responsible committee on human experimentation. Eligible women were recruited by the research coordinator who described the purpose, risks, and benefits of the study; advised on confidentiality, data collection, and intervention allocation procedures; and highlighted voluntary participation. The research coordinator addressed any concerns on the part of the participants before obtaining their written informed consent. Random allocation to the intervention and control groups was established using a web-based randomization plan generator (www.randomization.com). A single individual was responsible for the randomization process, and treatment assignments were revealed after each participant’s name had been entered. A physical therapist performed 6 sequential evaluations (T1-T6) at the time of participants’ medical follow-up appointments.
Intervention
The 12-week exercise intervention started 3 weeks postradiation and was composed of an initial 6-week program of low-level cardiovascular and resistance exercises that progressed to a set of more advanced exercises for the remaining 6 weeks. Participants were instructed to warm up for at least 10 minutes with a cardiovascular exercise of their choice (eg, a recumbent cross trainer, walking, or stairs) before doing a combined strength, endurance, and stretching exercise program for the upper body.20 The final portion of the exercise intervention included a period of light cool-down. Weight training resistance levels were based on a maximum 8 to 10 repetitions for strength and a maximum of 20 repetitions for endurance training exercises, which progressed gradually over the course of the 12-week exercise program to ensure participant safety.21,22 Participants in the intervention group were supervised at least once a week by an exercise physiologist at a center for oncology patients (Hope & Cope Wellness Centre), and patients were encouraged to perform the program at home 2 to 3 times a week. Those who were not able to exercise consistently at the center were provided with equipment and instructed on how to do the program safely at home.
By comparison, the control group received standard care, which included advice on the benefits of an active lifestyle, including exercise, but without a specific intervention. Participants were not restricted in their physical activity and/or sport participation levels, and their weekly activity levels were calculated using the Metabolic Equivalent of Task and recorded at each of the 6 time points.
Statistical analysis
Descriptive statistics were used to examine participant characteristics. The quantitative data collected through the BPI-SF measures were analyzed with JMP software (version 11.2; SAS Institute, Cary, NC). Continuous variables were tested for statistical significance (P ≤ .05) through the chi-square (categorical), analysis of variance, and nonparametric Wilcoxon tests. The analyses did not include missing data.
Results
A total of 59 young women were randomized into the intervention (n = 29) and control (n = 30) groups. Of those, 2 participants dropped out of the study because of family and time constraints, and 3 participants died, 2 from the control and 1 from the intervention group, after subsequently developing metastatic disease. Baseline data including comparative tumor characteristics, surgical interventions, and treatment interventions have been published in relation to other elements of this study.23,24 The participants had a mean age of 39.2 years (standard deviation [SD], 5.0). More than half of them had an invasive ductal carcinoma (69.5%) and were estrogen positive (78.0%), progesterone positive (74.6%), or HER2 positive (20.3%), whereas 10.2% were triple negative. Most of the participants had undergone breast-sparing procedures (86.4% lumpectomy), and 18.6% had a total mastectomy. By random chance, the intervention group had higher rates of total mastectomy (24.4% and 13.3%, respectively) and surgical reconstruction (12.2% and 6.7%, respectively) compared with the control group. Most of the women (71.2%) received chemotherapy, and all received radiation therapy. In the intervention group, 37.2% received radiation therapy localized to the axilla, and 88% received a boost of radiation to the surgical bed. Self-reported exercise diaries were returned by 15 of the 29 intervention participants, and training frequencies among them varied significantly (1-6 times a week).
The findings showed that there was little variance between the intervention and control groups in BPI-SF severity scores from T1 to T6, so the means and SDs of the BPI-SF scores were grouped at 6 time points (Table 1). There was no statistically significant difference between baseline measures at T1 (1.68; SD, 1.17) and measures at 18 months postintervention (T6: 1.46; SD, 1.37). At baseline, 87.7% of the women reported no pain (31.5%) or mild levels of pain (55.6%), and 13% reported moderate or severe pain. Over the duration of the study from T1 to T6, these primarily low levels of pain (BPI-SF, 0-4) remained consistent with a favorable shift toward having no pain (T1: 31.5%; T6: 24.4%). By 18 months postintervention, 95.7% of women reported no or mild pain, with 4.9% reporting moderate pain.
Similarly, there was little variance over time (T1-T6) and no statistically significant differences between the 2 groups in BPI-SF–measured levels of pain interference in daily activities (Table 2). Moreover, a domain analysis showed that there were no statistically significant differences in pain interference scores when comparing the type and extent of surgery (total mastectomy: 0.59 [1.17]; lumpectomy: 0.94 [1.96]). By chance – and not related directly to the objectives of this study – there was a statistically significant difference between the intervention and control groups in the interference of pain on the Enjoyment of Life domain in favor of the control group.
The sites of pain captured by the BPI-SF shed light on the preceding findings (Figure 1). At baseline (T1, postsurgery and preradiation), 37.0% of participants reported pain in the shoulder girdle–chest wall region, whereas 20.4% reported pain in the general neck–upper extremity region and 50% in other regions. Postradiation, shoulder girdle–chest wall pain was identified as the highest reported site of pain (49.1%; T2, postradiation and preintervention) and remained elevated at 3 months (T3) and 6 months (T4) postradiation (46.9% and 45.5%, respectively). At 12 and 18 months postradiation (T5 and T6), the principal focus of pain shifted once again to “other” regions at 30% and 32.5%, respectively, and the neck–upper extremity region at 10% and 15%, respectively. Shoulder girdle–chest wall pain concomitantly improved at those time points (15% and 25% respectively) but was not eliminated.
Pain levels recorded on physical examination for isolated shoulder range of movements were recently published,24 and they have been abbreviated and reproduced in this paper (Figure 2) to allow for a comparison of findings between the exercise intervention group and the control group to help determine the sensitivity of these tools for use in breast cancer patients. At baseline, pain levels with active movement were noted to be slightly greater in the intervention group for flexion and abduction.
Following the intervention, at 3 and 6 months postradiation (T3 and T4), the intervention group showed a steady decrease in pain levels in flexion and abduction, whereas the control group showed a 5-fold increase in pain with horizontal abduction. Furthermore, participants in the intervention group reported having no pain on movement 12 months postradiation (T5); however, recurrence of pain was apparent with all shoulder movements by 18 months postradiation (T6) in both the intervention and control groups.
Discussion
Previous studies have hypothesized that younger age (18-39 years), adjuvant radiotherapy, and axillary node dissection are risk factors for chronic pain in breast cancer survivors.22,25 Persistent pain is prevalent in 12% to 51% of breast cancer survivors, with up to one-third experiencing some pain more than 5 years after treatment,26,27 and our study outcomes concur with those findings. In our study, pain, as measured by the BPI-SF, was found to persist for most participants (75.6%) after the 18-month follow-up. The results of our trial showed that a 12-week exercise intervention administered postsurgery and postradiation had no statistically significant effect on long-term (18 months) pain severity and its interference in daily life. It is worth noting that body regions that had not been directly related to either surgical or radiation treatment for breast cancer were commonly identified as areas of pain but were not specifically targeted by our intervention. However, focusing on pain severity (BPI-SF), our findings suggest that the benefits of targeted upper-extremity exercise on pain in the intermediate time course of follow-up (T3, T4, and T5) was notable compared with the control group, which received standard care. The apparent recurrence of pain at 18 months in both groups was not anticipated and needs to be further investigated.
