LayerRx Mapping ID
205
Slot System
Featured Buckets
Featured Buckets Admin

Clip closure reduces postop bleeding risk after proximal polyp resection

Article Type
Changed
Mon, 09/30/2019 - 17:12

Closing mucosal defects with hemoclips after endoscopic resection of large polyps in the proximal colon may significantly reduce postoperative bleeding risk, according to investigators.

In a prospective study of almost 1,000 patients, this benefit was not influenced by polyp size, electrocautery setting, or concomitant use of antithrombotic medications, reported Heiko Pohl, MD, of Geisel School of Medicine at Dartmouth, Hanover, N.H., and colleagues.

“Endoscopic resection has replaced surgical resection as the primary treatment for large colon polyps due to a lower morbidity and less need for hospitalization,” the investigators wrote in Gastroenterology. “Postprocedure bleeding is the most common severe complication, occurring in 2%-24% of patients.” This risk is particularly common among patients with large polyps in the proximal colon.

Although previous trials have suggested that closing polyp resection sites with hemoclips could reduce the risk of postoperative bleeding, studies to date have been retrospective or uncontrolled, precluding definitive conclusions.

The prospective, controlled trial involved 44 endoscopists at 18 treatment centers. Enrollment included 919 patients with large, nonpedunculated colorectal polyps of at least 20 mm in diameter. Patients were randomized in an approximate 1:1 ratio into the clip group or control group and followed for at least 30 days after endoscopic polyp resection. The primary outcome was postoperative bleeding, defined as severe bleeding that required invasive intervention such as surgery or blood transfusion during follow-up. Subgroup analysis looked for associations between bleeding and polyp location, size, electrocautery setting, and medications.

Across the entire population, postoperative bleeding was significantly less common among patients who had their resection sites closed with clips, occurring at a rate of 3.5%, compared with 7.1% in the control group (P = .015). Serious adverse events were also less common in the clip group than the control group (4.8% vs. 9.5%; P = .006).

While the reduction of bleeding risk from clip closure was not influenced by polyp size, use of antithrombotic medications, or electrocautery setting, polyp location turned out to be a critical factor. Greatest reduction in risk of postoperative bleeding was seen among the 615 patients who had proximal polyps, based on a bleeding rate of 3.3% when clipped versus 9.6% among those who went without clips (P = .001). In contrast, clips in the distal colon were associated with a higher absolute risk of postoperative bleeding than no clips (4.0% vs. 1.4%); however, this difference was not statistically significant (P = .178).

“[T]his multicenter trial provides strong evidence that endoscopic clip closure of the mucosal defect after resection of large ... nonpedunculated colon polyps in the proximal colon significantly reduces the risk of postprocedure bleeding,” the investigators wrote.

They suggested that their study provides greater confidence in findings than similar trials previously conducted, enough to recommend that endoscopic techniques be altered accordingly. “[O]ur trial was methodologically rigorous, adequately powered, and all polyps were removed by endoscopic mucosal resection, which is considered the standard technique for large colon polyps in Western countries,” they wrote. “The results of the study are therefore broadly applicable to current practice. Furthermore, conduct of the study at different centers with multiple endoscopists strengthens generalizability of the findings.”

The investigators also speculated about why postoperative bleeding risk was increased when clips were used in the distal colon. “Potential explanations include a poorer quality of clipping, a shorter clip retention time, possible related to a thicker colon wall in the distal compared to the proximal colon,” they wrote, adding that “these considerations are worthy of further study.”

Indeed, more work remains to be done. “A formal cost-effectiveness analysis is needed to better understand the value of clip closure,” they wrote. “Such analysis can then also examine possible thresholds, for instance regarding the minimum proportion of polyp resections, for which complete closure should be achieved, or the maximum number of clips to close a defect.”

The study was funded by Boston Scientific. The investigators reported additional relationships with U.S. Endoscopy, Olympus, Medtronic, and others.

SOURCE: Pohl H et al. Gastroenterology. 2019 Mar 15. doi: 10.1053/j.gastro.2019.03.019.

Publications
Topics
Sections

Closing mucosal defects with hemoclips after endoscopic resection of large polyps in the proximal colon may significantly reduce postoperative bleeding risk, according to investigators.

In a prospective study of almost 1,000 patients, this benefit was not influenced by polyp size, electrocautery setting, or concomitant use of antithrombotic medications, reported Heiko Pohl, MD, of Geisel School of Medicine at Dartmouth, Hanover, N.H., and colleagues.

“Endoscopic resection has replaced surgical resection as the primary treatment for large colon polyps due to a lower morbidity and less need for hospitalization,” the investigators wrote in Gastroenterology. “Postprocedure bleeding is the most common severe complication, occurring in 2%-24% of patients.” This risk is particularly common among patients with large polyps in the proximal colon.

Although previous trials have suggested that closing polyp resection sites with hemoclips could reduce the risk of postoperative bleeding, studies to date have been retrospective or uncontrolled, precluding definitive conclusions.

The prospective, controlled trial involved 44 endoscopists at 18 treatment centers. Enrollment included 919 patients with large, nonpedunculated colorectal polyps of at least 20 mm in diameter. Patients were randomized in an approximate 1:1 ratio into the clip group or control group and followed for at least 30 days after endoscopic polyp resection. The primary outcome was postoperative bleeding, defined as severe bleeding that required invasive intervention such as surgery or blood transfusion during follow-up. Subgroup analysis looked for associations between bleeding and polyp location, size, electrocautery setting, and medications.

Across the entire population, postoperative bleeding was significantly less common among patients who had their resection sites closed with clips, occurring at a rate of 3.5%, compared with 7.1% in the control group (P = .015). Serious adverse events were also less common in the clip group than the control group (4.8% vs. 9.5%; P = .006).

While the reduction of bleeding risk from clip closure was not influenced by polyp size, use of antithrombotic medications, or electrocautery setting, polyp location turned out to be a critical factor. Greatest reduction in risk of postoperative bleeding was seen among the 615 patients who had proximal polyps, based on a bleeding rate of 3.3% when clipped versus 9.6% among those who went without clips (P = .001). In contrast, clips in the distal colon were associated with a higher absolute risk of postoperative bleeding than no clips (4.0% vs. 1.4%); however, this difference was not statistically significant (P = .178).

“[T]his multicenter trial provides strong evidence that endoscopic clip closure of the mucosal defect after resection of large ... nonpedunculated colon polyps in the proximal colon significantly reduces the risk of postprocedure bleeding,” the investigators wrote.

They suggested that their study provides greater confidence in findings than similar trials previously conducted, enough to recommend that endoscopic techniques be altered accordingly. “[O]ur trial was methodologically rigorous, adequately powered, and all polyps were removed by endoscopic mucosal resection, which is considered the standard technique for large colon polyps in Western countries,” they wrote. “The results of the study are therefore broadly applicable to current practice. Furthermore, conduct of the study at different centers with multiple endoscopists strengthens generalizability of the findings.”

The investigators also speculated about why postoperative bleeding risk was increased when clips were used in the distal colon. “Potential explanations include a poorer quality of clipping, a shorter clip retention time, possible related to a thicker colon wall in the distal compared to the proximal colon,” they wrote, adding that “these considerations are worthy of further study.”

Indeed, more work remains to be done. “A formal cost-effectiveness analysis is needed to better understand the value of clip closure,” they wrote. “Such analysis can then also examine possible thresholds, for instance regarding the minimum proportion of polyp resections, for which complete closure should be achieved, or the maximum number of clips to close a defect.”

The study was funded by Boston Scientific. The investigators reported additional relationships with U.S. Endoscopy, Olympus, Medtronic, and others.

SOURCE: Pohl H et al. Gastroenterology. 2019 Mar 15. doi: 10.1053/j.gastro.2019.03.019.

Closing mucosal defects with hemoclips after endoscopic resection of large polyps in the proximal colon may significantly reduce postoperative bleeding risk, according to investigators.

In a prospective study of almost 1,000 patients, this benefit was not influenced by polyp size, electrocautery setting, or concomitant use of antithrombotic medications, reported Heiko Pohl, MD, of Geisel School of Medicine at Dartmouth, Hanover, N.H., and colleagues.

“Endoscopic resection has replaced surgical resection as the primary treatment for large colon polyps due to a lower morbidity and less need for hospitalization,” the investigators wrote in Gastroenterology. “Postprocedure bleeding is the most common severe complication, occurring in 2%-24% of patients.” This risk is particularly common among patients with large polyps in the proximal colon.

Although previous trials have suggested that closing polyp resection sites with hemoclips could reduce the risk of postoperative bleeding, studies to date have been retrospective or uncontrolled, precluding definitive conclusions.

The prospective, controlled trial involved 44 endoscopists at 18 treatment centers. Enrollment included 919 patients with large, nonpedunculated colorectal polyps of at least 20 mm in diameter. Patients were randomized in an approximate 1:1 ratio into the clip group or control group and followed for at least 30 days after endoscopic polyp resection. The primary outcome was postoperative bleeding, defined as severe bleeding that required invasive intervention such as surgery or blood transfusion during follow-up. Subgroup analysis looked for associations between bleeding and polyp location, size, electrocautery setting, and medications.

Across the entire population, postoperative bleeding was significantly less common among patients who had their resection sites closed with clips, occurring at a rate of 3.5%, compared with 7.1% in the control group (P = .015). Serious adverse events were also less common in the clip group than the control group (4.8% vs. 9.5%; P = .006).

While the reduction of bleeding risk from clip closure was not influenced by polyp size, use of antithrombotic medications, or electrocautery setting, polyp location turned out to be a critical factor. Greatest reduction in risk of postoperative bleeding was seen among the 615 patients who had proximal polyps, based on a bleeding rate of 3.3% when clipped versus 9.6% among those who went without clips (P = .001). In contrast, clips in the distal colon were associated with a higher absolute risk of postoperative bleeding than no clips (4.0% vs. 1.4%); however, this difference was not statistically significant (P = .178).

“[T]his multicenter trial provides strong evidence that endoscopic clip closure of the mucosal defect after resection of large ... nonpedunculated colon polyps in the proximal colon significantly reduces the risk of postprocedure bleeding,” the investigators wrote.

They suggested that their study provides greater confidence in findings than similar trials previously conducted, enough to recommend that endoscopic techniques be altered accordingly. “[O]ur trial was methodologically rigorous, adequately powered, and all polyps were removed by endoscopic mucosal resection, which is considered the standard technique for large colon polyps in Western countries,” they wrote. “The results of the study are therefore broadly applicable to current practice. Furthermore, conduct of the study at different centers with multiple endoscopists strengthens generalizability of the findings.”

The investigators also speculated about why postoperative bleeding risk was increased when clips were used in the distal colon. “Potential explanations include a poorer quality of clipping, a shorter clip retention time, possible related to a thicker colon wall in the distal compared to the proximal colon,” they wrote, adding that “these considerations are worthy of further study.”

Indeed, more work remains to be done. “A formal cost-effectiveness analysis is needed to better understand the value of clip closure,” they wrote. “Such analysis can then also examine possible thresholds, for instance regarding the minimum proportion of polyp resections, for which complete closure should be achieved, or the maximum number of clips to close a defect.”

The study was funded by Boston Scientific. The investigators reported additional relationships with U.S. Endoscopy, Olympus, Medtronic, and others.

SOURCE: Pohl H et al. Gastroenterology. 2019 Mar 15. doi: 10.1053/j.gastro.2019.03.019.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM GASTROENTEROLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Colorectal Cancer Awareness Fair – Make Your Bottom Your Top Priority

Article Type
Changed
Thu, 09/05/2019 - 15:12

Background: The Comprehensive Cancer Program held a community Colorectal Cancer Awareness Fair on March 5, 2019 at the VAMC. The goal was to increase awareness of Colorectal Cancer and to engage veterans in educational opportunities about Colorectal Cancer.

Methods: The VAMC purchased an in atable “Megacolon” for veterans to walk through guided by nurses from the GI department. Cubicles were set-up for nursing education sessions, a provider station, a scheduling station, and a colonoscope table. A video loop “Before and After Colonoscopy” by Mechanisms in Medicine, Inc. (Thornhill, Ontario, Canada) played continuously in the waiting area by the provider and nurse’s cubicles. Providers in the GI department offered 2 educational presentations: “How to Stop Colon Cancer Before It Starts” by Carol Macaron, MD; and “Colonoscopy: The Good, Bad, and Ugly” by Edith Ho, MD. Additional education information was provided at staffed tables from VA General Surgery, GI, MOVE! Nutrition & Food Services, and Smoking Cessation. Also, in attendance were Crohn’s and Colitis Foundation, and the American Cancer Society. External Affairs advertised the fair on Facebook and Twitter. Medical Media created publicity posters and event flyers.

Results: The event was attended by 244 people—68 veterans, 170 employees, and 6 guests. Six colonoscopies were scheduled onsite. At least 7 veterans had questions regarding their colonoscopy surveillance in which reminder dates were given.

Author and Disclosure Information

Correspondence: Jennifer Dimick ([email protected])

Publications
Topics
Sections
Author and Disclosure Information

Correspondence: Jennifer Dimick ([email protected])

Author and Disclosure Information

Correspondence: Jennifer Dimick ([email protected])

Background: The Comprehensive Cancer Program held a community Colorectal Cancer Awareness Fair on March 5, 2019 at the VAMC. The goal was to increase awareness of Colorectal Cancer and to engage veterans in educational opportunities about Colorectal Cancer.

Methods: The VAMC purchased an in atable “Megacolon” for veterans to walk through guided by nurses from the GI department. Cubicles were set-up for nursing education sessions, a provider station, a scheduling station, and a colonoscope table. A video loop “Before and After Colonoscopy” by Mechanisms in Medicine, Inc. (Thornhill, Ontario, Canada) played continuously in the waiting area by the provider and nurse’s cubicles. Providers in the GI department offered 2 educational presentations: “How to Stop Colon Cancer Before It Starts” by Carol Macaron, MD; and “Colonoscopy: The Good, Bad, and Ugly” by Edith Ho, MD. Additional education information was provided at staffed tables from VA General Surgery, GI, MOVE! Nutrition & Food Services, and Smoking Cessation. Also, in attendance were Crohn’s and Colitis Foundation, and the American Cancer Society. External Affairs advertised the fair on Facebook and Twitter. Medical Media created publicity posters and event flyers.

Results: The event was attended by 244 people—68 veterans, 170 employees, and 6 guests. Six colonoscopies were scheduled onsite. At least 7 veterans had questions regarding their colonoscopy surveillance in which reminder dates were given.

Background: The Comprehensive Cancer Program held a community Colorectal Cancer Awareness Fair on March 5, 2019 at the VAMC. The goal was to increase awareness of Colorectal Cancer and to engage veterans in educational opportunities about Colorectal Cancer.

Methods: The VAMC purchased an in atable “Megacolon” for veterans to walk through guided by nurses from the GI department. Cubicles were set-up for nursing education sessions, a provider station, a scheduling station, and a colonoscope table. A video loop “Before and After Colonoscopy” by Mechanisms in Medicine, Inc. (Thornhill, Ontario, Canada) played continuously in the waiting area by the provider and nurse’s cubicles. Providers in the GI department offered 2 educational presentations: “How to Stop Colon Cancer Before It Starts” by Carol Macaron, MD; and “Colonoscopy: The Good, Bad, and Ugly” by Edith Ho, MD. Additional education information was provided at staffed tables from VA General Surgery, GI, MOVE! Nutrition & Food Services, and Smoking Cessation. Also, in attendance were Crohn’s and Colitis Foundation, and the American Cancer Society. External Affairs advertised the fair on Facebook and Twitter. Medical Media created publicity posters and event flyers.