More specific objective assessments of pain on active shoulder movement identified distinct patterns of pain that could not be isolated using the BPI-SF alone. The incidence and localization of pain on movement differed between the population of women who received a specific exercise intervention and those who received standard care (Figure 2). Patterns of pain over time fluctuated in the control group, whereas the intervention group reported a linear decrease in pain. Residual pain on shoulder movement remained apparent in both groups at 18-months postradiation, but that finding was not reflected in the BPI-SF results. The literature supports our findings on persistent pain among breast cancer survivors,3,7,8,28-30 and in our study of young women carefully screened and excluded for pre-existent shoulder conditions or comorbid medical conditions, recurrent articular pain was nonetheless prevalent. It seems that unidentified or multiple factors may be part of the etiology of pain in this young adult cohort.
Although the BPI-SF is a generic measurement tool commonly used to assess and measure cancer patients’ pain levels, the lack of variance in our BPI-SF severity and interference outcomes over time (T1-T6) (Table 1, Table 2), the variety of “other” unrelated regions (Figure 1) identified by the BPI-SF, and the contrast in our findings on specific physical examination emphasize the potential limitations of this clinical tool.
Moreover, the BPI-SF has not been validated specifically for breast cancer. Harrington and colleagues have recommended using the BPI-SF to assess pain in women with breast cancer,31 but the use of a more multidimensional measurement tool that evaluates axillary, chest, trunk, and upper-limb pain may prove to be more valuable in this population.
Limitations
Recruitment of young adult women was difficult because of our stringent inclusion criteria, the long-term follow-up, and the relatively small population of breast cancer patients in this age demographic. Therefore, the duration of the recruitment phase, despite our having access to a specialized young adult and adolescent clinic in our institute, greatly surpassed the expectations we had when we designed the study. In addition, there remains an inherent bias in participants who accept participation in a study that includes exercise interventions. Potential participants who exercise regularly or have a positive inclination toward doing exercise are more likely to participate. Despite the prescription of a targeted 12-week upper-limb intervention in this study, the general activity levels of both groups may have had an impact on the significance of this study. In addition, the low adherence to the use of self-reported logs failed to capture the true compliance rates of our participants because their lack of tracking does not indicate failure to comply with the program. The use of weekly or biweekly telephone calls to monitor compliance rates of activity more vigilantly may be used in future studies.
Conclusions
Advances in clinical management of breast cancer have improved survival outcomes, and morbidity over recent years, yet symptoms such as pain remain prevalent in this population. The results of this study showed that a targeted, 12-week upper-limb exercise intervention postradiation transiently improved levels of shoulder pain without a concomitant impact on chronic pain or any positive influence on activities of daily living 18 months posttreatment. Furthermore, future studies should use a variety of measurement tools to evaluate trunk and upper-limb pain in women with breast cancer and investigate the optimal timing of postradiation exercise interventions.
Acknowledgments
The authors thank Hope & Cope, the CURE foundation, and the Jewish General Hospital Foundation/Weekend to End Breast Cancer for providing the financial resources needed to sustain this research study. They also thank the McGill Adolescent and Young Adult program for its continued support. Previous oral presentations of research Muanza TM, et al. Randomized clinical trial of a progressive exercise program for young women with breast cancer undergoing radiation therapy. Int J Radiat Oncol Biol Phys. 2015;93(3):s35-s36.
Breast cancer is one of the most prevalent cancers in women worldwide, with more than 1 million new cases diagnosed annually.1 Prognosis for the disease has improved significantly, but 25% to 60% of women living with breast cancer experience some level of pain ranging from mild to severe, the nature of which can evolve from acute to chronic.2 Pre-, intra-, and post-treatment risk factors have been found to correlate with the development of acute and chronic pain and include young age, type of breast surgery (lumpectomy or total mastectomy), axillary node dissection, radiation therapy, and hormonal therapy.3-5 Chemotherapy, particularly anthracycline- and taxane-based regimens, has also been shown to induce pain, arthralgia, myalgia, and peripheral neuropathy during treatment.6 In particular, postradiation pain may result from subcutaneous fibrosis with fixation to underlying musculature and the development of fibrous flaps in the internal axilla.7 These tissue changes are commonly subclinical, occurring 4 to 12 months postradiation,8 and can progress undetected until pain and upper-limb disability develop.
The presence of persistent pain has a considerable impact on the quality of life in survivors of breast cancer: psychological distress is prevalent (anxiety, depression, worry, fear), the performance of daily activities is diminished (eg, bathing, dressing, preparing meals, shopping), and economic independence is compromised by the inability to work or reduced employment and income. These factors directly and indirectly contribute to an increase in the use of health care services.9,10
The management of pain is often characterized by pharmacologic-related treatment, such as the use of opioids and nonsteroidal anti-inflammatory medications, and nonpharmacologic-related treatment, such as exercise. Empirical evidence has shown that rehabilitative exercise programs, which commonly include a combination of resistance training and aerobic exercises, can effectively reduce pain in breast cancer survivors.10-12 Women living with breast cancer who are directed to rehabilitative exercise programs experience an improvement not only in pain levels but also in their ability to engage in activities of daily living, in their psychological health, and in their overall quality of life.13-15 However, despite evidence to support exercise programs to reduce pain related to breast cancer treatment, residual pain and upper-limb discomfort are common complaints in breast cancer survivors, and there is little focus on the duration of effectiveness of such programs for reducing pain after treatment for breast cancer. The objective of this study was to determine if an exercise program initiated postradiation would improve long-term pain levels in a carefully selected population of young women who were living with breast cancer and had no history of shoulder pathology or significant treatment complications.
Methods
Design
We used a pilot randomized control trial to compare the long-term effectiveness of a 12-week postradiation exercise program versus standard care on residual pain levels in young women (aged 18-45 years) living with breast cancer. The program was initiated 3 to 4 weeks postradiation to allow for acute inflammatory reactions to subside. Pain severity and interference were assessed using the Brief Pain Inventory-Short Form (BPI-SF), a tool for assessing cancer pain.16,17 Pain levels for isolated shoulder movements were also recorded on examination by a physical therapist. All measures were collected at 6 time points (T1-T6): postsurgery and preradiation (T1, baseline), postradiation and preintervention (T2), and 4 points during an 18-month period postradiation (T3-T6 at 3, 6, 12, and 18 months postradiation).
Sample
Young women living with breast cancer who met our eligibility criteria were identified from 2 clinics at the Jewish General Hospital – the Segal Cancer Center and the Department of Radiation Oncology in Montréal, Québec, Canada. Inclusion criteria included women with a diagnosis of stage I to stage III breast cancer, who were 18 to 45 years old, were scheduled for postoperative adjuvant radiation therapy, had an Eastern Cooperative Oncology Group Performance Status of 0 or 1 (normal ambulatory function, minimal symptoms), and who consented to participate in the study. Exclusion criteria included women with a metastatic (stage IV) diagnosis; significant musculoskeletal, cardiac, pulmonary, or metabolic comorbidities that would not allow for participation in physical activity; a previous breast cancer diagnosis with treatment to the ipsilateral or contralateral sides; postsurgical lymphedema; postsurgical capsulitis, tendonitis, or other shoulder inflammatory complications; and any contraindication to exercise. The recruitment goal was outlined as 50 patients per group; however, a protracted accrual time because of the stringent study criteria yielded a sample of 29 and 30 patients for the intervention and control groups, respectively, which was sufficient for significant testing of differences between the 2 study groups.18
Variables and measures
Clinical characteristics. We used standardized questions and chart review to document the participants’ clinical characteristics and to capture information on the following: the stage and subtype of breast cancer, hormonal and human epidermal growth factor receptors (HER2) (estrogen receptor, progesterone receptor, and HER2 status), extent of surgery (lumpectomy or total mastectomy), and other modalities of treatment (eg, chemotherapy, radiation therapy).
Pain assessment. The BPI-SF was used to assess participants’ cancer-related pain. Pain severity ranged from 0 (no pain), 1 to 4 (mild pain), 5 to 6 (moderate pain), to 7 to 10 (severe pain).18,19 The questionnaire also identifies the pain interference in daily activities using a Likert scale ranging from 0 (Does not interfere) to 10 (Completely interferes) in the following 7 domains or subscales: General Activity, Walking, Mood, Sleep, Work, Relations with Others, and Enjoyment of Life.16 For the purpose of this study, mean scores were tabulated using both pain intensity and interference scales.
Another important component of the BPI-SF instructs participants to localize pain by means of a body diagram. For purpose of analysis, 3 pain regions were established: shoulder girdle/chest wall on the affected side; neck and other upper extremity, including hand(s), forearm(s), wrist(s), and finger(s); and other regions, including abdominal discomfort, leg(s), hip(s), knee(s), ankle(s), lower back, and feet. In addition, pain levels on movement (Yes/No) were recorded for isolated shoulder flexion, abduction, and horizontal abduction (sitting and standing). The measurements were completed by a single physical therapist throughout the course of the study to minimize variance.
Procedure
The study protocol was approved by the Research Ethics Board at the Jewish General Hospital. Recruitment occurred from 2011 through 2015. The research was in accordance with the ethical standards of the responsible committee on human experimentation. Eligible women were recruited by the research coordinator who described the purpose, risks, and benefits of the study; advised on confidentiality, data collection, and intervention allocation procedures; and highlighted voluntary participation. The research coordinator addressed any concerns on the part of the participants before obtaining their written informed consent. Random allocation to the intervention and control groups was established using a web-based randomization plan generator (www.randomization.com). A single individual was responsible for the randomization process, and treatment assignments were revealed after each participant’s name had been entered. A physical therapist performed 6 sequential evaluations (T1-T6) at the time of participants’ medical follow-up appointments.
Intervention
The 12-week exercise intervention started 3 weeks postradiation and was composed of an initial 6-week program of low-level cardiovascular and resistance exercises that progressed to a set of more advanced exercises for the remaining 6 weeks. Participants were instructed to warm up for at least 10 minutes with a cardiovascular exercise of their choice (eg, a recumbent cross trainer, walking, or stairs) before doing a combined strength, endurance, and stretching exercise program for the upper body.20 The final portion of the exercise intervention included a period of light cool-down. Weight training resistance levels were based on a maximum 8 to 10 repetitions for strength and a maximum of 20 repetitions for endurance training exercises, which progressed gradually over the course of the 12-week exercise program to ensure participant safety.21,22 Participants in the intervention group were supervised at least once a week by an exercise physiologist at a center for oncology patients (Hope & Cope Wellness Centre), and patients were encouraged to perform the program at home 2 to 3 times a week. Those who were not able to exercise consistently at the center were provided with equipment and instructed on how to do the program safely at home.
By comparison, the control group received standard care, which included advice on the benefits of an active lifestyle, including exercise, but without a specific intervention. Participants were not restricted in their physical activity and/or sport participation levels, and their weekly activity levels were calculated using the Metabolic Equivalent of Task and recorded at each of the 6 time points.
Statistical analysis
Descriptive statistics were used to examine participant characteristics. The quantitative data collected through the BPI-SF measures were analyzed with JMP software (version 11.2; SAS Institute, Cary, NC). Continuous variables were tested for statistical significance (P ≤ .05) through the chi-square (categorical), analysis of variance, and nonparametric Wilcoxon tests. The analyses did not include missing data.
Results
A total of 59 young women were randomized into the intervention (n = 29) and control (n = 30) groups. Of those, 2 participants dropped out of the study because of family and time constraints, and 3 participants died, 2 from the control and 1 from the intervention group, after subsequently developing metastatic disease. Baseline data including comparative tumor characteristics, surgical interventions, and treatment interventions have been published in relation to other elements of this study.23,24 The participants had a mean age of 39.2 years (standard deviation [SD], 5.0). More than half of them had an invasive ductal carcinoma (69.5%) and were estrogen positive (78.0%), progesterone positive (74.6%), or HER2 positive (20.3%), whereas 10.2% were triple negative. Most of the participants had undergone breast-sparing procedures (86.4% lumpectomy), and 18.6% had a total mastectomy. By random chance, the intervention group had higher rates of total mastectomy (24.4% and 13.3%, respectively) and surgical reconstruction (12.2% and 6.7%, respectively) compared with the control group. Most of the women (71.2%) received chemotherapy, and all received radiation therapy. In the intervention group, 37.2% received radiation therapy localized to the axilla, and 88% received a boost of radiation to the surgical bed. Self-reported exercise diaries were returned by 15 of the 29 intervention participants, and training frequencies among them varied significantly (1-6 times a week).
The findings showed that there was little variance between the intervention and control groups in BPI-SF severity scores from T1 to T6, so the means and SDs of the BPI-SF scores were grouped at 6 time points (Table 1). There was no statistically significant difference between baseline measures at T1 (1.68; SD, 1.17) and measures at 18 months postintervention (T6: 1.46; SD, 1.37). At baseline, 87.7% of the women reported no pain (31.5%) or mild levels of pain (55.6%), and 13% reported moderate or severe pain. Over the duration of the study from T1 to T6, these primarily low levels of pain (BPI-SF, 0-4) remained consistent with a favorable shift toward having no pain (T1: 31.5%; T6: 24.4%). By 18 months postintervention, 95.7% of women reported no or mild pain, with 4.9% reporting moderate pain.
Similarly, there was little variance over time (T1-T6) and no statistically significant differences between the 2 groups in BPI-SF–measured levels of pain interference in daily activities (Table 2). Moreover, a domain analysis showed that there were no statistically significant differences in pain interference scores when comparing the type and extent of surgery (total mastectomy: 0.59 [1.17]; lumpectomy: 0.94 [1.96]). By chance – and not related directly to the objectives of this study – there was a statistically significant difference between the intervention and control groups in the interference of pain on the Enjoyment of Life domain in favor of the control group.
The sites of pain captured by the BPI-SF shed light on the preceding findings (Figure 1). At baseline (T1, postsurgery and preradiation), 37.0% of participants reported pain in the shoulder girdle–chest wall region, whereas 20.4% reported pain in the general neck–upper extremity region and 50% in other regions. Postradiation, shoulder girdle–chest wall pain was identified as the highest reported site of pain (49.1%; T2, postradiation and preintervention) and remained elevated at 3 months (T3) and 6 months (T4) postradiation (46.9% and 45.5%, respectively). At 12 and 18 months postradiation (T5 and T6), the principal focus of pain shifted once again to “other” regions at 30% and 32.5%, respectively, and the neck–upper extremity region at 10% and 15%, respectively. Shoulder girdle–chest wall pain concomitantly improved at those time points (15% and 25% respectively) but was not eliminated.
Pain levels recorded on physical examination for isolated shoulder range of movements were recently published,24 and they have been abbreviated and reproduced in this paper (Figure 2) to allow for a comparison of findings between the exercise intervention group and the control group to help determine the sensitivity of these tools for use in breast cancer patients. At baseline, pain levels with active movement were noted to be slightly greater in the intervention group for flexion and abduction.
Following the intervention, at 3 and 6 months postradiation (T3 and T4), the intervention group showed a steady decrease in pain levels in flexion and abduction, whereas the control group showed a 5-fold increase in pain with horizontal abduction. Furthermore, participants in the intervention group reported having no pain on movement 12 months postradiation (T5); however, recurrence of pain was apparent with all shoulder movements by 18 months postradiation (T6) in both the intervention and control groups.