Results: The event was attended by 244 people—68 veterans, 170 employees, and 6 guests. Six colonoscopies were scheduled onsite. At least 7 veterans had questions regarding their colonoscopy surveillance in which reminder dates were given.

Publications
Publications
Topics
Article Type
Sections
Citation Override
Abstract Presented at the 2019 Association of VA Hematology/Oncology Annual Meeting
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 09/04/2019 - 16:15
Un-Gate On Date
Wed, 09/04/2019 - 16:15
Use ProPublica
CFC Schedule Remove Status
Wed, 09/04/2019 - 16:15
Hide sidebar & use full width
render the right sidebar.

Surviving Colorectal Cancer, Now at Risk for Hypertension

Article Type
Changed
Tue, 05/03/2022 - 15:13
Are patients more at risk for hypertension and diabetes mellitus after surviving colorectal cancer? VA researchers investigate.

Colorectal cancer (CRC) survivor rates are improving, which means people are living long enough after the cancer to have other chronic conditions. CRC is the third most commonly diagnosed cancer among users of the US Department of Veterans Affairs (VA) health care system, according to VA researchers, and there is a high prevalence of cardiovascular disease (CVD). The researchers also say emerging evidence suggests that survivors of CRC may be more likely to develop diabetes mellitus (DM) in the 5 years following their cancer diagnosis. But they add that there is a paucity of research about control of CVD-related chronic conditions among survivors of CRC.

In a retrospective study, the researchers compared 9,758 nonmetastatic patients with CRC with 29,066 people who had not had cancer. At baseline, 69% of the survivors of CRC and the matched controls were diagnosed with hypertension, 52% with hyperlipidemia, and 37% with DM.

But somewhat contrary to expectations, the researchers found no significant differences between the 2 groups for DM in the year following the baseline assessment. The researchers point to the VA’s “strong history” of DM risk reduction research and 2 national programs targeting DM, although they do not know whether the people in their study participated in those.

The survivors of CRC also had half the odds of being diagnosed with hyperlipidemia. However, they did have 57% higher odds of being diagnosed with hypertension.

Although the researchers acknowledge that hypertension is a transient adverse effect of certain chemotherapy regimens, they found only 7 survivors of CRC and 11 controls were treated with bevacizumab during their first year postanchor date.

The relationship between nonmetastatic CRC and CVD risk-related chronic conditions is complex, the researchers say. But they share risk factors, including obesity, physical inactivity, and diet.

The researchers call behavioral change interventions that improve survivors of CRC physical activity, dietary habits, and body mass index a “promising beginning” but call for other similar interventions, particularly those targeting blood pressure management and adherence to antihypertensive medications (which was significantly lower among the survivors).

While the magnitude of the effect regarding hypertension seems relatively small, the researchers say, they believe it is still an important difference when considered from a population health perspective—and one that should be addressed. The researchers also note that nonmetastatic survivors of CRC and controls had very similar rates of primary care visits in the 3 years postanchor date and as a result similar opportunities to receive a hypertension diagnosis.

Publications
Topics
Sections
Are patients more at risk for hypertension and diabetes mellitus after surviving colorectal cancer? VA researchers investigate.
Are patients more at risk for hypertension and diabetes mellitus after surviving colorectal cancer? VA researchers investigate.

Colorectal cancer (CRC) survivor rates are improving, which means people are living long enough after the cancer to have other chronic conditions. CRC is the third most commonly diagnosed cancer among users of the US Department of Veterans Affairs (VA) health care system, according to VA researchers, and there is a high prevalence of cardiovascular disease (CVD). The researchers also say emerging evidence suggests that survivors of CRC may be more likely to develop diabetes mellitus (DM) in the 5 years following their cancer diagnosis. But they add that there is a paucity of research about control of CVD-related chronic conditions among survivors of CRC.

In a retrospective study, the researchers compared 9,758 nonmetastatic patients with CRC with 29,066 people who had not had cancer. At baseline, 69% of the survivors of CRC and the matched controls were diagnosed with hypertension, 52% with hyperlipidemia, and 37% with DM.

But somewhat contrary to expectations, the researchers found no significant differences between the 2 groups for DM in the year following the baseline assessment. The researchers point to the VA’s “strong history” of DM risk reduction research and 2 national programs targeting DM, although they do not know whether the people in their study participated in those.

The survivors of CRC also had half the odds of being diagnosed with hyperlipidemia. However, they did have 57% higher odds of being diagnosed with hypertension.

Although the researchers acknowledge that hypertension is a transient adverse effect of certain chemotherapy regimens, they found only 7 survivors of CRC and 11 controls were treated with bevacizumab during their first year postanchor date.

The relationship between nonmetastatic CRC and CVD risk-related chronic conditions is complex, the researchers say. But they share risk factors, including obesity, physical inactivity, and diet.

The researchers call behavioral change interventions that improve survivors of CRC physical activity, dietary habits, and body mass index a “promising beginning” but call for other similar interventions, particularly those targeting blood pressure management and adherence to antihypertensive medications (which was significantly lower among the survivors).

While the magnitude of the effect regarding hypertension seems relatively small, the researchers say, they believe it is still an important difference when considered from a population health perspective—and one that should be addressed. The researchers also note that nonmetastatic survivors of CRC and controls had very similar rates of primary care visits in the 3 years postanchor date and as a result similar opportunities to receive a hypertension diagnosis.

Colorectal cancer (CRC) survivor rates are improving, which means people are living long enough after the cancer to have other chronic conditions. CRC is the third most commonly diagnosed cancer among users of the US Department of Veterans Affairs (VA) health care system, according to VA researchers, and there is a high prevalence of cardiovascular disease (CVD). The researchers also say emerging evidence suggests that survivors of CRC may be more likely to develop diabetes mellitus (DM) in the 5 years following their cancer diagnosis. But they add that there is a paucity of research about control of CVD-related chronic conditions among survivors of CRC.

In a retrospective study, the researchers compared 9,758 nonmetastatic patients with CRC with 29,066 people who had not had cancer. At baseline, 69% of the survivors of CRC and the matched controls were diagnosed with hypertension, 52% with hyperlipidemia, and 37% with DM.

But somewhat contrary to expectations, the researchers found no significant differences between the 2 groups for DM in the year following the baseline assessment. The researchers point to the VA’s “strong history” of DM risk reduction research and 2 national programs targeting DM, although they do not know whether the people in their study participated in those.

The survivors of CRC also had half the odds of being diagnosed with hyperlipidemia. However, they did have 57% higher odds of being diagnosed with hypertension.

Although the researchers acknowledge that hypertension is a transient adverse effect of certain chemotherapy regimens, they found only 7 survivors of CRC and 11 controls were treated with bevacizumab during their first year postanchor date.

The relationship between nonmetastatic CRC and CVD risk-related chronic conditions is complex, the researchers say. But they share risk factors, including obesity, physical inactivity, and diet.

The researchers call behavioral change interventions that improve survivors of CRC physical activity, dietary habits, and body mass index a “promising beginning” but call for other similar interventions, particularly those targeting blood pressure management and adherence to antihypertensive medications (which was significantly lower among the survivors).

While the magnitude of the effect regarding hypertension seems relatively small, the researchers say, they believe it is still an important difference when considered from a population health perspective—and one that should be addressed. The researchers also note that nonmetastatic survivors of CRC and controls had very similar rates of primary care visits in the 3 years postanchor date and as a result similar opportunities to receive a hypertension diagnosis.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Gate On Date
Wed, 08/28/2019 - 11:15
Un-Gate On Date
Wed, 08/28/2019 - 11:15
Use ProPublica
CFC Schedule Remove Status
Wed, 08/28/2019 - 11:15
Hide sidebar & use full width
render the right sidebar.

Coordination of Care Between Primary Care and Oncology for Patients With Prostate Cancer (FULL)

Article Type
Changed
Thu, 12/15/2022 - 14:41
Display Headline
Coordination of Care Between Primary Care and Oncology for Patients With Prostate Cancer

The following is a lightly edited transcript of a teleconference recorded in July 2018. The teleconference brought together health care providers from the Greater Los Angeles VA Health Care System (GLAVAHCS) to discuss the real-world processes for managing the treatment of patients with prostate cancer as they move between primary and specialist care.

William J. Aronson, MD. We are fortunate in having a superb medical record system at the Department of Veterans Affairs (VA) where we can all communicate with each other through a number of methods. Let’s start our discussion by reviewing an index patient that we see in our practice who has been treated with either radical prostatectomy or radiation therapy. One question to address is: Is there a point when the Urology or Radiation Oncology service can transition the patient’s entire care back to the primary care team? And if so, what would be the optimal way to accomplish this?

Nick, is there some point at which you discharge the patient from the radiation oncology service and give specific directions to primary care, or is it primarily just back to urology in your case?

Nicholas G. Nickols, MD, PhD. I have not discharged any patient from my clinic after definitive prostate cancer treatment. During treatment, patients are seen every week. Subsequently, I see them 6 weeks posttreatment, and then every 4 months for the first year, then every 6 months for the next 4 years, and then yearly after that. Although I never formally discharged a patient from my clinic, you can see based on the frequency of visits, that the patient will see more often than their primary care provider (PCP) toward the beginning. And then, after some years, the patient sees their primary more than they me. So it’s not an immediate hand off but rather a gradual transition. It’s important that the PCP is aware of what to look for especially for the late recurrences, late potential side effects, probably more significantly than the early side effects, how to manage them when appropriate, and when to ask the patient to see our team more frequently in follow-up.

William Aronson. We have a number of patients who travel tremendous distances to see us, and I tend to think that many of our follow-up patients, once things are stabilized with regards to management of their side effects, really could see their primary care doctors if we can give them specific instructions on, for example, when to get a prostate-specific antigen (PSA) test and when to refer back to us.

Alison, can you think of some specific cases where you feel like we’ve successfully done that?

Alison Neymark, MS. For the most part we haven’t discharged people, either. What we have done is transitioned them over to a phone clinic. In our department, we have 4 nurse practitioners (NPs) who each have a half-day of phone clinic where they call patients with their test results. Some of those patients are prostate cancer patients that we have been following for years. We schedule them for a phone call, whether it’s every 3 months, every 6 months or every year, to review the updated PSA level and to just check in with them by phone. It’s a win-win because it’s a really quick phone call to reassure the veteran that the PSA level is being followed, and it frees up an in-person appointment slot for another veteran.

 

 

We still have patients that prefer face-to-face visits, even though they know we’re not doing anything except discussing a PSA level with them—they just want that security of seeing our face. Some patients are very nervous, and they don’t necessarily want to be discharged, so to speak, back to primary care. Also, for those patients that travel a long distance to clinic, we offer an appointment in the video chat clinic, with the community-based outpatient clinics in Bakersfield and Santa Maria, California.

PSA Levels

William Aronson. I probably see a patient about every 4 to 6 weeks who has a low PSA after about 10 years and has a long distance to travel and mobility and other problems that make it difficult to come in. 

And so, in a number of those cases, I refer the patients back to their PCP and recommend that they get a PSA test every 6 months to a year. Then they refer back to us if there’s any further issues or if there is a significant rise in the PSA level.

The challenge that I have is, what is that specific guideline to give with regards to the rise in PSA? I think it all depends on the patients prostate cancer clinical features and comorbidities.

Nicholas Nickols. If a patient has been seen by me in follow-up a number of times and there’s really no active issues and there’s a low suspicion of recurrence, then I offer the patient the option of a phone follow-up as an alternative to face to face. Some of them accept that, but I ask that they agree to also see either urology or their PCP face to face. I will also remotely ensure that they’re getting the right laboratory tests, and if not, I’ll put those orders in.

With regard to when to refer a patient back for a suspected recurrence after definitive radiation therapy, there is an accepted definition of biochemical failure called the Phoenix definition, which is an absolute rise in 2 ng/mL of PSA over their posttreatment nadir. Often the posttreatment nadir, especially if they were on hormone therapy, will be close to 0. If the PSA gets to 2, that is a good trigger for a referral back to me and/or urology to discuss restaging and workup for a suspected recurrence.

For patients that are postsurgery and then subsequently get salvage radiation, it is not as clear when a restaging workup should be initiated. Currently, the imaging that is routine care is not very sensitive for detecting PSA in that setting until the PSA is around 0.8 ng/mL, and that’s with the most modern imaging available. Over time that may improve.

William Aronson. The other index patient to think about would be the patient who is on watchful waiting for their prostate cancer, which is to be distinguished from active surveillance. If someone’s on active surveillance, we’re regularly doing prostate biopsies and doing very close monitoring; but we also have patients who have multiple other medical problems, have a limited life expectancy, don’t have aggressive prostate cancer, and it’s extremely reasonable not to do a biopsy in those patients.

 

 

Again, those are patients where we do follow the PSA generally every 6 months. And I think there’s also scenarios there where it’s reasonable to refer back to primary care with specific instructions. These, again, are patients who had difficulty getting in to see us or have mobility issues, but it is also a way to limit patient visits if that’s their desire.

Peter Glassman, MBBS, MSc: I’m trained as both a general internist and board certified in hospice and palliative medicine. I currently provide primary care as well as palliative care. I view prostate cancer from the diagnosis through the treatment spectrum as a continuum. It starts with the PCP with an elevated PSA level or if the digital rectal exam has an abnormality, and then the role of the genitourinary (GU) practitioner becomes more significant during the active treatment and diagnostic phases.

Primary care doesn’t disappear, and I think there are 2 major issues that go along with that. First of all, we in primary care, because we take care of patients that often have other comorbidities, need to work with the patient on those comorbidities. Secondly, we need the information shared between the GU and primary care providers so that we can answer questions from our patients and have an understanding of what they’re going through and when.

As time goes on, we go through various phases: We may reach a cure, a quiescent period, active therapy, watchful waiting, or recurrence. Primary care gets involved as time goes on when the disease either becomes quiescent, is just being followed, or is considered cured. Clearly when you have watchful waiting, active treatment, or are in a recurrence, then GU takes the forefront.

I view it as a wave function. Primary care to GU with primary in smaller letters and then primary, if you will, in larger letters, GU becomes a lesser participant unless there is active therapy, watchful waiting or recurrence.

In doing a little bit of research, I found 2 very good and very helpful documents. One is the American Cancer Society (ACS) prostate cancer survivorship care guidelines (Box). And the other is a synopsis of the guidelines. What I liked was that the guidelines focused not only on what should be done for the initial period of prostate cancer, but also for many of the ancillary issues which we often don’t give voice to. The guidelines provide a structure, a foundation to work with our patients over time on their prostate cancer-related issues while, at the same time, being cognizant that we need to deal with their other comorbid conditions.

Modes of Communication

Alison Neymark. We find that including parameters for PSA monitoring in our Progress Notes in the electronic health record (EHR) the best way to communicate with other providers. We’ll say, “If PSA gets to this level, please refer back.” We try to make it clear because with the VA being a training facility, it could be a different resident/attending physician team that’s going to see the patient the next time he is in primary care.

 

 

Peter Glassman. Yes, we’re very lucky, as Bill talked about earlier and Alison just mentioned. We have the EHR, and Bill may remember this. Before the EHR, we were constantly fishing to find the most relevant notes. If a patient saw a GU practitioner the day before they saw me, I was often asking the patient what was said. Now we can just review the notes.

It’s a double-edged sword though because there are, of course, many notes in a medical record; and you have to look for the specific items. The EHR and documenting the medical record probably plays the primary role in getting information across. When you want to have an active handoff, or you need to communicate with each other, we have a variety of mechanisms, ranging from the phone to the Microsoft Skype Link (Redmond, WA) system that allows us to tap a message to a colleague.

And I’ve been here long enough that I’ve seen most permutations of how prostate cancer is diagnosed as well as shared among providers. Bill and I have shared patients. Alison and I have shared patients, not necessarily with prostate cancer, although that too. But we know how to communicate with each other. And of course, there’s paging if you need something more urgently.