Discussion
Previous studies have hypothesized that younger age (18-39 years), adjuvant radiotherapy, and axillary node dissection are risk factors for chronic pain in breast cancer survivors.22,25 Persistent pain is prevalent in 12% to 51% of breast cancer survivors, with up to one-third experiencing some pain more than 5 years after treatment,26,27 and our study outcomes concur with those findings. In our study, pain, as measured by the BPI-SF, was found to persist for most participants (75.6%) after the 18-month follow-up. The results of our trial showed that a 12-week exercise intervention administered postsurgery and postradiation had no statistically significant effect on long-term (18 months) pain severity and its interference in daily life. It is worth noting that body regions that had not been directly related to either surgical or radiation treatment for breast cancer were commonly identified as areas of pain but were not specifically targeted by our intervention. However, focusing on pain severity (BPI-SF), our findings suggest that the benefits of targeted upper-extremity exercise on pain in the intermediate time course of follow-up (T3, T4, and T5) was notable compared with the control group, which received standard care. The apparent recurrence of pain at 18 months in both groups was not anticipated and needs to be further investigated.
More specific objective assessments of pain on active shoulder movement identified distinct patterns of pain that could not be isolated using the BPI-SF alone. The incidence and localization of pain on movement differed between the population of women who received a specific exercise intervention and those who received standard care (Figure 2). Patterns of pain over time fluctuated in the control group, whereas the intervention group reported a linear decrease in pain. Residual pain on shoulder movement remained apparent in both groups at 18-months postradiation, but that finding was not reflected in the BPI-SF results. The literature supports our findings on persistent pain among breast cancer survivors,3,7,8,28-30 and in our study of young women carefully screened and excluded for pre-existent shoulder conditions or comorbid medical conditions, recurrent articular pain was nonetheless prevalent. It seems that unidentified or multiple factors may be part of the etiology of pain in this young adult cohort.
Although the BPI-SF is a generic measurement tool commonly used to assess and measure cancer patients’ pain levels, the lack of variance in our BPI-SF severity and interference outcomes over time (T1-T6) (Table 1, Table 2), the variety of “other” unrelated regions (Figure 1) identified by the BPI-SF, and the contrast in our findings on specific physical examination emphasize the potential limitations of this clinical tool.
Moreover, the BPI-SF has not been validated specifically for breast cancer. Harrington and colleagues have recommended using the BPI-SF to assess pain in women with breast cancer,31 but the use of a more multidimensional measurement tool that evaluates axillary, chest, trunk, and upper-limb pain may prove to be more valuable in this population.
Limitations
Recruitment of young adult women was difficult because of our stringent inclusion criteria, the long-term follow-up, and the relatively small population of breast cancer patients in this age demographic. Therefore, the duration of the recruitment phase, despite our having access to a specialized young adult and adolescent clinic in our institute, greatly surpassed the expectations we had when we designed the study. In addition, there remains an inherent bias in participants who accept participation in a study that includes exercise interventions. Potential participants who exercise regularly or have a positive inclination toward doing exercise are more likely to participate. Despite the prescription of a targeted 12-week upper-limb intervention in this study, the general activity levels of both groups may have had an impact on the significance of this study. In addition, the low adherence to the use of self-reported logs failed to capture the true compliance rates of our participants because their lack of tracking does not indicate failure to comply with the program. The use of weekly or biweekly telephone calls to monitor compliance rates of activity more vigilantly may be used in future studies.
Conclusions
Advances in clinical management of breast cancer have improved survival outcomes, and morbidity over recent years, yet symptoms such as pain remain prevalent in this population. The results of this study showed that a targeted, 12-week upper-limb exercise intervention postradiation transiently improved levels of shoulder pain without a concomitant impact on chronic pain or any positive influence on activities of daily living 18 months posttreatment. Furthermore, future studies should use a variety of measurement tools to evaluate trunk and upper-limb pain in women with breast cancer and investigate the optimal timing of postradiation exercise interventions.
Acknowledgments
The authors thank Hope & Cope, the CURE foundation, and the Jewish General Hospital Foundation/Weekend to End Breast Cancer for providing the financial resources needed to sustain this research study. They also thank the McGill Adolescent and Young Adult program for its continued support. Previous oral presentations of research Muanza TM, et al. Randomized clinical trial of a progressive exercise program for young women with breast cancer undergoing radiation therapy. Int J Radiat Oncol Biol Phys. 2015;93(3):s35-s36.
1. World Health Organization. Breast cancer: prevention and control. www.who.int/cancer/detection/breastcancer/en/. Updated 2017. Accessed September 16, 2016.
2. Andersen KG, Kehlet H. Persistent pain after breast cancer treatment: a critical review of risk factors and strategies for prevention. J Pain. 2011;12(7):725-746.
3. Ernst MF, Voogd AC, Balder W, Klinkenbijl JH, Roukema JA. Early and late morbidity associated with axillary levels I-III dissection in breast cancer. J Surg Oncol. 2002;79(3):151-155; discussion 156.
4. Gulluoglu BM, Cingi A, Cakir T, Gercek A, Barlas A, Eti Z. Factors related to post-treatment chronic pain in breast cancer survivors: the interference of pain with life functions. Int J Fertil Womens Med. 2006;51(2):75-82.
5. Jung BF, Ahrendt GM, Oaklander AL, Dworkin RH. Neuropathic pain following breast cancer surgery: proposed classification and research update. Pain. 2003;104(1-2):1-13.
6. Saibil S, Fitzgerald B, Freedman OC, et al. Incidence of taxane-induced pain and distress in patients receiving chemotherapy for early-stage breast cancer: a retrospective, outcomes-based survey. Curr Oncol. 2010;17(4):42-47.
7. Tengrup I, Tennvall-Nittby L, Christiansson I, Laurin M. Arm morbidity after breast-conserving therapy for breast cancer. Acta Oncol. 2000;39(3):393-397.
8. Johansen J, Overgaard J, Blichert-Toft M, Overgaard M. Treatment of morbidity associated with the management of the axilla in breast-conserving therapy. Acta Oncol. 2000;39(3):349-354.
9. Mittmann N, Porter JM, Rangrej J, et al. Health system costs for stage-specific breast cancer: a population-based approach. Curr Oncol. 2014;21(6):281-293.
10. Page A. Keeping patients safe: transforming the work environment of nurses. Washington, DC: National Academies Press; 2004.
11. McNeely ML, Campbell K, Ospina M, et al. Exercise interventions for upper-limb dysfunction due to breast cancer treatment. Cochrane Database Syst Rev. 2010;(6):CD005211. doi:10.1002/14651858.CD005211.pub2
12. Wong P, Muanza T, Hijal T, et al. Effect of exercise in reducing breast and chest-wall pain in patients with breast cancer: a pilot study. Curr Oncol. 2012;19(3):e129-e135.
13. Fernández-Lao C, Cantarero-Villanueva I, Fernández-de-Las-Peñas C, del Moral-Ávila R, Castro-Sánchez AM, Arroyo-Morales M. Effectiveness of a multidimensional physical therapy program on pain, pressure hypersensitivity, and trigger points in breast cancer survivors: a randomized controlled clinical trial. Clin J Pain. 2012;28(2):113-121.
14. Courneya KS, Mackey JR, Bell GJ, Jones LW, Field CJ, Fairey AS. Randomized controlled trial of exercise training in postmenopausal breast cancer survivors: cardiopulmonary and quality of life outcomes. J Clin Oncol. 2003;21(9):1660-1668.
15. Segal R, Evans W, Johnson D, et al. Structured exercise improves physical functioning in women with stages I and II breast cancer: results of a randomized controlled trial. J Clin Oncol. 2001;19(3):657-665.
16. Cleeland CS, Ryan KM. Pain assessment: global use of the Brief Pain Inventory. Ann Acad Med Singapore. 1994;23(2):129-138.
17. Kumar SP. Utilization of Brief Pain Inventory as an assessment tool for pain in patients with cancer: a focused review. Indian J Palliat Care. 2011;17(2):108-115.
18. Van Voorhis CRW, Morgan BL. Understanding power and rules of thumb for determining sample sizes. Tutor Quant Methods Psychol. 2007;3(2):43-50.
19. Serlin RC, Mendoza TR, Nakamura Y, Edwards KR, Cleeland CS. When is cancer pain mild, moderate or severe? Grading pain severity by its interference with function. Pain. 1995;61(2):277-284.
20. Lee TS, Kilbreath SL, Refshauge KM, Pendlebury SC, Beith JM, Lee MJ. Pectoral stretching program for women undergoing radiotherapy for breast cancer. Breast Cancer Res Treat. 2007;102(3):313-321.
21. Schmitz KH, Courneya KS, Matthews C, et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42(7):1409-1426.
22. Pollock ML, Gaesser GA, Butcher JD, et al. ACSM position stand: the recommended quantity and quality of exercise for developing and maintaining cardiorespiratory and muscular fitness, and flexibility in healthy adults. Med Sci Sports Exerc. 1998;30(6):975-991.
23. Ibrahim M, Muanza T, Smirnow N, et al. Time course of upper limb function and return-to-work post-radiotherapy in young adults with breast cancer: a pilot randomized control trial on effects of targeted exercise program. J Cancer Surviv. 2017;11(6):791-799.
24. Ibrahim M, Muanza T, Smirnow N, et al. A pilot randomized controlled trial on the effects of a progressive exercise program on the range of motion and upper extremity grip strength in young adults with breast cancer. Clin Breast Cancer. 2018;18(1):e55-e64.
25. Gärtner R, Jensen MB, Nielsen J, Ewertz M, Kroman N, Kehlet H. Prevalence of and factors associated with persistent pain following breast cancer surgery. JAMA. 2009;302(18):1985-1992.
26. Hayes SC, Johansson K, Stout NL, et al. Upper-body morbidity after breast cancer: incidence and evidence for evaluation, prevention, and management within a prospective surveillance model of care. Cancer. 2012;118(suppl 8):2237-2249.
27. Kärki A, Simonen R, Mälkiä E, Selfe J. Impairments, activity limitations and participation restrictions 6 and 12 months after breast cancer operation. J Rehabil Med. 2005;37(3):180-188.
28. Katz J, Poleshuck EL, Andrus CH, et al. Risk factors for acute pain and its persistence following breast cancer surgery. Pain. 2005;119(1-3):16-25.
29. Tasmuth T, von Smitten K, Hietanen P, Kataja M, Kalso E. Pain and other symptoms after different treatment modalities of breast cancer. Ann Oncol. 1995;6(5):453-459.
30. Whelan TJ, Levine M, Julian J, Kirkbride P, Skingley P. The effects of radiation therapy on quality of life of women with breast carcinoma: results of a randomized trial. Ontario Clinical Oncology Group. Cancer. 2000;88(10):2260-2266.
31. Harrington S, Gilchrist L, Sander A. Breast cancer EDGE task force outcomes: clinical measures of pain. Rehabil Oncol. 2014;32(1):13-21.
1. World Health Organization. Breast cancer: prevention and control. www.who.int/cancer/detection/breastcancer/en/. Updated 2017. Accessed September 16, 2016.
2. Andersen KG, Kehlet H. Persistent pain after breast cancer treatment: a critical review of risk factors and strategies for prevention. J Pain. 2011;12(7):725-746.
3. Ernst MF, Voogd AC, Balder W, Klinkenbijl JH, Roukema JA. Early and late morbidity associated with axillary levels I-III dissection in breast cancer. J Surg Oncol. 2002;79(3):151-155; discussion 156.
4. Gulluoglu BM, Cingi A, Cakir T, Gercek A, Barlas A, Eti Z. Factors related to post-treatment chronic pain in breast cancer survivors: the interference of pain with life functions. Int J Fertil Womens Med. 2006;51(2):75-82.
5. Jung BF, Ahrendt GM, Oaklander AL, Dworkin RH. Neuropathic pain following breast cancer surgery: proposed classification and research update. Pain. 2003;104(1-2):1-13.
6. Saibil S, Fitzgerald B, Freedman OC, et al. Incidence of taxane-induced pain and distress in patients receiving chemotherapy for early-stage breast cancer: a retrospective, outcomes-based survey. Curr Oncol. 2010;17(4):42-47.
7. Tengrup I, Tennvall-Nittby L, Christiansson I, Laurin M. Arm morbidity after breast-conserving therapy for breast cancer. Acta Oncol. 2000;39(3):393-397.
8. Johansen J, Overgaard J, Blichert-Toft M, Overgaard M. Treatment of morbidity associated with the management of the axilla in breast-conserving therapy. Acta Oncol. 2000;39(3):349-354.
9. Mittmann N, Porter JM, Rangrej J, et al. Health system costs for stage-specific breast cancer: a population-based approach. Curr Oncol. 2014;21(6):281-293.
10. Page A. Keeping patients safe: transforming the work environment of nurses. Washington, DC: National Academies Press; 2004.
11. McNeely ML, Campbell K, Ospina M, et al. Exercise interventions for upper-limb dysfunction due to breast cancer treatment. Cochrane Database Syst Rev. 2010;(6):CD005211. doi:10.1002/14651858.CD005211.pub2
12. Wong P, Muanza T, Hijal T, et al. Effect of exercise in reducing breast and chest-wall pain in patients with breast cancer: a pilot study. Curr Oncol. 2012;19(3):e129-e135.
13. Fernández-Lao C, Cantarero-Villanueva I, Fernández-de-Las-Peñas C, del Moral-Ávila R, Castro-Sánchez AM, Arroyo-Morales M. Effectiveness of a multidimensional physical therapy program on pain, pressure hypersensitivity, and trigger points in breast cancer survivors: a randomized controlled clinical trial. Clin J Pain. 2012;28(2):113-121.
14. Courneya KS, Mackey JR, Bell GJ, Jones LW, Field CJ, Fairey AS. Randomized controlled trial of exercise training in postmenopausal breast cancer survivors: cardiopulmonary and quality of life outcomes. J Clin Oncol. 2003;21(9):1660-1668.
15. Segal R, Evans W, Johnson D, et al. Structured exercise improves physical functioning in women with stages I and II breast cancer: results of a randomized controlled trial. J Clin Oncol. 2001;19(3):657-665.
16. Cleeland CS, Ryan KM. Pain assessment: global use of the Brief Pain Inventory. Ann Acad Med Singapore. 1994;23(2):129-138.
17. Kumar SP. Utilization of Brief Pain Inventory as an assessment tool for pain in patients with cancer: a focused review. Indian J Palliat Care. 2011;17(2):108-115.
18. Van Voorhis CRW, Morgan BL. Understanding power and rules of thumb for determining sample sizes. Tutor Quant Methods Psychol. 2007;3(2):43-50.
19. Serlin RC, Mendoza TR, Nakamura Y, Edwards KR, Cleeland CS. When is cancer pain mild, moderate or severe? Grading pain severity by its interference with function. Pain. 1995;61(2):277-284.
20. Lee TS, Kilbreath SL, Refshauge KM, Pendlebury SC, Beith JM, Lee MJ. Pectoral stretching program for women undergoing radiotherapy for breast cancer. Breast Cancer Res Treat. 2007;102(3):313-321.
21. Schmitz KH, Courneya KS, Matthews C, et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42(7):1409-1426.
22. Pollock ML, Gaesser GA, Butcher JD, et al. ACSM position stand: the recommended quantity and quality of exercise for developing and maintaining cardiorespiratory and muscular fitness, and flexibility in healthy adults. Med Sci Sports Exerc. 1998;30(6):975-991.
23. Ibrahim M, Muanza T, Smirnow N, et al. Time course of upper limb function and return-to-work post-radiotherapy in young adults with breast cancer: a pilot randomized control trial on effects of targeted exercise program. J Cancer Surviv. 2017;11(6):791-799.