William Aronson. We also use Microsoft Outlook e-mail, and encrypt the messages to keep them confidential and private. The other nice thing we have is there is a nationwide urology Outlook e-mail, so if any of us have any specific questions, through one e-mail we can send it around the country; and there’s usually multiple very useful responses. That’s another real strength of our system within the VA that helps patient care enormously.

Nicholas Nickols. Sometimes, if there’s a critical note that I absolutely want someone on the care team to read, I’ll add them as a cosigner; and that will pop up when they log in to the Computerized Patient Record System (CPRS) as something that they need to read.

If the patient lives particularly far or gets his care at another VA medical center and laboratory tests are needed, then I will reach out to their PCP via e-mail. If contact is not confirmed, I will reach out via phone or Skype.

Peter Glassman. The most helpful notes are those that are very specific as to what primary care is being asked to do and/or what urology is going to be doing. So, the more specific we get in the notes as to what is being addressed, I think that’s very helpful.

I have been here long enough that I’ve known both Alison and Bill; and if they have an issue, they will tap me a message. It wasn’t long ago that Bill sent a message to me, and we worked on a patient with prostate cancer who was going to be on long-term hormone therapy. We talked about osteoporosis management, and between us we worked out who was going to do what. Those are the kind of shared decision-making situations that are very, very helpful.
 

 

 

Alison Neymark. Also, GLAVAHCS has a home-based primary care team (HBPC), and a lot of the PCPs for that team are NPs. They know that they can contact me for their patients because a lot of those patients are on watchful waiting, and we do not necessarily need to see them face to face in clinic. Our urology team just needs to review updated lab results and how they are doing clinically. The HBPC NP who knows them best can contact me every 6 months or so, and we’ll discuss the case, which avoids making the patient come in, especially when they’re homebound. Those of us that have been working at the VA for many years have established good relationships. We feel very comfortable reaching out and talking to each other about these patients

Peter Glassman. Alison, I agree. When I can talk to my patients and say, “You know, we had that question about,” whatever the question might be, “and I contacted urology, and this is what they said.” It gives the patient confidence that we’re following up on the issues that they have and that we’re communicating with each other in a way that is to their benefit. And I think it’s very appreciated both by the provider as well as the patient.

William Aronson. Not infrequently I’ll have patients who have nonurologic issues, which I may first detect, or who have specific issues with their prostate cancer that can be comanaged. And I have found that when I send an encrypted e-mail to the PCP, it has been an extremely satisfying interaction; and we really get to the heart of the matter quickly for the sake of the veteran.

Veterans With Comorbidities

William Aronson. Posttraumatic stress disorder (PTSD) is a very significant and unique aspect of our patients, which is enormously important to recognize. For example, the side effects of prostate treatments can be very significant, whether radiation or surgery. Our patients understandably can be very fearful of the prostate cancer diagnosis and treatment side effects.

We know, for example, after a patient gets a diagnosis of prostate cancer, they’re at increased risk of cardiac death. That’s an especially important issue for our patients that there be an ongoing interaction between urology and primary care.

The ACS guidelines that Dr. Glassman referred to were enlightening. In many cases, primary care can look at the whole patient and their circumstances better than we can and may detect, for example, specific psychological issues that either they can manage or refer to other specialists.

Peter Glassman. One of the things that was highlighted in the ACS guideline is that in any population of men who have this disease, there’s going to be distress, anxiety, and full-fledged depression. Of course, there are psychosocial aspects of prostate cancer, such as sexual activity and intimacy with a partner that we often don’t explore but are probably playing an important role in the overall health of our patients. We need to be mindful of these psychosocial aspects and at least periodically ask them, “How are you doing with this? How are things at home?” And of course, we already use screeners for depression. As the article noted, distress and anxiety and other factors can make somebody’s life less optimal with poorer quality of life.

 

 

Dual Care Patients

Alison Neymark. Many patients whether they have Medicare, insurance through their spouse, or Kaiser Permanente through their job, choose to go to both places. The challenge is communicating with the non-VA providers because here at the VA we can communicate easily through Skype, Outlook e-mail, or CPRS, but for dual care patients who’s in charge? I encourage the veterans to choose whom they want to manage their care; we’re always here and happy to treat them, but they need to decide who’s in charge because I don’t want them to get into a situation where the differing opinions lead to a delay in care.

Nicholas Nickols. The communication when the patient is receiving care outside VA, either on a continuous basis or temporarily, is more of a challenge. We obviously can’t rely upon the messaging system, face-to-face contact is difficult, and they may not be able to use e-mail as well. So in those situations, usually a phone call is the best approach. I have found that the outside providers are happy to speak on the phone to coordinate care.

Peter Glassman. I agree, it does add a layer of complexity because we don’t readily have the notes, any information in front of us. That said, a lot of our patients can and do bring in information from outside specialists, and I’m hopeful that they share the information that we provide back to their outside doctors as well.

William Aronson. Some patient get nervous. They might decide they want care elsewhere, but they still want the VA available for them. I always let them know they should proceed in whatever way they prefer, but we’re always available and here for them. I try to empower them to make their own decisions and feel comfortable with them.

Nicholas Nickols. Notes from the outside, if they’re being referred for VA Choice or community care, do get uploaded into VistA Imaging and can be accessed, although it’s not instantaneous. Sometimes there’s a delay, but I have been able to access outside notes most of the time. If a patient goes through a clinic at the VA, the note is written in real time, and you can read it immediately.

Peter Glassman. That is true for patients that are within the VA system who receive contracted care either through Choice or through non-VA care that is contracted through VA. For somebody who is choosing to use 2 health care systems, that can provide more of a challenge because those notes don’t come to us. Over time, most of my patients have brought test results to me.

The thing with oncologic care, of course, is it’s a lot more complex. And it’s hard to know without reasonable documentation what’s been going on. At some level, you have to trust that the outside provider is doing whatever they need to do, or you have to take it upon yourself to do it within the system.

 

 

Alison Neymark. In my experience with the Choice Program, it really depends on the outside providers and how comfortable they are with the system that has been established to share records. Not all providers are going into that system and accessing it. I have had cases where I will see the non-VA provider’s note and it’ll say, “No documentation available for this consultation.” It just happens that they didn’t go into the system to review it. So it can be a challenge.

I’ve had good communication with the providers who use the system correctly. In some cases, just to make it easier, I will go ahead and communicate with them through encrypted e-mail, or I’ll talk to their care coordinators directly by phone. 

That way we can make sure everything is expedited to avoid any delay in the patient’s care. It is more time consuming, but it’s important because some of the procedures we don’t offer at the VA, and that’s why we’re using the Choice system.

Peter Glassman. Many, if not most, PCPs are going to take care of these patients, certainly within the VA, with their GU colleagues. And most of us feel comfortable using the current documentation system in a way that allows us to share information or at least to gather information about these patients.

One of the things that I think came out for me in looking at this was that there are guidelines or there are ideas out there on how to take better care of these patients. And I for one learned a fair bit just by going through these documents, which I’m very appreciative of. But it does highlight to me that we can give good care and provide good shared care for prostate cancer survivors. I think that is something that perhaps this discussion will highlight that not only are people doing that, but there are resources they can utilize that will help them get a more comprehensive picture of taking care of prostate cancer survivors in the primary care clinic.

The beauty of the VA system as a system is that as these issues come up that might affect the overall health of the veteran with prostate cancer, for example, psychosocial issues, we have many people that can address this that are experts in their area. And one of the great beauties of having an all-encompassing healthcare system is being able to use resources within the system, whether that be for other medical problems or other social or other psychological issues, that we ourselves are not expert in. We can reach out to our other colleagues and ask them for assistance. We have that available to help the patients. It’s really holistic.

We even have integrated medicine where we can help patients, hopefully, get back into a healthy lifestyle, for example, whereas we may not have that expertise or knowledge. We often think of this as sort of a shared decision between GU and primary care. But, in fact, it’s really the responsibility of many, many people of the system at large. We are very lucky to have that.

Article PDF
Author and Disclosure Information

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Issue
Federal Practitioner - 35(5)s
Publications
Topics
Page Number
S18-S23
Sections
Author and Disclosure Information

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Author and Disclosure Information

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Article PDF
Article PDF

The following is a lightly edited transcript of a teleconference recorded in July 2018. The teleconference brought together health care providers from the Greater Los Angeles VA Health Care System (GLAVAHCS) to discuss the real-world processes for managing the treatment of patients with prostate cancer as they move between primary and specialist care.

William J. Aronson, MD. We are fortunate in having a superb medical record system at the Department of Veterans Affairs (VA) where we can all communicate with each other through a number of methods. Let’s start our discussion by reviewing an index patient that we see in our practice who has been treated with either radical prostatectomy or radiation therapy. One question to address is: Is there a point when the Urology or Radiation Oncology service can transition the patient’s entire care back to the primary care team? And if so, what would be the optimal way to accomplish this?

Nick, is there some point at which you discharge the patient from the radiation oncology service and give specific directions to primary care, or is it primarily just back to urology in your case?

Nicholas G. Nickols, MD, PhD. I have not discharged any patient from my clinic after definitive prostate cancer treatment. During treatment, patients are seen every week. Subsequently, I see them 6 weeks posttreatment, and then every 4 months for the first year, then every 6 months for the next 4 years, and then yearly after that. Although I never formally discharged a patient from my clinic, you can see based on the frequency of visits, that the patient will see more often than their primary care provider (PCP) toward the beginning. And then, after some years, the patient sees their primary more than they me. So it’s not an immediate hand off but rather a gradual transition. It’s important that the PCP is aware of what to look for especially for the late recurrences, late potential side effects, probably more significantly than the early side effects, how to manage them when appropriate, and when to ask the patient to see our team more frequently in follow-up.

William Aronson. We have a number of patients who travel tremendous distances to see us, and I tend to think that many of our follow-up patients, once things are stabilized with regards to management of their side effects, really could see their primary care doctors if we can give them specific instructions on, for example, when to get a prostate-specific antigen (PSA) test and when to refer back to us.

Alison, can you think of some specific cases where you feel like we’ve successfully done that?

Alison Neymark, MS. For the most part we haven’t discharged people, either. What we have done is transitioned them over to a phone clinic. In our department, we have 4 nurse practitioners (NPs) who each have a half-day of phone clinic where they call patients with their test results. Some of those patients are prostate cancer patients that we have been following for years. We schedule them for a phone call, whether it’s every 3 months, every 6 months or every year, to review the updated PSA level and to just check in with them by phone. It’s a win-win because it’s a really quick phone call to reassure the veteran that the PSA level is being followed, and it frees up an in-person appointment slot for another veteran.

 

 

We still have patients that prefer face-to-face visits, even though they know we’re not doing anything except discussing a PSA level with them—they just want that security of seeing our face. Some patients are very nervous, and they don’t necessarily want to be discharged, so to speak, back to primary care. Also, for those patients that travel a long distance to clinic, we offer an appointment in the video chat clinic, with the community-based outpatient clinics in Bakersfield and Santa Maria, California.

PSA Levels

William Aronson. I probably see a patient about every 4 to 6 weeks who has a low PSA after about 10 years and has a long distance to travel and mobility and other problems that make it difficult to come in. 

And so, in a number of those cases, I refer the patients back to their PCP and recommend that they get a PSA test every 6 months to a year. Then they refer back to us if there’s any further issues or if there is a significant rise in the PSA level.

The challenge that I have is, what is that specific guideline to give with regards to the rise in PSA? I think it all depends on the patients prostate cancer clinical features and comorbidities.

Nicholas Nickols. If a patient has been seen by me in follow-up a number of times and there’s really no active issues and there’s a low suspicion of recurrence, then I offer the patient the option of a phone follow-up as an alternative to face to face. Some of them accept that, but I ask that they agree to also see either urology or their PCP face to face. I will also remotely ensure that they’re getting the right laboratory tests, and if not, I’ll put those orders in.

With regard to when to refer a patient back for a suspected recurrence after definitive radiation therapy, there is an accepted definition of biochemical failure called the Phoenix definition, which is an absolute rise in 2 ng/mL of PSA over their posttreatment nadir. Often the posttreatment nadir, especially if they were on hormone therapy, will be close to 0. If the PSA gets to 2, that is a good trigger for a referral back to me and/or urology to discuss restaging and workup for a suspected recurrence.

For patients that are postsurgery and then subsequently get salvage radiation, it is not as clear when a restaging workup should be initiated. Currently, the imaging that is routine care is not very sensitive for detecting PSA in that setting until the PSA is around 0.8 ng/mL, and that’s with the most modern imaging available. Over time that may improve.

William Aronson. The other index patient to think about would be the patient who is on watchful waiting for their prostate cancer, which is to be distinguished from active surveillance. If someone’s on active surveillance, we’re regularly doing prostate biopsies and doing very close monitoring; but we also have patients who have multiple other medical problems, have a limited life expectancy, don’t have aggressive prostate cancer, and it’s extremely reasonable not to do a biopsy in those patients.

 

 

Again, those are patients where we do follow the PSA generally every 6 months. And I think there’s also scenarios there where it’s reasonable to refer back to primary care with specific instructions. These, again, are patients who had difficulty getting in to see us or have mobility issues, but it is also a way to limit patient visits if that’s their desire.

Peter Glassman, MBBS, MSc: I’m trained as both a general internist and board certified in hospice and palliative medicine. I currently provide primary care as well as palliative care. I view prostate cancer from the diagnosis through the treatment spectrum as a continuum. It starts with the PCP with an elevated PSA level or if the digital rectal exam has an abnormality, and then the role of the genitourinary (GU) practitioner becomes more significant during the active treatment and diagnostic phases.

Primary care doesn’t disappear, and I think there are 2 major issues that go along with that. First of all, we in primary care, because we take care of patients that often have other comorbidities, need to work with the patient on those comorbidities. Secondly, we need the information shared between the GU and primary care providers so that we can answer questions from our patients and have an understanding of what they’re going through and when.

As time goes on, we go through various phases: We may reach a cure, a quiescent period, active therapy, watchful waiting, or recurrence. Primary care gets involved as time goes on when the disease either becomes quiescent, is just being followed, or is considered cured. Clearly when you have watchful waiting, active treatment, or are in a recurrence, then GU takes the forefront.

I view it as a wave function. Primary care to GU with primary in smaller letters and then primary, if you will, in larger letters, GU becomes a lesser participant unless there is active therapy, watchful waiting or recurrence.

In doing a little bit of research, I found 2 very good and very helpful documents. One is the American Cancer Society (ACS) prostate cancer survivorship care guidelines (Box). And the other is a synopsis of the guidelines. What I liked was that the guidelines focused not only on what should be done for the initial period of prostate cancer, but also for many of the ancillary issues which we often don’t give voice to. The guidelines provide a structure, a foundation to work with our patients over time on their prostate cancer-related issues while, at the same time, being cognizant that we need to deal with their other comorbid conditions.

Modes of Communication

Alison Neymark. We find that including parameters for PSA monitoring in our Progress Notes in the electronic health record (EHR) the best way to communicate with other providers. We’ll say, “If PSA gets to this level, please refer back.” We try to make it clear because with the VA being a training facility, it could be a different resident/attending physician team that’s going to see the patient the next time he is in primary care.

 

 

Peter Glassman. Yes, we’re very lucky, as Bill talked about earlier and Alison just mentioned. We have the EHR, and Bill may remember this. Before the EHR, we were constantly fishing to find the most relevant notes. If a patient saw a GU practitioner the day before they saw me, I was often asking the patient what was said. Now we can just review the notes.

It’s a double-edged sword though because there are, of course, many notes in a medical record; and you have to look for the specific items. The EHR and documenting the medical record probably plays the primary role in getting information across. When you want to have an active handoff, or you need to communicate with each other, we have a variety of mechanisms, ranging from the phone to the Microsoft Skype Link (Redmond, WA) system that allows us to tap a message to a colleague.

And I’ve been here long enough that I’ve seen most permutations of how prostate cancer is diagnosed as well as shared among providers. Bill and I have shared patients. Alison and I have shared patients, not necessarily with prostate cancer, although that too. But we know how to communicate with each other. And of course, there’s paging if you need something more urgently.

William Aronson. We also use Microsoft Outlook e-mail, and encrypt the messages to keep them confidential and private. The other nice thing we have is there is a nationwide urology Outlook e-mail, so if any of us have any specific questions, through one e-mail we can send it around the country; and there’s usually multiple very useful responses. That’s another real strength of our system within the VA that helps patient care enormously.

Nicholas Nickols. Sometimes, if there’s a critical note that I absolutely want someone on the care team to read, I’ll add them as a cosigner; and that will pop up when they log in to the Computerized Patient Record System (CPRS) as something that they need to read.

If the patient lives particularly far or gets his care at another VA medical center and laboratory tests are needed, then I will reach out to their PCP via e-mail. If contact is not confirmed, I will reach out via phone or Skype.

Peter Glassman. The most helpful notes are those that are very specific as to what primary care is being asked to do and/or what urology is going to be doing. So, the more specific we get in the notes as to what is being addressed, I think that’s very helpful.

I have been here long enough that I’ve known both Alison and Bill; and if they have an issue, they will tap me a message. It wasn’t long ago that Bill sent a message to me, and we worked on a patient with prostate cancer who was going to be on long-term hormone therapy. We talked about osteoporosis management, and between us we worked out who was going to do what. Those are the kind of shared decision-making situations that are very, very helpful.
 

 

 

Alison Neymark. Also, GLAVAHCS has a home-based primary care team (HBPC), and a lot of the PCPs for that team are NPs. They know that they can contact me for their patients because a lot of those patients are on watchful waiting, and we do not necessarily need to see them face to face in clinic. Our urology team just needs to review updated lab results and how they are doing clinically. The HBPC NP who knows them best can contact me every 6 months or so, and we’ll discuss the case, which avoids making the patient come in, especially when they’re homebound. Those of us that have been working at the VA for many years have established good relationships. We feel very comfortable reaching out and talking to each other about these patients

Peter Glassman. Alison, I agree. When I can talk to my patients and say, “You know, we had that question about,” whatever the question might be, “and I contacted urology, and this is what they said.” It gives the patient confidence that we’re following up on the issues that they have and that we’re communicating with each other in a way that is to their benefit. And I think it’s very appreciated both by the provider as well as the patient.

William Aronson. Not infrequently I’ll have patients who have nonurologic issues, which I may first detect, or who have specific issues with their prostate cancer that can be comanaged. And I have found that when I send an encrypted e-mail to the PCP, it has been an extremely satisfying interaction; and we really get to the heart of the matter quickly for the sake of the veteran.

Veterans With Comorbidities

William Aronson. Posttraumatic stress disorder (PTSD) is a very significant and unique aspect of our patients, which is enormously important to recognize. For example, the side effects of prostate treatments can be very significant, whether radiation or surgery. Our patients understandably can be very fearful of the prostate cancer diagnosis and treatment side effects.

We know, for example, after a patient gets a diagnosis of prostate cancer, they’re at increased risk of cardiac death. That’s an especially important issue for our patients that there be an ongoing interaction between urology and primary care.

The ACS guidelines that Dr. Glassman referred to were enlightening. In many cases, primary care can look at the whole patient and their circumstances better than we can and may detect, for example, specific psychological issues that either they can manage or refer to other specialists.

Peter Glassman. One of the things that was highlighted in the ACS guideline is that in any population of men who have this disease, there’s going to be distress, anxiety, and full-fledged depression. Of course, there are psychosocial aspects of prostate cancer, such as sexual activity and intimacy with a partner that we often don’t explore but are probably playing an important role in the overall health of our patients. We need to be mindful of these psychosocial aspects and at least periodically ask them, “How are you doing with this? How are things at home?” And of course, we already use screeners for depression. As the article noted, distress and anxiety and other factors can make somebody’s life less optimal with poorer quality of life.

 

 

Dual Care Patients

Alison Neymark. Many patients whether they have Medicare, insurance through their spouse, or Kaiser Permanente through their job, choose to go to both places. The challenge is communicating with the non-VA providers because here at the VA we can communicate easily through Skype, Outlook e-mail, or CPRS, but for dual care patients who’s in charge? I encourage the veterans to choose whom they want to manage their care; we’re always here and happy to treat them, but they need to decide who’s in charge because I don’t want them to get into a situation where the differing opinions lead to a delay in care.

Nicholas Nickols. The communication when the patient is receiving care outside VA, either on a continuous basis or temporarily, is more of a challenge. We obviously can’t rely upon the messaging system, face-to-face contact is difficult, and they may not be able to use e-mail as well. So in those situations, usually a phone call is the best approach. I have found that the outside providers are happy to speak on the phone to coordinate care.

Peter Glassman. I agree, it does add a layer of complexity because we don’t readily have the notes, any information in front of us. That said, a lot of our patients can and do bring in information from outside specialists, and I’m hopeful that they share the information that we provide back to their outside doctors as well.

William Aronson. Some patient get nervous. They might decide they want care elsewhere, but they still want the VA available for them. I always let them know they should proceed in whatever way they prefer, but we’re always available and here for them. I try to empower them to make their own decisions and feel comfortable with them.

Nicholas Nickols. Notes from the outside, if they’re being referred for VA Choice or community care, do get uploaded into VistA Imaging and can be accessed, although it’s not instantaneous. Sometimes there’s a delay, but I have been able to access outside notes most of the time. If a patient goes through a clinic at the VA, the note is written in real time, and you can read it immediately.

Peter Glassman. That is true for patients that are within the VA system who receive contracted care either through Choice or through non-VA care that is contracted through VA. For somebody who is choosing to use 2 health care systems, that can provide more of a challenge because those notes don’t come to us. Over time, most of my patients have brought test results to me.

The thing with oncologic care, of course, is it’s a lot more complex. And it’s hard to know without reasonable documentation what’s been going on. At some level, you have to trust that the outside provider is doing whatever they need to do, or you have to take it upon yourself to do it within the system.

 

 

Alison Neymark. In my experience with the Choice Program, it really depends on the outside providers and how comfortable they are with the system that has been established to share records. Not all providers are going into that system and accessing it. I have had cases where I will see the non-VA provider’s note and it’ll say, “No documentation available for this consultation.” It just happens that they didn’t go into the system to review it. So it can be a challenge.

I’ve had good communication with the providers who use the system correctly. In some cases, just to make it easier, I will go ahead and communicate with them through encrypted e-mail, or I’ll talk to their care coordinators directly by phone. 

That way we can make sure everything is expedited to avoid any delay in the patient’s care. It is more time consuming, but it’s important because some of the procedures we don’t offer at the VA, and that’s why we’re using the Choice system.

Peter Glassman. Many, if not most, PCPs are going to take care of these patients, certainly within the VA, with their GU colleagues. And most of us feel comfortable using the current documentation system in a way that allows us to share information or at least to gather information about these patients.

One of the things that I think came out for me in looking at this was that there are guidelines or there are ideas out there on how to take better care of these patients. And I for one learned a fair bit just by going through these documents, which I’m very appreciative of. But it does highlight to me that we can give good care and provide good shared care for prostate cancer survivors. I think that is something that perhaps this discussion will highlight that not only are people doing that, but there are resources they can utilize that will help them get a more comprehensive picture of taking care of prostate cancer survivors in the primary care clinic.

The beauty of the VA system as a system is that as these issues come up that might affect the overall health of the veteran with prostate cancer, for example, psychosocial issues, we have many people that can address this that are experts in their area. And one of the great beauties of having an all-encompassing healthcare system is being able to use resources within the system, whether that be for other medical problems or other social or other psychological issues, that we ourselves are not expert in. We can reach out to our other colleagues and ask them for assistance. We have that available to help the patients. It’s really holistic.

We even have integrated medicine where we can help patients, hopefully, get back into a healthy lifestyle, for example, whereas we may not have that expertise or knowledge. We often think of this as sort of a shared decision between GU and primary care. But, in fact, it’s really the responsibility of many, many people of the system at large. We are very lucky to have that.

The following is a lightly edited transcript of a teleconference recorded in July 2018. The teleconference brought together health care providers from the Greater Los Angeles VA Health Care System (GLAVAHCS) to discuss the real-world processes for managing the treatment of patients with prostate cancer as they move between primary and specialist care.

William J. Aronson, MD. We are fortunate in having a superb medical record system at the Department of Veterans Affairs (VA) where we can all communicate with each other through a number of methods. Let’s start our discussion by reviewing an index patient that we see in our practice who has been treated with either radical prostatectomy or radiation therapy. One question to address is: Is there a point when the Urology or Radiation Oncology service can transition the patient’s entire care back to the primary care team? And if so, what would be the optimal way to accomplish this?

Nick, is there some point at which you discharge the patient from the radiation oncology service and give specific directions to primary care, or is it primarily just back to urology in your case?

Nicholas G. Nickols, MD, PhD. I have not discharged any patient from my clinic after definitive prostate cancer treatment. During treatment, patients are seen every week. Subsequently, I see them 6 weeks posttreatment, and then every 4 months for the first year, then every 6 months for the next 4 years, and then yearly after that. Although I never formally discharged a patient from my clinic, you can see based on the frequency of visits, that the patient will see more often than their primary care provider (PCP) toward the beginning. And then, after some years, the patient sees their primary more than they me. So it’s not an immediate hand off but rather a gradual transition. It’s important that the PCP is aware of what to look for especially for the late recurrences, late potential side effects, probably more significantly than the early side effects, how to manage them when appropriate, and when to ask the patient to see our team more frequently in follow-up.

William Aronson. We have a number of patients who travel tremendous distances to see us, and I tend to think that many of our follow-up patients, once things are stabilized with regards to management of their side effects, really could see their primary care doctors if we can give them specific instructions on, for example, when to get a prostate-specific antigen (PSA) test and when to refer back to us.

Alison, can you think of some specific cases where you feel like we’ve successfully done that?

Alison Neymark, MS. For the most part we haven’t discharged people, either. What we have done is transitioned them over to a phone clinic. In our department, we have 4 nurse practitioners (NPs) who each have a half-day of phone clinic where they call patients with their test results. Some of those patients are prostate cancer patients that we have been following for years. We schedule them for a phone call, whether it’s every 3 months, every 6 months or every year, to review the updated PSA level and to just check in with them by phone. It’s a win-win because it’s a really quick phone call to reassure the veteran that the PSA level is being followed, and it frees up an in-person appointment slot for another veteran.

 

 

We still have patients that prefer face-to-face visits, even though they know we’re not doing anything except discussing a PSA level with them—they just want that security of seeing our face. Some patients are very nervous, and they don’t necessarily want to be discharged, so to speak, back to primary care. Also, for those patients that travel a long distance to clinic, we offer an appointment in the video chat clinic, with the community-based outpatient clinics in Bakersfield and Santa Maria, California.

PSA Levels

William Aronson. I probably see a patient about every 4 to 6 weeks who has a low PSA after about 10 years and has a long distance to travel and mobility and other problems that make it difficult to come in. 

And so, in a number of those cases, I refer the patients back to their PCP and recommend that they get a PSA test every 6 months to a year. Then they refer back to us if there’s any further issues or if there is a significant rise in the PSA level.

The challenge that I have is, what is that specific guideline to give with regards to the rise in PSA? I think it all depends on the patients prostate cancer clinical features and comorbidities.

Nicholas Nickols. If a patient has been seen by me in follow-up a number of times and there’s really no active issues and there’s a low suspicion of recurrence, then I offer the patient the option of a phone follow-up as an alternative to face to face. Some of them accept that, but I ask that they agree to also see either urology or their PCP face to face. I will also remotely ensure that they’re getting the right laboratory tests, and if not, I’ll put those orders in.

With regard to when to refer a patient back for a suspected recurrence after definitive radiation therapy, there is an accepted definition of biochemical failure called the Phoenix definition, which is an absolute rise in 2 ng/mL of PSA over their posttreatment nadir. Often the posttreatment nadir, especially if they were on hormone therapy, will be close to 0. If the PSA gets to 2, that is a good trigger for a referral back to me and/or urology to discuss restaging and workup for a suspected recurrence.

For patients that are postsurgery and then subsequently get salvage radiation, it is not as clear when a restaging workup should be initiated. Currently, the imaging that is routine care is not very sensitive for detecting PSA in that setting until the PSA is around 0.8 ng/mL, and that’s with the most modern imaging available. Over time that may improve.

William Aronson. The other index patient to think about would be the patient who is on watchful waiting for their prostate cancer, which is to be distinguished from active surveillance. If someone’s on active surveillance, we’re regularly doing prostate biopsies and doing very close monitoring; but we also have patients who have multiple other medical problems, have a limited life expectancy, don’t have aggressive prostate cancer, and it’s extremely reasonable not to do a biopsy in those patients.

 

 

Again, those are patients where we do follow the PSA generally every 6 months. And I think there’s also scenarios there where it’s reasonable to refer back to primary care with specific instructions. These, again, are patients who had difficulty getting in to see us or have mobility issues, but it is also a way to limit patient visits if that’s their desire.

Peter Glassman, MBBS, MSc: I’m trained as both a general internist and board certified in hospice and palliative medicine. I currently provide primary care as well as palliative care. I view prostate cancer from the diagnosis through the treatment spectrum as a continuum. It starts with the PCP with an elevated PSA level or if the digital rectal exam has an abnormality, and then the role of the genitourinary (GU) practitioner becomes more significant during the active treatment and diagnostic phases.

Primary care doesn’t disappear, and I think there are 2 major issues that go along with that. First of all, we in primary care, because we take care of patients that often have other comorbidities, need to work with the patient on those comorbidities. Secondly, we need the information shared between the GU and primary care providers so that we can answer questions from our patients and have an understanding of what they’re going through and when.

As time goes on, we go through various phases: We may reach a cure, a quiescent period, active therapy, watchful waiting, or recurrence. Primary care gets involved as time goes on when the disease either becomes quiescent, is just being followed, or is considered cured. Clearly when you have watchful waiting, active treatment, or are in a recurrence, then GU takes the forefront.

I view it as a wave function. Primary care to GU with primary in smaller letters and then primary, if you will, in larger letters, GU becomes a lesser participant unless there is active therapy, watchful waiting or recurrence.

In doing a little bit of research, I found 2 very good and very helpful documents. One is the American Cancer Society (ACS) prostate cancer survivorship care guidelines (Box). And the other is a synopsis of the guidelines. What I liked was that the guidelines focused not only on what should be done for the initial period of prostate cancer, but also for many of the ancillary issues which we often don’t give voice to. The guidelines provide a structure, a foundation to work with our patients over time on their prostate cancer-related issues while, at the same time, being cognizant that we need to deal with their other comorbid conditions.

Modes of Communication

Alison Neymark. We find that including parameters for PSA monitoring in our Progress Notes in the electronic health record (EHR) the best way to communicate with other providers. We’ll say, “If PSA gets to this level, please refer back.” We try to make it clear because with the VA being a training facility, it could be a different resident/attending physician team that’s going to see the patient the next time he is in primary care.

 

 

Peter Glassman. Yes, we’re very lucky, as Bill talked about earlier and Alison just mentioned. We have the EHR, and Bill may remember this. Before the EHR, we were constantly fishing to find the most relevant notes. If a patient saw a GU practitioner the day before they saw me, I was often asking the patient what was said. Now we can just review the notes.