24. Ibrahim M, Muanza T, Smirnow N, et al. A pilot randomized controlled trial on the effects of a progressive exercise program on the range of motion and upper extremity grip strength in young adults with breast cancer. Clin Breast Cancer. 2018;18(1):e55-e64.
25. Gärtner R, Jensen MB, Nielsen J, Ewertz M, Kroman N, Kehlet H. Prevalence of and factors associated with persistent pain following breast cancer surgery. JAMA. 2009;302(18):1985-1992.
26. Hayes SC, Johansson K, Stout NL, et al. Upper-body morbidity after breast cancer: incidence and evidence for evaluation, prevention, and management within a prospective surveillance model of care. Cancer. 2012;118(suppl 8):2237-2249.
27. Kärki A, Simonen R, Mälkiä E, Selfe J. Impairments, activity limitations and participation restrictions 6 and 12 months after breast cancer operation. J Rehabil Med. 2005;37(3):180-188.
28. Katz J, Poleshuck EL, Andrus CH, et al. Risk factors for acute pain and its persistence following breast cancer surgery. Pain. 2005;119(1-3):16-25.
29. Tasmuth T, von Smitten K, Hietanen P, Kataja M, Kalso E. Pain and other symptoms after different treatment modalities of breast cancer. Ann Oncol. 1995;6(5):453-459.
30. Whelan TJ, Levine M, Julian J, Kirkbride P, Skingley P. The effects of radiation therapy on quality of life of women with breast carcinoma: results of a randomized trial. Ontario Clinical Oncology Group. Cancer. 2000;88(10):2260-2266.
31. Harrington S, Gilchrist L, Sander A. Breast cancer EDGE task force outcomes: clinical measures of pain. Rehabil Oncol. 2014;32(1):13-21.
Testicular Pain Leads to a Rare Diagnosis
A 26-year-old man presented to uroligy clinicians with right testicular pain and a right epididymal mass. It was a challenge to diagnose the cause—until he revealed some essential clues.
The differential diagnosis included testicular malignancy and lymphoma. However, tumor markers were within normal limits. Tests for HIV and syphilis were negative. The clinicians also considered granulomatous or chronic orchitis, but after treatment with nonsteroidal anti-inflammatory drugs, the pain and palpable epididymal mass had resolved. Then, follow-up testicular ultrasound images showed new diffuse heterogeneous hypoechoic lesions in the right testis.
The patient elected to have a right radical orchiectomy with sperm cryopreserved. He recovered well, and semen analysis did not show any abnormalities.
Pathology of the surgical specimen revealed necrotizing and nonnecrotizing granulomas. At this point, the patient recollected that he had developed cervical lymphadenopathy and oral ulcers several weeks after traveling to South America, 8 months before presenting with the testicular symptoms.
Combined with another clue—frequent exposure to cats during his South America trip—the patient’s symptoms now created a clearer picture. He had been diagnosed with toxoplasmosis at the time but had not received treatment because he was immunocompetent. The patient’s symptoms had resolved spontaneously, and he said he had been in his usual health between then and when he developed the testicular pain.
Based on this new information, the clinicians conducted immunohistochemical tests, which revealed isolated cysts about 20 µm in diameter, confirming a diagnosis of testicular toxoplasmosis. They started him on systemic toxoplasmosis treatment; he has been in good health since.
The clinicians note that toxoplasmosis is highly prevalent, infecting up to 30% of the world’s population. Cat feces is one source of infection with Toxoplasma gondii, which is typically asymptomatic. In immunocompetent patients, it tends to present as an acute infection that is benign and self-limited.
Only a few cases of testicular toxoplasmosis have been reported in the literature, and all have been in immunocompromised patients. Because this patient was immunocompetent, the case is unique, being the only one reported as yet. Given the immunocompetence, the clinicians say, the patient demonstrates the need for clinicians to have a high index of suspicion.
Source:
Wong V, Amarasekera C, Kundu S. BMJ Case Rep. 2018;2018. pii: bcr-2018-224962.
A 26-year-old man presented to uroligy clinicians with right testicular pain and a right epididymal mass. It was a challenge to diagnose the cause—until he revealed some essential clues.
The differential diagnosis included testicular malignancy and lymphoma. However, tumor markers were within normal limits. Tests for HIV and syphilis were negative. The clinicians also considered granulomatous or chronic orchitis, but after treatment with nonsteroidal anti-inflammatory drugs, the pain and palpable epididymal mass had resolved. Then, follow-up testicular ultrasound images showed new diffuse heterogeneous hypoechoic lesions in the right testis.
The patient elected to have a right radical orchiectomy with sperm cryopreserved. He recovered well, and semen analysis did not show any abnormalities.
Pathology of the surgical specimen revealed necrotizing and nonnecrotizing granulomas. At this point, the patient recollected that he had developed cervical lymphadenopathy and oral ulcers several weeks after traveling to South America, 8 months before presenting with the testicular symptoms.
Combined with another clue—frequent exposure to cats during his South America trip—the patient’s symptoms now created a clearer picture. He had been diagnosed with toxoplasmosis at the time but had not received treatment because he was immunocompetent. The patient’s symptoms had resolved spontaneously, and he said he had been in his usual health between then and when he developed the testicular pain.
Based on this new information, the clinicians conducted immunohistochemical tests, which revealed isolated cysts about 20 µm in diameter, confirming a diagnosis of testicular toxoplasmosis. They started him on systemic toxoplasmosis treatment; he has been in good health since.
The clinicians note that toxoplasmosis is highly prevalent, infecting up to 30% of the world’s population. Cat feces is one source of infection with Toxoplasma gondii, which is typically asymptomatic. In immunocompetent patients, it tends to present as an acute infection that is benign and self-limited.
Only a few cases of testicular toxoplasmosis have been reported in the literature, and all have been in immunocompromised patients. Because this patient was immunocompetent, the case is unique, being the only one reported as yet. Given the immunocompetence, the clinicians say, the patient demonstrates the need for clinicians to have a high index of suspicion.
Source:
Wong V, Amarasekera C, Kundu S. BMJ Case Rep. 2018;2018. pii: bcr-2018-224962.
A 26-year-old man presented to uroligy clinicians with right testicular pain and a right epididymal mass. It was a challenge to diagnose the cause—until he revealed some essential clues.
The differential diagnosis included testicular malignancy and lymphoma. However, tumor markers were within normal limits. Tests for HIV and syphilis were negative. The clinicians also considered granulomatous or chronic orchitis, but after treatment with nonsteroidal anti-inflammatory drugs, the pain and palpable epididymal mass had resolved. Then, follow-up testicular ultrasound images showed new diffuse heterogeneous hypoechoic lesions in the right testis.
The patient elected to have a right radical orchiectomy with sperm cryopreserved. He recovered well, and semen analysis did not show any abnormalities.
Pathology of the surgical specimen revealed necrotizing and nonnecrotizing granulomas. At this point, the patient recollected that he had developed cervical lymphadenopathy and oral ulcers several weeks after traveling to South America, 8 months before presenting with the testicular symptoms.
Combined with another clue—frequent exposure to cats during his South America trip—the patient’s symptoms now created a clearer picture. He had been diagnosed with toxoplasmosis at the time but had not received treatment because he was immunocompetent. The patient’s symptoms had resolved spontaneously, and he said he had been in his usual health between then and when he developed the testicular pain.
Based on this new information, the clinicians conducted immunohistochemical tests, which revealed isolated cysts about 20 µm in diameter, confirming a diagnosis of testicular toxoplasmosis. They started him on systemic toxoplasmosis treatment; he has been in good health since.
The clinicians note that toxoplasmosis is highly prevalent, infecting up to 30% of the world’s population. Cat feces is one source of infection with Toxoplasma gondii, which is typically asymptomatic. In immunocompetent patients, it tends to present as an acute infection that is benign and self-limited.