It’s a double-edged sword though because there are, of course, many notes in a medical record; and you have to look for the specific items. The EHR and documenting the medical record probably plays the primary role in getting information across. When you want to have an active handoff, or you need to communicate with each other, we have a variety of mechanisms, ranging from the phone to the Microsoft Skype Link (Redmond, WA) system that allows us to tap a message to a colleague.

And I’ve been here long enough that I’ve seen most permutations of how prostate cancer is diagnosed as well as shared among providers. Bill and I have shared patients. Alison and I have shared patients, not necessarily with prostate cancer, although that too. But we know how to communicate with each other. And of course, there’s paging if you need something more urgently.

William Aronson. We also use Microsoft Outlook e-mail, and encrypt the messages to keep them confidential and private. The other nice thing we have is there is a nationwide urology Outlook e-mail, so if any of us have any specific questions, through one e-mail we can send it around the country; and there’s usually multiple very useful responses. That’s another real strength of our system within the VA that helps patient care enormously.

Nicholas Nickols. Sometimes, if there’s a critical note that I absolutely want someone on the care team to read, I’ll add them as a cosigner; and that will pop up when they log in to the Computerized Patient Record System (CPRS) as something that they need to read.

If the patient lives particularly far or gets his care at another VA medical center and laboratory tests are needed, then I will reach out to their PCP via e-mail. If contact is not confirmed, I will reach out via phone or Skype.

Peter Glassman. The most helpful notes are those that are very specific as to what primary care is being asked to do and/or what urology is going to be doing. So, the more specific we get in the notes as to what is being addressed, I think that’s very helpful.

I have been here long enough that I’ve known both Alison and Bill; and if they have an issue, they will tap me a message. It wasn’t long ago that Bill sent a message to me, and we worked on a patient with prostate cancer who was going to be on long-term hormone therapy. We talked about osteoporosis management, and between us we worked out who was going to do what. Those are the kind of shared decision-making situations that are very, very helpful.
 

 

 

Alison Neymark. Also, GLAVAHCS has a home-based primary care team (HBPC), and a lot of the PCPs for that team are NPs. They know that they can contact me for their patients because a lot of those patients are on watchful waiting, and we do not necessarily need to see them face to face in clinic. Our urology team just needs to review updated lab results and how they are doing clinically. The HBPC NP who knows them best can contact me every 6 months or so, and we’ll discuss the case, which avoids making the patient come in, especially when they’re homebound. Those of us that have been working at the VA for many years have established good relationships. We feel very comfortable reaching out and talking to each other about these patients

Peter Glassman. Alison, I agree. When I can talk to my patients and say, “You know, we had that question about,” whatever the question might be, “and I contacted urology, and this is what they said.” It gives the patient confidence that we’re following up on the issues that they have and that we’re communicating with each other in a way that is to their benefit. And I think it’s very appreciated both by the provider as well as the patient.

William Aronson. Not infrequently I’ll have patients who have nonurologic issues, which I may first detect, or who have specific issues with their prostate cancer that can be comanaged. And I have found that when I send an encrypted e-mail to the PCP, it has been an extremely satisfying interaction; and we really get to the heart of the matter quickly for the sake of the veteran.

Veterans With Comorbidities

William Aronson. Posttraumatic stress disorder (PTSD) is a very significant and unique aspect of our patients, which is enormously important to recognize. For example, the side effects of prostate treatments can be very significant, whether radiation or surgery. Our patients understandably can be very fearful of the prostate cancer diagnosis and treatment side effects.

We know, for example, after a patient gets a diagnosis of prostate cancer, they’re at increased risk of cardiac death. That’s an especially important issue for our patients that there be an ongoing interaction between urology and primary care.

The ACS guidelines that Dr. Glassman referred to were enlightening. In many cases, primary care can look at the whole patient and their circumstances better than we can and may detect, for example, specific psychological issues that either they can manage or refer to other specialists.

Peter Glassman. One of the things that was highlighted in the ACS guideline is that in any population of men who have this disease, there’s going to be distress, anxiety, and full-fledged depression. Of course, there are psychosocial aspects of prostate cancer, such as sexual activity and intimacy with a partner that we often don’t explore but are probably playing an important role in the overall health of our patients. We need to be mindful of these psychosocial aspects and at least periodically ask them, “How are you doing with this? How are things at home?” And of course, we already use screeners for depression. As the article noted, distress and anxiety and other factors can make somebody’s life less optimal with poorer quality of life.

 

 

Dual Care Patients

Alison Neymark. Many patients whether they have Medicare, insurance through their spouse, or Kaiser Permanente through their job, choose to go to both places. The challenge is communicating with the non-VA providers because here at the VA we can communicate easily through Skype, Outlook e-mail, or CPRS, but for dual care patients who’s in charge? I encourage the veterans to choose whom they want to manage their care; we’re always here and happy to treat them, but they need to decide who’s in charge because I don’t want them to get into a situation where the differing opinions lead to a delay in care.

Nicholas Nickols. The communication when the patient is receiving care outside VA, either on a continuous basis or temporarily, is more of a challenge. We obviously can’t rely upon the messaging system, face-to-face contact is difficult, and they may not be able to use e-mail as well. So in those situations, usually a phone call is the best approach. I have found that the outside providers are happy to speak on the phone to coordinate care.

Peter Glassman. I agree, it does add a layer of complexity because we don’t readily have the notes, any information in front of us. That said, a lot of our patients can and do bring in information from outside specialists, and I’m hopeful that they share the information that we provide back to their outside doctors as well.

William Aronson. Some patient get nervous. They might decide they want care elsewhere, but they still want the VA available for them. I always let them know they should proceed in whatever way they prefer, but we’re always available and here for them. I try to empower them to make their own decisions and feel comfortable with them.

Nicholas Nickols. Notes from the outside, if they’re being referred for VA Choice or community care, do get uploaded into VistA Imaging and can be accessed, although it’s not instantaneous. Sometimes there’s a delay, but I have been able to access outside notes most of the time. If a patient goes through a clinic at the VA, the note is written in real time, and you can read it immediately.

Peter Glassman. That is true for patients that are within the VA system who receive contracted care either through Choice or through non-VA care that is contracted through VA. For somebody who is choosing to use 2 health care systems, that can provide more of a challenge because those notes don’t come to us. Over time, most of my patients have brought test results to me.

The thing with oncologic care, of course, is it’s a lot more complex. And it’s hard to know without reasonable documentation what’s been going on. At some level, you have to trust that the outside provider is doing whatever they need to do, or you have to take it upon yourself to do it within the system.

 

 

Alison Neymark. In my experience with the Choice Program, it really depends on the outside providers and how comfortable they are with the system that has been established to share records. Not all providers are going into that system and accessing it. I have had cases where I will see the non-VA provider’s note and it’ll say, “No documentation available for this consultation.” It just happens that they didn’t go into the system to review it. So it can be a challenge.

I’ve had good communication with the providers who use the system correctly. In some cases, just to make it easier, I will go ahead and communicate with them through encrypted e-mail, or I’ll talk to their care coordinators directly by phone. 

That way we can make sure everything is expedited to avoid any delay in the patient’s care. It is more time consuming, but it’s important because some of the procedures we don’t offer at the VA, and that’s why we’re using the Choice system.

Peter Glassman. Many, if not most, PCPs are going to take care of these patients, certainly within the VA, with their GU colleagues. And most of us feel comfortable using the current documentation system in a way that allows us to share information or at least to gather information about these patients.

One of the things that I think came out for me in looking at this was that there are guidelines or there are ideas out there on how to take better care of these patients. And I for one learned a fair bit just by going through these documents, which I’m very appreciative of. But it does highlight to me that we can give good care and provide good shared care for prostate cancer survivors. I think that is something that perhaps this discussion will highlight that not only are people doing that, but there are resources they can utilize that will help them get a more comprehensive picture of taking care of prostate cancer survivors in the primary care clinic.

The beauty of the VA system as a system is that as these issues come up that might affect the overall health of the veteran with prostate cancer, for example, psychosocial issues, we have many people that can address this that are experts in their area. And one of the great beauties of having an all-encompassing healthcare system is being able to use resources within the system, whether that be for other medical problems or other social or other psychological issues, that we ourselves are not expert in. We can reach out to our other colleagues and ask them for assistance. We have that available to help the patients. It’s really holistic.

We even have integrated medicine where we can help patients, hopefully, get back into a healthy lifestyle, for example, whereas we may not have that expertise or knowledge. We often think of this as sort of a shared decision between GU and primary care. But, in fact, it’s really the responsibility of many, many people of the system at large. We are very lucky to have that.

Issue
Federal Practitioner - 35(5)s
Issue
Federal Practitioner - 35(5)s
Page Number
S18-S23
Page Number
S18-S23
Publications
Publications
Topics
Article Type
Display Headline
Coordination of Care Between Primary Care and Oncology for Patients With Prostate Cancer
Display Headline
Coordination of Care Between Primary Care and Oncology for Patients With Prostate Cancer
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Immune Checkpoint Inhibitors for Urothelial Cancer: An Update on New Therapies (FULL)

Article Type
Changed
Thu, 12/15/2022 - 14:41
Display Headline
Immune Checkpoint Inhibitors for Urothelial Cancer: An Update on New Therapies
Ongoing trials are evaluating immune checkpoint inhibitors—used alone, in combination with cytotoxic, targeted, radiation therapies, or with other such inhibitors—for therapy in patients with advanced bladder cancer.

An essential feature of cancer is its ability to evade the immune system. Multiple mechanisms are used for this purpose, including the disruption of antigen presentation and suppression of the immune response. The latter mechanism involves the activation of T-cell inhibition by recruiting regulatory T cells that weaken this response. Recent progress in understanding the ability of cancer to evade the immune system has paved the way to develop strategies to reverse this process and reactivate the immune system. Particularly, immune checkpoint signaling between T cells and tumor cells has been targeted with a new class of drug, immune checkpoint inhibitors. Immunotherapy has been an established and effective treatment in bladder cancer since 1976 when Morales and colleagues demonstrated that intravesical treatments with bacillus Calmette-Guérin can treat carcinoma in situ and prevent nonmuscle invasive urothelial cancer recurrence.1,2 This treatment elicits a cytotoxic response via antigenic presentation by bladder tumor cells.

Cytotoxic T-lymphocyte-associated protein (CTLA)-4, programmed death-1 (PD-1) and programmed death-ligand-1 (PD-L1) are molecules that downregulate the immune response and are targets of therapeutic antibodies that have demonstrated clinical efficacy across a wide range of malignancies. Five such agents—pembrolizumab, atezolizumab, nivolumab, avelumab and durvalumab—were recently approved by the US Food and Drug Administration (FDA) for clinical use in patients with advanced urothelial cancers.3 This class of agents also has been approved for several other malignancies, most notably in melanoma, non-small cell lung cancer, and renal cell carcinoma.3

Immune Biology

CTLA-4 is expressed on activated CD4 and CD8 T cells and competes with CD28 on T cells to interact with the costimulatory B7 proteins on antigen presenting cells. The CD28/B7 interaction promotes T-cell activation and effector functions, and the CTLA-4/B7 interaction inhibits them. In addition, PD-1 is a receptor expressed on CD4 and CD8 T cells, T regulatory (Treg) cells, B cells and natural killer (NK) cells that interacts with its ligand PD-L1 to suppress the immune response. Urothelial cancer possesses features that make it an adequate target for immunotherapeutic agents. Primarily, it is characterized by a high-mutation load, which lends itself to an increased expression of immunogenic antigens on tumor cells.4

Immunotherapy Treatments in Cisplatin-Ineligible Patients

Cisplatin-based chemotherapy is the first-line treatment and standard of care in unresectable or metastatic urothelial cancer. However, many patients are unable to receive cisplatin secondary to renal dysfunction, poor performance status, or other comorbidities. Alternative cytotoxic therapies in the first-line setting such as carboplatin-based regimens are associated with inferior outcomes and poor tolerability. There is, therefore, a need for effective and well-tolerated therapies in cisplatin-ineligible patients (Table).

In the phase 2 Keynote-052 trial, 370 cisplatin-ineligible patients were treated with the anti-PD-1 antibody pembrolizumab 200 mg every 3 weeks for up to 2 years.5At a median follow-up of 9.5 months, the objective response rate (+ORR) was 29% for the entire cohort, with a 7% complete response (CR) rate, and a 22% partial response (PR) rate.5 The median duration of response had not been reached at the time of analysis. Responses were seen regardless of PD-L1 expression, although high response rates were noted in patients whose tumors had PD-L1 expression > 10%. Pembrolizumab had an acceptable tolerability profile in this population. The most common grade 3 or 4 treatment-related adverse event (AE) was fatigue at 2%; 5% of patients discontinued therapy due to treatment related AEs, whereas 17% of patients had immune-mediated AEs.5

Similarly, in a single-arm phase 2 trial, atezolizumab, an anti-PD-L1 antibody, dosed at 1,200 mg every 3 weeks was used as first-line therapy in 119 patients with advanced urothelial cancer who were cisplatin ineligible. At a median follow-up of 17 months, the ORR was 23%, with a 9% CR rate. The median duration of response had not been reached. Median progression free survival (PFS) was 2.7 months, whereas overall survival (OS) was 16 months. Eight percent of patients had an AE leading to treatment discontinuation, and 17% had immune-mediated AEs.6 Both pembrolizumab and atezolizumab were granted FDA approval in 2017 for patients with locally advanced or metastatic urothelial carcinoma who are not eligible for cisplatin-based chemotherapy.3

 

 

Immunotherapy Treatments After Progression With Cisplatin

Cytotoxic chemotherapy in the second-line setting with disease progression following platinum-based treatment has shown dismal responses, with a median OS of about 6 to 7 months.7 Immunotherapy provides an effective and a much-needed option in this scenario.

Five antibodies targeting the PD-1/PD-L1 pathway, pembrolizumab, nivolumab, atezolizumab, avelumab and durvalumab, have been granted FDA approval for patients who have progressed during or after platinum-based therapy (Table).3 In the phase 3 Keynote-045 trial, 542 patients were randomly assigned to receive either pembrolizumab 200 mg administered every 3 weeks or investigator’s choice chemotherapy (paclitaxel, docetaxel, or vinflunine).7 Median OS was 10.3 months in the pembrolizumab group and 7.4 months in the chemotherapy group (hazard ratio for death, 0.73; P = .002). Serious (grade 3 or above) treatment-related AEs were significantly less frequent with pembrolizumab (15% vs 49.4%).7 In a phase 2 trial, 270 patients were treated with nivolumab, a PD-1 inhibitor, at a dose of 3 mg/kg given every 2 weeks.8 The ORR was 19.6%, while the median OS for the entire cohort was 7 months. Responses were seen at all levels of PD-L1 expression, although in patients whose tumor expressed PD-L1 ≥1%, median OS was 11.3 months.8

It should be noted that in a large phase 3 trial comparing atezolizumab with chemotherapy in the second-line setting, ORR and OS were not statistically different between the 2 groups, although the duration of response was longer with atezolizumab.9 In early phase trials, avelumab and durvalumab, both PD-L1 inhibitors showed an ORR of about 17%, with higher ORR seen in patients with tumors positive for PD-L1 expression.10,11 The AE profile of immune checkpoint inhibitors is relatively favorable in clinical trials. The American Society of Clinical Oncology and National Comprehensive Cancer Network have jointly published evidence-based guidelines for the management of their immune related AEs.12

Future Directions

Several challenges have emerged with immunotherapy treatments. One issue is the relatively low ORRs for immune checkpoint inhibitors, ranging from 13.4% to 24% depending on the trial. Therefore, there is a need to identify reliable biomarkers and selection criteria to predict their efficacy and improve patient selection. Although tumor PD-L1 expression has shown some usefulness in this setting, responses have been noted in patients whose tumors have low or no expression of PD-L1. This low predictive accuracy is caused by several factors, including PD-L1 intratumor expression heterogeneity, primary vs metastatic site PD-L1 expression heterogeneity, lack of consensus on which PD-L1 assays and which value cutoffs to use, and the differences seen in marker expression depending on the freshness of the tissue specimen.

Other predictive biomarkers with potential include tumor gene expression profiles/tumor mutational load, T-cell and B-cell signatures. The optimal imaging modality and timing of this imaging for response assessment also is uncertain. So-called tumor pseudo-progression seen on imaging after treatment with these agents as a result of the immune/inflammatory response to the tumor is now a well-recognized phenomenon, but it can be challenging to differentiate from true disease progression. Other challenges include deciding on which immune checkpoint inhibitor to use given a lack of head-to-head comparisons of these immunotherapeutic agents, finding the proper drug doses to maximize efficacy, as well as determining the optimal duration of treatment in patients with continued response to immunotherapy. Many oncologists continue these treatments for up to 2 years in the setting of a significant or complete response.

 

 

Conclusion

Immune checkpoint inhibitors have emerged as pivotal treatments for patients with advanced urothelial cancer who are unfit to receive cisplatin in the first-line setting or who experience disease progression after cisplatin-based chemotherapy. This field continues to expand at a rapid pace due to multiple ongoing clinical trials assessing these agents, whether alone, in combination with cytotoxic, targeted, radiation therapies, or with other immune checkpoint inhibitors, both in the advanced as well as the neoadjuvant/adjuvant settings.

References

1. Morales A, Eidinger D, Bruce AW. Intracavitary bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J Urol. 1976;116(2):180-183.

2. Morales A. Treatment of carcinoma in situ of the bladder with BCG. Cancer Immunol Immunother. 1980;9 (1-2):69-72.

3. US Food and drug administration. FDA approved drug products. www.accessdata.fda.gov/scripts/cder/daf/index.cfm. Accessed July 5, 2018.

4. Farina MS, Lundgren KT, Bellmunt J. Immunotherapy in urothelial cancer: recent results and future perspectives. Drugs. 2017;77(10):1077-1089.

5. Balar AV, Castellano DE, O’Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18(11):1483-1492.

6. Balar AV, Galsky MD, Rosenberg JE, et al; IMvigor210 Study Group. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389(10064):67-76.

7. Bellmunt J, de Wit R, Vaughn DJ, et al; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376(11):1015-1026.

8. Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18(3):312-322.

9. Powles T, Durán I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748-757.

10. Patel MR, Ellerton J, Infante JR, et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol. 2018;19(1):51-64.

11. Powles T, O’Donnell PH, Massard C, et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 2017;3(9):e172411.

12. Brahmer JR, Lacchetti C, Schneider BJ, et al; National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2018;36(17):1714-1768.

Article PDF
Author and Disclosure Information

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Dr. El-Mouallem is a Fellow in Hematology and Medical Oncology at the Hunter Holmes McGuire VA Medical Center in Richmond, Virginia, and a Fellow in the Division of Hematology, Oncology, and Palliative Care at Virginia Commonwealth University (VCU) in Richmond. Dr. Paul is Assistant Clinical Professor in the Division of Hematology, Oncology, and Palliative Care at Massey Cancer Center at VCU Medical Center.
Correspondence: Dr. Paul ([email protected])

Issue
Federal Practitioner - 35(5)s
Publications
Topics
Page Number
S62-S64
Sections
Author and Disclosure Information

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Dr. El-Mouallem is a Fellow in Hematology and Medical Oncology at the Hunter Holmes McGuire VA Medical Center in Richmond, Virginia, and a Fellow in the Division of Hematology, Oncology, and Palliative Care at Virginia Commonwealth University (VCU) in Richmond. Dr. Paul is Assistant Clinical Professor in the Division of Hematology, Oncology, and Palliative Care at Massey Cancer Center at VCU Medical Center.
Correspondence: Dr. Paul ([email protected])

Author and Disclosure Information

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies. This article may discuss unlabeled or investigational use of certain drugs. Please review the complete prescribing information for specific drugs or drug combinations—including indications, contraindications, warnings, and adverse effects—before administering pharmacologic therapy to patients.

Dr. El-Mouallem is a Fellow in Hematology and Medical Oncology at the Hunter Holmes McGuire VA Medical Center in Richmond, Virginia, and a Fellow in the Division of Hematology, Oncology, and Palliative Care at Virginia Commonwealth University (VCU) in Richmond. Dr. Paul is Assistant Clinical Professor in the Division of Hematology, Oncology, and Palliative Care at Massey Cancer Center at VCU Medical Center.
Correspondence: Dr. Paul ([email protected])

Article PDF
Article PDF
Ongoing trials are evaluating immune checkpoint inhibitors—used alone, in combination with cytotoxic, targeted, radiation therapies, or with other such inhibitors—for therapy in patients with advanced bladder cancer.
Ongoing trials are evaluating immune checkpoint inhibitors—used alone, in combination with cytotoxic, targeted, radiation therapies, or with other such inhibitors—for therapy in patients with advanced bladder cancer.

An essential feature of cancer is its ability to evade the immune system. Multiple mechanisms are used for this purpose, including the disruption of antigen presentation and suppression of the immune response. The latter mechanism involves the activation of T-cell inhibition by recruiting regulatory T cells that weaken this response. Recent progress in understanding the ability of cancer to evade the immune system has paved the way to develop strategies to reverse this process and reactivate the immune system. Particularly, immune checkpoint signaling between T cells and tumor cells has been targeted with a new class of drug, immune checkpoint inhibitors. Immunotherapy has been an established and effective treatment in bladder cancer since 1976 when Morales and colleagues demonstrated that intravesical treatments with bacillus Calmette-Guérin can treat carcinoma in situ and prevent nonmuscle invasive urothelial cancer recurrence.1,2 This treatment elicits a cytotoxic response via antigenic presentation by bladder tumor cells.

Cytotoxic T-lymphocyte-associated protein (CTLA)-4, programmed death-1 (PD-1) and programmed death-ligand-1 (PD-L1) are molecules that downregulate the immune response and are targets of therapeutic antibodies that have demonstrated clinical efficacy across a wide range of malignancies. Five such agents—pembrolizumab, atezolizumab, nivolumab, avelumab and durvalumab—were recently approved by the US Food and Drug Administration (FDA) for clinical use in patients with advanced urothelial cancers.3 This class of agents also has been approved for several other malignancies, most notably in melanoma, non-small cell lung cancer, and renal cell carcinoma.3

Immune Biology

CTLA-4 is expressed on activated CD4 and CD8 T cells and competes with CD28 on T cells to interact with the costimulatory B7 proteins on antigen presenting cells. The CD28/B7 interaction promotes T-cell activation and effector functions, and the CTLA-4/B7 interaction inhibits them. In addition, PD-1 is a receptor expressed on CD4 and CD8 T cells, T regulatory (Treg) cells, B cells and natural killer (NK) cells that interacts with its ligand PD-L1 to suppress the immune response. Urothelial cancer possesses features that make it an adequate target for immunotherapeutic agents. Primarily, it is characterized by a high-mutation load, which lends itself to an increased expression of immunogenic antigens on tumor cells.4

Immunotherapy Treatments in Cisplatin-Ineligible Patients

Cisplatin-based chemotherapy is the first-line treatment and standard of care in unresectable or metastatic urothelial cancer. However, many patients are unable to receive cisplatin secondary to renal dysfunction, poor performance status, or other comorbidities. Alternative cytotoxic therapies in the first-line setting such as carboplatin-based regimens are associated with inferior outcomes and poor tolerability. There is, therefore, a need for effective and well-tolerated therapies in cisplatin-ineligible patients (Table).

In the phase 2 Keynote-052 trial, 370 cisplatin-ineligible patients were treated with the anti-PD-1 antibody pembrolizumab 200 mg every 3 weeks for up to 2 years.5At a median follow-up of 9.5 months, the objective response rate (+ORR) was 29% for the entire cohort, with a 7% complete response (CR) rate, and a 22% partial response (PR) rate.5 The median duration of response had not been reached at the time of analysis. Responses were seen regardless of PD-L1 expression, although high response rates were noted in patients whose tumors had PD-L1 expression > 10%. Pembrolizumab had an acceptable tolerability profile in this population. The most common grade 3 or 4 treatment-related adverse event (AE) was fatigue at 2%; 5% of patients discontinued therapy due to treatment related AEs, whereas 17% of patients had immune-mediated AEs.5

Similarly, in a single-arm phase 2 trial, atezolizumab, an anti-PD-L1 antibody, dosed at 1,200 mg every 3 weeks was used as first-line therapy in 119 patients with advanced urothelial cancer who were cisplatin ineligible. At a median follow-up of 17 months, the ORR was 23%, with a 9% CR rate. The median duration of response had not been reached. Median progression free survival (PFS) was 2.7 months, whereas overall survival (OS) was 16 months. Eight percent of patients had an AE leading to treatment discontinuation, and 17% had immune-mediated AEs.6 Both pembrolizumab and atezolizumab were granted FDA approval in 2017 for patients with locally advanced or metastatic urothelial carcinoma who are not eligible for cisplatin-based chemotherapy.3

 

 

Immunotherapy Treatments After Progression With Cisplatin

Cytotoxic chemotherapy in the second-line setting with disease progression following platinum-based treatment has shown dismal responses, with a median OS of about 6 to 7 months.7 Immunotherapy provides an effective and a much-needed option in this scenario.

Five antibodies targeting the PD-1/PD-L1 pathway, pembrolizumab, nivolumab, atezolizumab, avelumab and durvalumab, have been granted FDA approval for patients who have progressed during or after platinum-based therapy (Table).3 In the phase 3 Keynote-045 trial, 542 patients were randomly assigned to receive either pembrolizumab 200 mg administered every 3 weeks or investigator’s choice chemotherapy (paclitaxel, docetaxel, or vinflunine).7 Median OS was 10.3 months in the pembrolizumab group and 7.4 months in the chemotherapy group (hazard ratio for death, 0.73; P = .002). Serious (grade 3 or above) treatment-related AEs were significantly less frequent with pembrolizumab (15% vs 49.4%).7 In a phase 2 trial, 270 patients were treated with nivolumab, a PD-1 inhibitor, at a dose of 3 mg/kg given every 2 weeks.8 The ORR was 19.6%, while the median OS for the entire cohort was 7 months. Responses were seen at all levels of PD-L1 expression, although in patients whose tumor expressed PD-L1 ≥1%, median OS was 11.3 months.8

It should be noted that in a large phase 3 trial comparing atezolizumab with chemotherapy in the second-line setting, ORR and OS were not statistically different between the 2 groups, although the duration of response was longer with atezolizumab.9 In early phase trials, avelumab and durvalumab, both PD-L1 inhibitors showed an ORR of about 17%, with higher ORR seen in patients with tumors positive for PD-L1 expression.10,11 The AE profile of immune checkpoint inhibitors is relatively favorable in clinical trials. The American Society of Clinical Oncology and National Comprehensive Cancer Network have jointly published evidence-based guidelines for the management of their immune related AEs.12

Future Directions

Several challenges have emerged with immunotherapy treatments. One issue is the relatively low ORRs for immune checkpoint inhibitors, ranging from 13.4% to 24% depending on the trial. Therefore, there is a need to identify reliable biomarkers and selection criteria to predict their efficacy and improve patient selection. Although tumor PD-L1 expression has shown some usefulness in this setting, responses have been noted in patients whose tumors have low or no expression of PD-L1. This low predictive accuracy is caused by several factors, including PD-L1 intratumor expression heterogeneity, primary vs metastatic site PD-L1 expression heterogeneity, lack of consensus on which PD-L1 assays and which value cutoffs to use, and the differences seen in marker expression depending on the freshness of the tissue specimen.

Other predictive biomarkers with potential include tumor gene expression profiles/tumor mutational load, T-cell and B-cell signatures. The optimal imaging modality and timing of this imaging for response assessment also is uncertain. So-called tumor pseudo-progression seen on imaging after treatment with these agents as a result of the immune/inflammatory response to the tumor is now a well-recognized phenomenon, but it can be challenging to differentiate from true disease progression. Other challenges include deciding on which immune checkpoint inhibitor to use given a lack of head-to-head comparisons of these immunotherapeutic agents, finding the proper drug doses to maximize efficacy, as well as determining the optimal duration of treatment in patients with continued response to immunotherapy. Many oncologists continue these treatments for up to 2 years in the setting of a significant or complete response.

 

 

Conclusion

Immune checkpoint inhibitors have emerged as pivotal treatments for patients with advanced urothelial cancer who are unfit to receive cisplatin in the first-line setting or who experience disease progression after cisplatin-based chemotherapy. This field continues to expand at a rapid pace due to multiple ongoing clinical trials assessing these agents, whether alone, in combination with cytotoxic, targeted, radiation therapies, or with other immune checkpoint inhibitors, both in the advanced as well as the neoadjuvant/adjuvant settings.

An essential feature of cancer is its ability to evade the immune system. Multiple mechanisms are used for this purpose, including the disruption of antigen presentation and suppression of the immune response. The latter mechanism involves the activation of T-cell inhibition by recruiting regulatory T cells that weaken this response. Recent progress in understanding the ability of cancer to evade the immune system has paved the way to develop strategies to reverse this process and reactivate the immune system. Particularly, immune checkpoint signaling between T cells and tumor cells has been targeted with a new class of drug, immune checkpoint inhibitors. Immunotherapy has been an established and effective treatment in bladder cancer since 1976 when Morales and colleagues demonstrated that intravesical treatments with bacillus Calmette-Guérin can treat carcinoma in situ and prevent nonmuscle invasive urothelial cancer recurrence.1,2 This treatment elicits a cytotoxic response via antigenic presentation by bladder tumor cells.

Cytotoxic T-lymphocyte-associated protein (CTLA)-4, programmed death-1 (PD-1) and programmed death-ligand-1 (PD-L1) are molecules that downregulate the immune response and are targets of therapeutic antibodies that have demonstrated clinical efficacy across a wide range of malignancies. Five such agents—pembrolizumab, atezolizumab, nivolumab, avelumab and durvalumab—were recently approved by the US Food and Drug Administration (FDA) for clinical use in patients with advanced urothelial cancers.3 This class of agents also has been approved for several other malignancies, most notably in melanoma, non-small cell lung cancer, and renal cell carcinoma.3

Immune Biology

CTLA-4 is expressed on activated CD4 and CD8 T cells and competes with CD28 on T cells to interact with the costimulatory B7 proteins on antigen presenting cells. The CD28/B7 interaction promotes T-cell activation and effector functions, and the CTLA-4/B7 interaction inhibits them. In addition, PD-1 is a receptor expressed on CD4 and CD8 T cells, T regulatory (Treg) cells, B cells and natural killer (NK) cells that interacts with its ligand PD-L1 to suppress the immune response. Urothelial cancer possesses features that make it an adequate target for immunotherapeutic agents. Primarily, it is characterized by a high-mutation load, which lends itself to an increased expression of immunogenic antigens on tumor cells.4

Immunotherapy Treatments in Cisplatin-Ineligible Patients

Cisplatin-based chemotherapy is the first-line treatment and standard of care in unresectable or metastatic urothelial cancer. However, many patients are unable to receive cisplatin secondary to renal dysfunction, poor performance status, or other comorbidities. Alternative cytotoxic therapies in the first-line setting such as carboplatin-based regimens are associated with inferior outcomes and poor tolerability. There is, therefore, a need for effective and well-tolerated therapies in cisplatin-ineligible patients (Table).