Only a few cases of testicular toxoplasmosis have been reported in the literature, and all have been in immunocompromised patients. Because this patient was immunocompetent, the case is unique, being the only one reported as yet. Given the immunocompetence, the clinicians say, the patient demonstrates the need for clinicians to have a high index of suspicion.
Source:
Wong V, Amarasekera C, Kundu S. BMJ Case Rep. 2018;2018. pii: bcr-2018-224962.
MAb doubles ORR, PFS in rel/ref MM
STOCKHOLM—Adding elotuzumab (E) to treatment with pomalidomide (P) and low-dose dexamethasone (d) can produce “clinically meaningful” results in patients with relapsed/refractory multiple myeloma (MM), according to an investigator for the ELOQUENT-3 trial.
In this phase 2 trial, patients who received EPd had double the overall response rate (ORR) and median progression-free survival (PFS) of patients who received Pd.
Additionally, adverse events (AEs) were comparable between the treatment arms.
Meletios Dimopoulos, MD, of National and Kapodistrian University of Athens in Greece, presented these results as a late-breaking abstract (LB2606) at the 23rd Congress of the European Hematology Association (EHA).
The research was sponsored by Bristol-Myers Squibb.
The ELOQUENT-3 trial enrolled MM patients who had refractory or relapsed and refractory MM. They had to have received lenalidomide and a proteasome inhibitor (PI).
The patients were randomized to receive EPd (n=60) or Pd (n=57) in 28-day cycles until disease progression or unacceptable toxicity.
Pomalidomide was given orally at 4 mg on days 1 to 21 of each cycle. In the Pd arm, dexamethasone was given as a 20 mg (for patients older than 75) or 40 mg (75 and younger) tablet weekly.
In the EPd arm, dexamethasone was split between oral (8 mg, 20 mg, or 40 mg tablets) and intravenous doses (8 mg or 28 mg).
Elotuzumab was given at 10 mg/kg intravenously weekly for the first 2 cycles and 20 mg/kg monthly from cycle 3 on.
Patient characteristics
The patients’ median age was 69 (range, 43-81) in the EPd arm and 66 (range, 36-81) in the Pd arm. They were a median of 4.8 years (EPd) or 4.4 years (Pd) from diagnosis.
The median number of prior therapies was 3 (range, 2-8) in both groups.
Ninety percent of patients in the EPd arm and 84% of those in the Pd arm were refractory to lenalidomide. Seventy-eight percent and 82%, respectively, were refractory to a PI. And 68% and 72%, respectively, were refractory to both lenalidomide and a PI.
Treatment duration
Dr Dimopoulos noted that twice as many patients remained on treatment with EPd compared to Pd at the time of database lock (February 21, 2018). Forty percent of EPd patients (n=24) and 20% of Pd patients (n=11) were still on treatment at that time.
Patients’ primary reason for treatment discontinuation was disease progression—43% of EPd recipients and 56% of Pd recipients. Two percent of EPd recipients and 4% of Pd recipients withdrew due to treatment-related toxicity. Four percent of patients in the Pd arm (and none in the EPd arm) withdrew due to maximum clinical benefit.
The median number of treatment cycles was 9 (range, 4-13) in the EPd arm and 5 (range, 3-10) in the Pd arm.
Efficacy
The ORR was 53% in the EPd arm and 26% in the Pd arm. The odds ratio was 3.25 (P=0.0029).
Eight percent of patients in the EPd arm had a complete response or stringent complete response, as did 2% of patients in the Pd arm.
The median duration of response was 8.3 months in the Pd arm and has not been reached in the EPd arm.
“Elotuzumab with pomalidomide and dexamethasone showed a significant and clinically meaningful 46% reduction in the risk of progression or death,” Dr Dimopoulos said.
The median PFS was 10.3 months with EPd and 4.7 months with Pd (hazard ratio=0.54, P=0.0078).
Although overall survival data are not yet mature, there was a trend favoring EPd over Pd (hazard ratio=0.62). There were 13 deaths in the EPd arm and 18 deaths in the Pd arm.
Safety
Dr Dimopoulos said AEs were comparable between the treatment arms. He pointed out that neutropenia was less common with EPd compared to Pd, despite similar pomalidomide dose intensity. And exposure-adjusted hematologic AEs and infections were lower with EPd than with Pd.
Ninety-seven percent of patients in the EPd arm and 95% in the Pd arm had at least 1 AE.
Grade 3-4 nonhematologic AEs (in the EPd and Pd arms, respectively) included constipation (2% and 0%), hyperglycemia (8% and 7%), bone pain (3% and 0%), dyspnea (3% and 2%), fatigue (0% and 4%), respiratory tract infection (0% and 2%), and upper respiratory tract infection (0% and 2%).
Grade 3-4 hematologic AEs (in the EPd and Pd arms, respectively) included anemia (10% and 20%), neutropenia (13% and 27%), thrombocytopenia (8% and 5%), and lymphopenia (8% and 2%).
Grade 3-4 AEs of special interest (in the EPd and Pd arms, respectively) included infections (13% and 22%), vascular disorders (3% and 0%), cardiac disorders (7% and 4%), and neoplasms (2% and 11%).
There were 5 grade 5 AEs in the EPd arm and 8 in the Pd arm.
In the EPd arm, grade 5 AEs included infection (n=3), cardiac failure, and general physical health deterioration.
In the Pd arm, grade 5 AEs included malignant neoplasm progression (n=4), infection, multiple organ failure and infection, myocardial infarction, and plasma cell myeloma.
STOCKHOLM—Adding elotuzumab (E) to treatment with pomalidomide (P) and low-dose dexamethasone (d) can produce “clinically meaningful” results in patients with relapsed/refractory multiple myeloma (MM), according to an investigator for the ELOQUENT-3 trial.
In this phase 2 trial, patients who received EPd had double the overall response rate (ORR) and median progression-free survival (PFS) of patients who received Pd.
Additionally, adverse events (AEs) were comparable between the treatment arms.
Meletios Dimopoulos, MD, of National and Kapodistrian University of Athens in Greece, presented these results as a late-breaking abstract (LB2606) at the 23rd Congress of the European Hematology Association (EHA).
The research was sponsored by Bristol-Myers Squibb.
The ELOQUENT-3 trial enrolled MM patients who had refractory or relapsed and refractory MM. They had to have received lenalidomide and a proteasome inhibitor (PI).
The patients were randomized to receive EPd (n=60) or Pd (n=57) in 28-day cycles until disease progression or unacceptable toxicity.
Pomalidomide was given orally at 4 mg on days 1 to 21 of each cycle. In the Pd arm, dexamethasone was given as a 20 mg (for patients older than 75) or 40 mg (75 and younger) tablet weekly.
In the EPd arm, dexamethasone was split between oral (8 mg, 20 mg, or 40 mg tablets) and intravenous doses (8 mg or 28 mg).
Elotuzumab was given at 10 mg/kg intravenously weekly for the first 2 cycles and 20 mg/kg monthly from cycle 3 on.
Patient characteristics
The patients’ median age was 69 (range, 43-81) in the EPd arm and 66 (range, 36-81) in the Pd arm. They were a median of 4.8 years (EPd) or 4.4 years (Pd) from diagnosis.
The median number of prior therapies was 3 (range, 2-8) in both groups.
Ninety percent of patients in the EPd arm and 84% of those in the Pd arm were refractory to lenalidomide. Seventy-eight percent and 82%, respectively, were refractory to a PI. And 68% and 72%, respectively, were refractory to both lenalidomide and a PI.
Treatment duration
Dr Dimopoulos noted that twice as many patients remained on treatment with EPd compared to Pd at the time of database lock (February 21, 2018). Forty percent of EPd patients (n=24) and 20% of Pd patients (n=11) were still on treatment at that time.