In the phase 2 Keynote-052 trial, 370 cisplatin-ineligible patients were treated with the anti-PD-1 antibody pembrolizumab 200 mg every 3 weeks for up to 2 years.5At a median follow-up of 9.5 months, the objective response rate (+ORR) was 29% for the entire cohort, with a 7% complete response (CR) rate, and a 22% partial response (PR) rate.5 The median duration of response had not been reached at the time of analysis. Responses were seen regardless of PD-L1 expression, although high response rates were noted in patients whose tumors had PD-L1 expression > 10%. Pembrolizumab had an acceptable tolerability profile in this population. The most common grade 3 or 4 treatment-related adverse event (AE) was fatigue at 2%; 5% of patients discontinued therapy due to treatment related AEs, whereas 17% of patients had immune-mediated AEs.5

Similarly, in a single-arm phase 2 trial, atezolizumab, an anti-PD-L1 antibody, dosed at 1,200 mg every 3 weeks was used as first-line therapy in 119 patients with advanced urothelial cancer who were cisplatin ineligible. At a median follow-up of 17 months, the ORR was 23%, with a 9% CR rate. The median duration of response had not been reached. Median progression free survival (PFS) was 2.7 months, whereas overall survival (OS) was 16 months. Eight percent of patients had an AE leading to treatment discontinuation, and 17% had immune-mediated AEs.6 Both pembrolizumab and atezolizumab were granted FDA approval in 2017 for patients with locally advanced or metastatic urothelial carcinoma who are not eligible for cisplatin-based chemotherapy.3

 

 

Immunotherapy Treatments After Progression With Cisplatin

Cytotoxic chemotherapy in the second-line setting with disease progression following platinum-based treatment has shown dismal responses, with a median OS of about 6 to 7 months.7 Immunotherapy provides an effective and a much-needed option in this scenario.

Five antibodies targeting the PD-1/PD-L1 pathway, pembrolizumab, nivolumab, atezolizumab, avelumab and durvalumab, have been granted FDA approval for patients who have progressed during or after platinum-based therapy (Table).3 In the phase 3 Keynote-045 trial, 542 patients were randomly assigned to receive either pembrolizumab 200 mg administered every 3 weeks or investigator’s choice chemotherapy (paclitaxel, docetaxel, or vinflunine).7 Median OS was 10.3 months in the pembrolizumab group and 7.4 months in the chemotherapy group (hazard ratio for death, 0.73; P = .002). Serious (grade 3 or above) treatment-related AEs were significantly less frequent with pembrolizumab (15% vs 49.4%).7 In a phase 2 trial, 270 patients were treated with nivolumab, a PD-1 inhibitor, at a dose of 3 mg/kg given every 2 weeks.8 The ORR was 19.6%, while the median OS for the entire cohort was 7 months. Responses were seen at all levels of PD-L1 expression, although in patients whose tumor expressed PD-L1 ≥1%, median OS was 11.3 months.8

It should be noted that in a large phase 3 trial comparing atezolizumab with chemotherapy in the second-line setting, ORR and OS were not statistically different between the 2 groups, although the duration of response was longer with atezolizumab.9 In early phase trials, avelumab and durvalumab, both PD-L1 inhibitors showed an ORR of about 17%, with higher ORR seen in patients with tumors positive for PD-L1 expression.10,11 The AE profile of immune checkpoint inhibitors is relatively favorable in clinical trials. The American Society of Clinical Oncology and National Comprehensive Cancer Network have jointly published evidence-based guidelines for the management of their immune related AEs.12

Future Directions

Several challenges have emerged with immunotherapy treatments. One issue is the relatively low ORRs for immune checkpoint inhibitors, ranging from 13.4% to 24% depending on the trial. Therefore, there is a need to identify reliable biomarkers and selection criteria to predict their efficacy and improve patient selection. Although tumor PD-L1 expression has shown some usefulness in this setting, responses have been noted in patients whose tumors have low or no expression of PD-L1. This low predictive accuracy is caused by several factors, including PD-L1 intratumor expression heterogeneity, primary vs metastatic site PD-L1 expression heterogeneity, lack of consensus on which PD-L1 assays and which value cutoffs to use, and the differences seen in marker expression depending on the freshness of the tissue specimen.

Other predictive biomarkers with potential include tumor gene expression profiles/tumor mutational load, T-cell and B-cell signatures. The optimal imaging modality and timing of this imaging for response assessment also is uncertain. So-called tumor pseudo-progression seen on imaging after treatment with these agents as a result of the immune/inflammatory response to the tumor is now a well-recognized phenomenon, but it can be challenging to differentiate from true disease progression. Other challenges include deciding on which immune checkpoint inhibitor to use given a lack of head-to-head comparisons of these immunotherapeutic agents, finding the proper drug doses to maximize efficacy, as well as determining the optimal duration of treatment in patients with continued response to immunotherapy. Many oncologists continue these treatments for up to 2 years in the setting of a significant or complete response.

 

 

Conclusion

Immune checkpoint inhibitors have emerged as pivotal treatments for patients with advanced urothelial cancer who are unfit to receive cisplatin in the first-line setting or who experience disease progression after cisplatin-based chemotherapy. This field continues to expand at a rapid pace due to multiple ongoing clinical trials assessing these agents, whether alone, in combination with cytotoxic, targeted, radiation therapies, or with other immune checkpoint inhibitors, both in the advanced as well as the neoadjuvant/adjuvant settings.

References

1. Morales A, Eidinger D, Bruce AW. Intracavitary bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J Urol. 1976;116(2):180-183.

2. Morales A. Treatment of carcinoma in situ of the bladder with BCG. Cancer Immunol Immunother. 1980;9 (1-2):69-72.

3. US Food and drug administration. FDA approved drug products. www.accessdata.fda.gov/scripts/cder/daf/index.cfm. Accessed July 5, 2018.

4. Farina MS, Lundgren KT, Bellmunt J. Immunotherapy in urothelial cancer: recent results and future perspectives. Drugs. 2017;77(10):1077-1089.

5. Balar AV, Castellano DE, O’Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18(11):1483-1492.

6. Balar AV, Galsky MD, Rosenberg JE, et al; IMvigor210 Study Group. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389(10064):67-76.

7. Bellmunt J, de Wit R, Vaughn DJ, et al; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376(11):1015-1026.

8. Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18(3):312-322.

9. Powles T, Durán I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748-757.

10. Patel MR, Ellerton J, Infante JR, et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol. 2018;19(1):51-64.

11. Powles T, O’Donnell PH, Massard C, et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 2017;3(9):e172411.

12. Brahmer JR, Lacchetti C, Schneider BJ, et al; National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2018;36(17):1714-1768.

References

1. Morales A, Eidinger D, Bruce AW. Intracavitary bacillus Calmette-Guerin in the treatment of superficial bladder tumors. J Urol. 1976;116(2):180-183.

2. Morales A. Treatment of carcinoma in situ of the bladder with BCG. Cancer Immunol Immunother. 1980;9 (1-2):69-72.

3. US Food and drug administration. FDA approved drug products. www.accessdata.fda.gov/scripts/cder/daf/index.cfm. Accessed July 5, 2018.

4. Farina MS, Lundgren KT, Bellmunt J. Immunotherapy in urothelial cancer: recent results and future perspectives. Drugs. 2017;77(10):1077-1089.

5. Balar AV, Castellano DE, O’Donnell PH, et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): a multicentre, single-arm, phase 2 study. Lancet Oncol. 2017;18(11):1483-1492.

6. Balar AV, Galsky MD, Rosenberg JE, et al; IMvigor210 Study Group. Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial. Lancet. 2017;389(10064):67-76.

7. Bellmunt J, de Wit R, Vaughn DJ, et al; KEYNOTE-045 Investigators. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 2017;376(11):1015-1026.

8. Sharma P, Retz M, Siefker-Radtke A, et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol. 2017;18(3):312-322.

9. Powles T, Durán I, van der Heijden MS, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2018;391(10122):748-757.

10. Patel MR, Ellerton J, Infante JR, et al. Avelumab in metastatic urothelial carcinoma after platinum failure (JAVELIN Solid Tumor): pooled results from two expansion cohorts of an open-label, phase 1 trial. Lancet Oncol. 2018;19(1):51-64.

11. Powles T, O’Donnell PH, Massard C, et al. Efficacy and safety of durvalumab in locally advanced or metastatic urothelial carcinoma: updated results from a phase 1/2 open-label study. JAMA Oncol. 2017;3(9):e172411.

12. Brahmer JR, Lacchetti C, Schneider BJ, et al; National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2018;36(17):1714-1768.

Issue
Federal Practitioner - 35(5)s
Issue
Federal Practitioner - 35(5)s
Page Number
S62-S64
Page Number
S62-S64
Publications
Publications
Topics
Article Type
Display Headline
Immune Checkpoint Inhibitors for Urothelial Cancer: An Update on New Therapies
Display Headline
Immune Checkpoint Inhibitors for Urothelial Cancer: An Update on New Therapies
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Article PDF Media

Accuracy of Endoscopic Ultrasound in Staging of Early Rectal Cancer (FULL)

Article Type
Changed
Thu, 12/15/2022 - 14:42
Display Headline
Accuracy of Endoscopic Ultrasound in Staging of Early Rectal Cancer

Endoscopic ultrasound can be highly accurate for the staging of neoplasms in early rectal cancer.

Colorectal cancer is the second most common cause of cancer death in the US, with one-third of all colorectal cancers occurring within the rectum. Each year, an estimated 40000 Americans are diagnosed with rectal cancer (RC).1,2 The prognosis and treatment of RC depends on both T and N stage at the time of diagnosis.3-5 According to the most recent National Comprehensive Cancer Network guidelines from May 2019, patients with T1 to T2N0 tumors should undergo transanal or transabdominal surgery upfront, whereas patients with T3 to T4N0 or any TN1 to 2 should start with neoadjuvant therapy for better locoregional control, followed by surgery.6 Therefore, the appropriate management of RC requires adequate staging.

Endoscopic ultrasound (EUS), magnetic resonance imaging (MRI), and computed tomography (CT) are the imaging techniques currently used to stage RC. In a meta-analysis of 90 articles published between 1985 and 2002 that compared the 3 radiologic modalities, Bipat and colleagues found that MRI and EUS had a similar sensitivity of 94%, whereas the specificity of EUS (86%) was significantly higher than that of MRI (69%) for muscularis propria invasion.7 CT was performed only in a limited number of trials because CT was considered inadequate to assess early T stage. For perirectal tissue invasion, the sensitivity of EUS was statistically higher than that of CT and MRI imaging: 90% compared with 79% and 82%, respectively. The specificity estimates for EUS, CT, and MRI were comparable: 75%, 78%, and 76%, respectively. The respective sensitivity and specificity of the 3 imaging modalities to evaluate lymph nodes were also comparable: EUS, 67% and 78%; CT, 55% and 74%; and MRI, 66% and 76%.

The role of EUS in the diagnosis and treatment of RC has long been validated.1,2-5 A meta-analysis of 42 studies involving 5039 patients found EUS to be highly accurate for differentiating various T stages.8 However, EUS cannot assess iliac and mesenteric lymph nodes or posterior tumor extension beyond endopelvic fascia in advanced RC. Notable heterogeneity was found among the studies in the meta-analyses with regard to the type of equipment used for staging, as well as the criteria used to assess the depth of penetration and nodal status. The recent introduction of phased-array coils and the development of T2-weighted fast spin sequences have improved the resolution of MRI. The MERCURY trial showed that extension of tumor to within 1 mm of the circumferential margin on high-resolution MRI correctly predicted margin involvement at the time of surgery in 92% of the patients.9 In the retrospective study by Balyasnikova and colleagues, MRI was found to correctly identify partial submucosal invasion and suitability for local excision in 89% of the cases.10

Therefore, both EUS and MRI are useful, more so than CT, in assessment of the depth of tumor invasion, nodal staging, and predicting the circumferential resection margin. The use of EUS, however, does not preclude the use of MRI, or vice versa. Rather, the 2 modalities can complement each other in staging and proper patient selection for treatment.11

Despite data supporting the value of EUS in staging RC, its use is limited by a high degree of operator dependence and a substantial learning curve,12-17 which may explain the low EUS accuracy observed in some reports.7,13,15 Given the presence of recognized alternatives such as MRI, we decided to reevaluate EUS accuracy for the staging of RC outside high-volume specialized centers and prospective clinical trials.

 

 

Methods

A retrospective chart review was performed that included all consecutive patients undergoing rectal ultrasound from January 2011 to August 2015 at the US Department of Veterans Affairs Medical Center (VAMC) in Memphis, Tennessee. Sixty-five patients with short-stocked or sessile lesions < 15 cm from anal margin staged T2N0M0 or lower by endorectal ultrasound (ERUS) were included. The patients with neoplasms staged in excess of T2 or N0 were excluded from the study because treatment protocol dictates immediate neoadjuvant treatment, the administration of which would affect subsequent histopathology.

For the 37 patients included in the final analysis, ERUS results were compared with surgical pathology to ascertain accuracy. The resections were performed endoscopically or surgically with a goal of obtaining clear margins. The choice of procedure depended on size, shape, location, and depth of invasion. All patients underwent clinical and endoscopic surveillance with flexible sigmoidoscopy/EUS every 3 to 6 months for the first 2 years. We used 2 different gold standards for surveillance depending on the type of procedure performed to remove the lesion. A pathology report was the gold standard used for patients who underwent surgery. In patients who underwent endoscopic resection, we used the lack of recurrent disease, determined by normal endoscopic and endoscopic ultrasound examination, to signify complete endoscopic resection and therefore adequate staging as an early neoplasm.

Results

From January 2011 to August 2015, 65 rectal ultrasounds were performed. All EUS procedures were performed by 1 physician (C Ruben Tombazzi). All patients had previous endoscopic evaluation and tissue diagnoses. Twenty-eight patients were excluded: 18 had T3 or N1 disease, 2 had T2N0 but refused surgery, 2 had anal cancer, 3 patients with suspected cancer had benign nonneoplastic disease (2 radiation proctitis, 1 normal rectal wall), and 3 underwent EUS for benign tumors (1 ganglioneuroma and 2 lipomas).

Thirty-seven patients were included in the study, 3 of whom were staged as T2N0 and 34 as T1N0 or lower by EUS. All patients were men ranging in age from 43 to 73 years (mean, 59 years). All 37 patients underwent endoscopic or surgical resection of their early rectal neoplasm. The final pathologic evaluation of the specimens demonstrated 14 carcinoid tumors, 11 adenocarcinomas, 6 tubular adenomas with high-grade dysplasia, and 6 benign adenomas. The preoperative EUS staging was confirmed for all patients, with 100% sensitivity, specificity, and accuracy. None of the patients who underwent endoscopic or surgical transanal resection had recurrence, determined by normal endoscopic and endoscopic ultrasound appearance, during a mean of 32.6 months surveillance.

Discussion

EUS has long been a recognized method for T and N staging of RC.1,3-5,7,8 Our data confirm that, in experienced hands, EUS is highly accurate in the staging of early rectal cancers.

The impact of EUS on the management of RC was demonstrated in a Mayo Clinic prospective blinded study.1 In that cohort of 80 consecutive patients who had previously had a CT for staging, EUS altered patient management in about 30% of cases. The most common change precipatated by EUS was the indication for additional neoadjuvant treatment.