Patients’ primary reason for treatment discontinuation was disease progression—43% of EPd recipients and 56% of Pd recipients. Two percent of EPd recipients and 4% of Pd recipients withdrew due to treatment-related toxicity. Four percent of patients in the Pd arm (and none in the EPd arm) withdrew due to maximum clinical benefit.
The median number of treatment cycles was 9 (range, 4-13) in the EPd arm and 5 (range, 3-10) in the Pd arm.
Efficacy
The ORR was 53% in the EPd arm and 26% in the Pd arm. The odds ratio was 3.25 (P=0.0029).
Eight percent of patients in the EPd arm had a complete response or stringent complete response, as did 2% of patients in the Pd arm.
The median duration of response was 8.3 months in the Pd arm and has not been reached in the EPd arm.
“Elotuzumab with pomalidomide and dexamethasone showed a significant and clinically meaningful 46% reduction in the risk of progression or death,” Dr Dimopoulos said.
The median PFS was 10.3 months with EPd and 4.7 months with Pd (hazard ratio=0.54, P=0.0078).
Although overall survival data are not yet mature, there was a trend favoring EPd over Pd (hazard ratio=0.62). There were 13 deaths in the EPd arm and 18 deaths in the Pd arm.
Safety
Dr Dimopoulos said AEs were comparable between the treatment arms. He pointed out that neutropenia was less common with EPd compared to Pd, despite similar pomalidomide dose intensity. And exposure-adjusted hematologic AEs and infections were lower with EPd than with Pd.
Ninety-seven percent of patients in the EPd arm and 95% in the Pd arm had at least 1 AE.
Grade 3-4 nonhematologic AEs (in the EPd and Pd arms, respectively) included constipation (2% and 0%), hyperglycemia (8% and 7%), bone pain (3% and 0%), dyspnea (3% and 2%), fatigue (0% and 4%), respiratory tract infection (0% and 2%), and upper respiratory tract infection (0% and 2%).
Grade 3-4 hematologic AEs (in the EPd and Pd arms, respectively) included anemia (10% and 20%), neutropenia (13% and 27%), thrombocytopenia (8% and 5%), and lymphopenia (8% and 2%).
Grade 3-4 AEs of special interest (in the EPd and Pd arms, respectively) included infections (13% and 22%), vascular disorders (3% and 0%), cardiac disorders (7% and 4%), and neoplasms (2% and 11%).
There were 5 grade 5 AEs in the EPd arm and 8 in the Pd arm.
In the EPd arm, grade 5 AEs included infection (n=3), cardiac failure, and general physical health deterioration.
In the Pd arm, grade 5 AEs included malignant neoplasm progression (n=4), infection, multiple organ failure and infection, myocardial infarction, and plasma cell myeloma.
STOCKHOLM—Adding elotuzumab (E) to treatment with pomalidomide (P) and low-dose dexamethasone (d) can produce “clinically meaningful” results in patients with relapsed/refractory multiple myeloma (MM), according to an investigator for the ELOQUENT-3 trial.
In this phase 2 trial, patients who received EPd had double the overall response rate (ORR) and median progression-free survival (PFS) of patients who received Pd.
Additionally, adverse events (AEs) were comparable between the treatment arms.
Meletios Dimopoulos, MD, of National and Kapodistrian University of Athens in Greece, presented these results as a late-breaking abstract (LB2606) at the 23rd Congress of the European Hematology Association (EHA).
The research was sponsored by Bristol-Myers Squibb.
The ELOQUENT-3 trial enrolled MM patients who had refractory or relapsed and refractory MM. They had to have received lenalidomide and a proteasome inhibitor (PI).
The patients were randomized to receive EPd (n=60) or Pd (n=57) in 28-day cycles until disease progression or unacceptable toxicity.
Pomalidomide was given orally at 4 mg on days 1 to 21 of each cycle. In the Pd arm, dexamethasone was given as a 20 mg (for patients older than 75) or 40 mg (75 and younger) tablet weekly.
In the EPd arm, dexamethasone was split between oral (8 mg, 20 mg, or 40 mg tablets) and intravenous doses (8 mg or 28 mg).
Elotuzumab was given at 10 mg/kg intravenously weekly for the first 2 cycles and 20 mg/kg monthly from cycle 3 on.
Patient characteristics
The patients’ median age was 69 (range, 43-81) in the EPd arm and 66 (range, 36-81) in the Pd arm. They were a median of 4.8 years (EPd) or 4.4 years (Pd) from diagnosis.
The median number of prior therapies was 3 (range, 2-8) in both groups.
Ninety percent of patients in the EPd arm and 84% of those in the Pd arm were refractory to lenalidomide. Seventy-eight percent and 82%, respectively, were refractory to a PI. And 68% and 72%, respectively, were refractory to both lenalidomide and a PI.
Treatment duration
Dr Dimopoulos noted that twice as many patients remained on treatment with EPd compared to Pd at the time of database lock (February 21, 2018). Forty percent of EPd patients (n=24) and 20% of Pd patients (n=11) were still on treatment at that time.
Patients’ primary reason for treatment discontinuation was disease progression—43% of EPd recipients and 56% of Pd recipients. Two percent of EPd recipients and 4% of Pd recipients withdrew due to treatment-related toxicity. Four percent of patients in the Pd arm (and none in the EPd arm) withdrew due to maximum clinical benefit.
The median number of treatment cycles was 9 (range, 4-13) in the EPd arm and 5 (range, 3-10) in the Pd arm.
Efficacy
The ORR was 53% in the EPd arm and 26% in the Pd arm. The odds ratio was 3.25 (P=0.0029).
Eight percent of patients in the EPd arm had a complete response or stringent complete response, as did 2% of patients in the Pd arm.
The median duration of response was 8.3 months in the Pd arm and has not been reached in the EPd arm.
“Elotuzumab with pomalidomide and dexamethasone showed a significant and clinically meaningful 46% reduction in the risk of progression or death,” Dr Dimopoulos said.
The median PFS was 10.3 months with EPd and 4.7 months with Pd (hazard ratio=0.54, P=0.0078).
Although overall survival data are not yet mature, there was a trend favoring EPd over Pd (hazard ratio=0.62). There were 13 deaths in the EPd arm and 18 deaths in the Pd arm.
Safety
Dr Dimopoulos said AEs were comparable between the treatment arms. He pointed out that neutropenia was less common with EPd compared to Pd, despite similar pomalidomide dose intensity. And exposure-adjusted hematologic AEs and infections were lower with EPd than with Pd.
Ninety-seven percent of patients in the EPd arm and 95% in the Pd arm had at least 1 AE.
Grade 3-4 nonhematologic AEs (in the EPd and Pd arms, respectively) included constipation (2% and 0%), hyperglycemia (8% and 7%), bone pain (3% and 0%), dyspnea (3% and 2%), fatigue (0% and 4%), respiratory tract infection (0% and 2%), and upper respiratory tract infection (0% and 2%).
Grade 3-4 hematologic AEs (in the EPd and Pd arms, respectively) included anemia (10% and 20%), neutropenia (13% and 27%), thrombocytopenia (8% and 5%), and lymphopenia (8% and 2%).
Grade 3-4 AEs of special interest (in the EPd and Pd arms, respectively) included infections (13% and 22%), vascular disorders (3% and 0%), cardiac disorders (7% and 4%), and neoplasms (2% and 11%).
There were 5 grade 5 AEs in the EPd arm and 8 in the Pd arm.
In the EPd arm, grade 5 AEs included infection (n=3), cardiac failure, and general physical health deterioration.
In the Pd arm, grade 5 AEs included malignant neoplasm progression (n=4), infection, multiple organ failure and infection, myocardial infarction, and plasma cell myeloma.