However, the results have not been as encouraging when ERUS is performed outside of strict research protocol. A multicenter, prospective, country-wide quality assurance study from > 300 German hospitals was designed to assess the diagnostic accuracy of EUS in RC.13 Of 29206 patients, 7096 underwent surgery, without neoadjuvant treatment, and were included in the final analysis. The correspondence of tumor invasion with histopathology was 64.7%, with understaging of 18% and overstaging of 17.3%.13 These numbers were better in hospitals with greater experience performing ERUS: 73% accuracy in the centers with a case load of > 30 cases per year compared with 63.2% accuracy for the centers with < 10 cases a year. Marusch and colleagues had previously demonstrated an EUS accuracy of 63.3% in a study of 1463 patients with RC in Germany.14 Another study based out of the UK had similar findings. Ashraf and colleagues performed a database analyses from 20 UK centers and identified 165 patients with RC who underwent ERUS and endoscopic microsurgery.15 Compared with histopathology, EUS had 57.1% sensitivity, 73% specificity, and 42.9% accuracy for T1 cancers; EUS accuracy was 50% for T2 and 58% for T3 tumors. The authors concluded that the general accuracy of EUS in determining stage was around 50%, the statistical equivalent of flipping a coin.

The low accuracy of EUS observed by German and British multicenter studies13-15 was attributed to the difference that may exist in clinical trials at specialized centers compared with wider use of EUS in a community setting. As seen by our data, the Memphis VAMC is not a high-volume center for the treatment of RC. However, all our EUS procedures were performed and interpreted by a single operator (C. Ruben Tombazzi) with 18 years of EUS experience. We cannot conclude that no patient was overstaged, as patients receiving a stage of T3N0 or T > N0 received neoadjuvant treatment and were not included. However, we can conclude that no patient was understaged. All patients deemed to be T1 to T2N0 included in our study received accurate staging. Our results are consistent with the high accuracy of EUS reported from other centers with experience in diagnosis and treatment of RC.1,3-5,17,18

Although EUS is accurate in differentiating T1 from T2 tumors, it cannot reliably differentiate T1 from T0 lesions. In one study, 57.6% of adenomas and 30.7% of carcinomas in situ were staged as T1 on EUS, while almost half of T1 cancers were interpreted as T0.17 This drawback is a well-known limitation of EUS; although, the misinterpretation does not affect treatment, as both T0 and T1 lesions can be treated successfully by local excision alone, which was the algorithm used for our patients. The choice of the specific procedure for local excision was left to the clinicians and included transanal endoscopic or surgical resections. At a mean follow-up of 32.6 months, none of the 37 patients who underwent endoscopic or surgical transanal resection had evidence of recurrent disease.

A limitation of EUS, or any other imaging modality, is differentiating tumor invasion from peritumoral inflammation. The inflammation can render images of tumor borders ill-defined and irregular, which hinders precise staging. However, the accurate identification of tumors with deep involvement of the submucosa (T1sm3) is of importance, because these tumors are more advanced than the superficial and intermediate T1 lesions (T1sm1 and T1sm2, respectively).

Patients with RC whose lesions are considered T1sm3 are at higher risk of harboring lymph node metastases.18 Nascimbeni and colleagues had shown that the invasion into the lower third of the submucosa (sm3) was an independent risk factor for lower cancer-free survival among patients with T1 RC.19We did not measure the distance of the tumor to muscular layer in our study, but we relied on EUS to predict the circumferential tumor margins and guide the surgical resection. Of the 11 patients with T1 rectal adenocarcinomas and the 6 patients with tubular adenoma with high-grade dysplasia, all treated by local excision, none developed a local or distant recurrence during follow-up.

Unlike rectal adenocarcinomas, the prognosis for carcinoid tumors correlates not only with the depth of invasion but also with the size of the tumor. The other adverse prognostic features include poor differentiation, high mitosis index, and lymphovascular invasion.20

EUS had been shown to be highly accurate in determining the precise carcinoid tumor size, depth of invasion, and lymph node metastases.20,21 In a study of 66 resected rectal carcinoid tumors by Ishii and colleagues, 57 lesions had a diameter of ≤ 10 mm and 9 lesions had a diameter of > 10 mm.21 All of the 57 carcinoid tumors with a diameter of ≤ 10 mm were confined to the submucosa. In contrast, 5 of the 9 lesions > 10 mm invaded the muscularis propria, 6 had a lymphovascular invasion, 4 were lymph node metastases, and 1 was a liver metastasis.

In our series, 4 of the 14 carcinoid tumors were > 10 mm but none were > 20 mm. None of the carcinoids with a diameter ≤ 10 mm invaded the muscularis propria. Of the 4 carcinoids > 10 mm, 1 was T2N0 and 3 were T1N0. All carcinoid tumors in our series were low grade and with low proliferation indexes, and all were treated successfully by local excision.

Conclusion

We believe our study shows that EUS can be highly accurate in staging rectal lesions, specifically lesions that are T1-T2N0, be they adenocarcinoma or carcinoid. Although we could not assess overstaging for lesions that were staged > T2 or > N0, we were able to determine no understaging for all of our patients. In experienced hands, EUS remains a highly accurate staging tool for early rectal carcinoma.

References

1. Harewood GC, Wiersema MJ, Nelson H, et al. A prospective, blinded assessment of the impact of preoperative staging on the management of rectal cancer. Gastroenterology. 2002;123(1):24-32.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5-29.

3. Ahuja NK, Sauer BG, Wang AY, et al. Performance of endoscopic ultrasound in staging rectal adenocarcinoma appropriate for primary surgical resection. Clin Gastroenterol Hepatol. 2015;13:339-44.

4. Doornebosch PG, Bronkhorst PJ, Hop WC, Bode WA, Sing AK, de Graaf EJ. The role of endorectal ultrasound in therapeutic decision-making for local vs. transabdominal resection of rectal tumors. Dis Colon Rectum. 2008;51(1):38-42.

5. Santoro GA, Gizzi G, Pellegrini L, Battistella G, Di Falco G. The value of high-resolution three-dimensional endorectal ultrasonography in the management of submucosal invasive rectal tumors. Dis Colon Rectum. 2009;52(11):1837-1843.

6. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: rectal cancer, version 2.2019. https://www.nccn.org/professionals/physician_gls/pdf/rectal.pdf. Published May 15, 2019. Accessed July 19, 2019.

7. Bipat S, Glas AS, Slors FJ, Zwinderman AH, Bossuyt PM, Stoker J. Rectal cancer: local staging and assessment of lymph node involvement with endoluminal US, CT, and MR imaging—a meta-analysis. Radiology. 2004;232(3):773-783.

8. Puli SR, Bechtold ML, Reddy JB, Choudhary A, Antillon MR, Brugge WR. How good is endoscopic ultrasound in differentiating various T stages of rectal cancer? Meta-analysis and systematic review. Ann Surg Oncol. 2009;16(2):254-265.

9. MERCURY Study Group. Diagnostic accuracy of preoperative magnetic resonance imaging in predicting curative resection of rectal cancer: prospective observational study. BMJ. 2006;333(7572):779.

10. Balyasnikova S, Read J, Wotherspoon A, et al. Diagnostic accuracy of high-resolution MRI as a method to predict potentially safe endoscopic and surgical planes in patient with early rectal cancer. BMJ Open Gastroenterol. 2017;4(1):e000151.

11. Frasson M, Garcia-Granero E, Roda D, et al. Preoperative chemoradiation may not always be needed for patients with T3 and T2N+ rectal cancer. Cancer. 2011;117(14):3118-3125.

12. Rafaelsen SR, Sørensen T, Jakobsen A, Bisgaard C, Lindebjerg J. Transrectal ultrasonography and magnetic resonance imaging in the staging of rectal cancer. Effect of experience. Scand J Gastroenterol. 2008;43(4):440-446.

13. Marusch F, Ptok H, Sahm M, et al. Endorectal ultrasound in rectal carcinoma – do the literature results really correspond to the realities of routine clinical care? Endoscopy. 2011;43(5):425-431.

14. Marusch F, Koch A, Schmidt U, et al. Routine use of transrectal ultrasound in rectal carcinoma: results of a prospective multicenter study. Endoscopy. 2002;34(5):385-390.

15. Ashraf S, Hompes R, Slater A, et al; Association of Coloproctology of Great Britain and Ireland Transanal Endoscopic Microsurgery (TEM) Collaboration. A critical appraisal of endorectal ultrasound and transanal endoscopic microsurgery and decision-making in early rectal cancer. Colorectal Dis. 2012;14(7):821-826.

16. Harewood GC. Assessment of clinical impact of endoscopic ultrasound on rectal cancer. Am J Gastroenterol. 2004;99(4):623-627.

17. Zorcolo L, Fantola G, Cabras F, Marongiu L, D’Alia G, Casula G. Preoperative staging of patients with rectal tumors suitable for transanal endoscopic microsurgery (TEM): comparison of endorectal ultrasound and histopathologic findings. Surg Endosc. 2009;23(6):1384-1389.

18. Akasu T, Kondo H, Moriya Y, et al. Endoscopic ultrasonography and treatment of early stage rectal cancer. World J Surg. 2000;24(9):1061-1068.

19. Nascimbeni R, Nivatvongs S, Larson DR, Burgart LJ. Long-term survival after local excision for T1 carcinoma of the rectum. Dis Colon Rectum. 2004;47(11):1773-1779.

20. Park CH, Cheon JH, Kim JO, et al. Criteria for decision making after endoscopic resection of well-differentiated rectal carcinoids with regard to potential lymphatic spread. Endoscopy. 2011;43(9):790-795.

21. Ishii N, Horiki N, Itoh T, et al. Endoscopic submucosal dissection and preoperative assessment with endoscopic ultrasonography for the treatment of rectal carcinoid tumors. Surg Endosc. 2010;24(6):1413-1419.

Article PDF
Author and Disclosure Information

Claudio Roberto Tombazzi is an Internal Medicine Resident in the Department of Medicine at Vanderbilt University in Nashville, Tennessee. Parker Loy is a Medical Student, Victor Bondar is an Assistant Professor, Bradford Waters is a Professor, and Claudio Ruben Tombazzi is an Associate Professor, all at the University of Tennessee Health Science Center in Memphis, Tennessee. Jose Ruiz is a Resident of Internal Medicine at The University of Puerto Rico in San Juan.
Correspondence: Claudio Ruben Tombazzi ([email protected])

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Issue
Federal Practitioner - 36(5)s
Publications
Topics
Page Number
S26-S29
Sections
Author and Disclosure Information

Claudio Roberto Tombazzi is an Internal Medicine Resident in the Department of Medicine at Vanderbilt University in Nashville, Tennessee. Parker Loy is a Medical Student, Victor Bondar is an Assistant Professor, Bradford Waters is a Professor, and Claudio Ruben Tombazzi is an Associate Professor, all at the University of Tennessee Health Science Center in Memphis, Tennessee. Jose Ruiz is a Resident of Internal Medicine at The University of Puerto Rico in San Juan.
Correspondence: Claudio Ruben Tombazzi ([email protected])

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Author and Disclosure Information

Claudio Roberto Tombazzi is an Internal Medicine Resident in the Department of Medicine at Vanderbilt University in Nashville, Tennessee. Parker Loy is a Medical Student, Victor Bondar is an Assistant Professor, Bradford Waters is a Professor, and Claudio Ruben Tombazzi is an Associate Professor, all at the University of Tennessee Health Science Center in Memphis, Tennessee. Jose Ruiz is a Resident of Internal Medicine at The University of Puerto Rico in San Juan.
Correspondence: Claudio Ruben Tombazzi ([email protected])

Author disclosures
The authors report no actual or potential conflicts of interest with regard to this article.

Disclaimer
The opinions expressed herein are those of the authors and do not necessarily reflect those of Federal Practitioner, Frontline Medical Communications Inc., the US Government, or any of its agencies.

Article PDF
Article PDF
Related Articles

Endoscopic ultrasound can be highly accurate for the staging of neoplasms in early rectal cancer.

Endoscopic ultrasound can be highly accurate for the staging of neoplasms in early rectal cancer.

Colorectal cancer is the second most common cause of cancer death in the US, with one-third of all colorectal cancers occurring within the rectum. Each year, an estimated 40000 Americans are diagnosed with rectal cancer (RC).1,2 The prognosis and treatment of RC depends on both T and N stage at the time of diagnosis.3-5 According to the most recent National Comprehensive Cancer Network guidelines from May 2019, patients with T1 to T2N0 tumors should undergo transanal or transabdominal surgery upfront, whereas patients with T3 to T4N0 or any TN1 to 2 should start with neoadjuvant therapy for better locoregional control, followed by surgery.6 Therefore, the appropriate management of RC requires adequate staging.

Endoscopic ultrasound (EUS), magnetic resonance imaging (MRI), and computed tomography (CT) are the imaging techniques currently used to stage RC. In a meta-analysis of 90 articles published between 1985 and 2002 that compared the 3 radiologic modalities, Bipat and colleagues found that MRI and EUS had a similar sensitivity of 94%, whereas the specificity of EUS (86%) was significantly higher than that of MRI (69%) for muscularis propria invasion.7 CT was performed only in a limited number of trials because CT was considered inadequate to assess early T stage. For perirectal tissue invasion, the sensitivity of EUS was statistically higher than that of CT and MRI imaging: 90% compared with 79% and 82%, respectively. The specificity estimates for EUS, CT, and MRI were comparable: 75%, 78%, and 76%, respectively. The respective sensitivity and specificity of the 3 imaging modalities to evaluate lymph nodes were also comparable: EUS, 67% and 78%; CT, 55% and 74%; and MRI, 66% and 76%.

The role of EUS in the diagnosis and treatment of RC has long been validated.1,2-5 A meta-analysis of 42 studies involving 5039 patients found EUS to be highly accurate for differentiating various T stages.8 However, EUS cannot assess iliac and mesenteric lymph nodes or posterior tumor extension beyond endopelvic fascia in advanced RC. Notable heterogeneity was found among the studies in the meta-analyses with regard to the type of equipment used for staging, as well as the criteria used to assess the depth of penetration and nodal status. The recent introduction of phased-array coils and the development of T2-weighted fast spin sequences have improved the resolution of MRI. The MERCURY trial showed that extension of tumor to within 1 mm of the circumferential margin on high-resolution MRI correctly predicted margin involvement at the time of surgery in 92% of the patients.9 In the retrospective study by Balyasnikova and colleagues, MRI was found to correctly identify partial submucosal invasion and suitability for local excision in 89% of the cases.10

Therefore, both EUS and MRI are useful, more so than CT, in assessment of the depth of tumor invasion, nodal staging, and predicting the circumferential resection margin. The use of EUS, however, does not preclude the use of MRI, or vice versa. Rather, the 2 modalities can complement each other in staging and proper patient selection for treatment.11

Despite data supporting the value of EUS in staging RC, its use is limited by a high degree of operator dependence and a substantial learning curve,12-17 which may explain the low EUS accuracy observed in some reports.7,13,15 Given the presence of recognized alternatives such as MRI, we decided to reevaluate EUS accuracy for the staging of RC outside high-volume specialized centers and prospective clinical trials.

 

 

Methods

A retrospective chart review was performed that included all consecutive patients undergoing rectal ultrasound from January 2011 to August 2015 at the US Department of Veterans Affairs Medical Center (VAMC) in Memphis, Tennessee. Sixty-five patients with short-stocked or sessile lesions < 15 cm from anal margin staged T2N0M0 or lower by endorectal ultrasound (ERUS) were included. The patients with neoplasms staged in excess of T2 or N0 were excluded from the study because treatment protocol dictates immediate neoadjuvant treatment, the administration of which would affect subsequent histopathology.

For the 37 patients included in the final analysis, ERUS results were compared with surgical pathology to ascertain accuracy. The resections were performed endoscopically or surgically with a goal of obtaining clear margins. The choice of procedure depended on size, shape, location, and depth of invasion. All patients underwent clinical and endoscopic surveillance with flexible sigmoidoscopy/EUS every 3 to 6 months for the first 2 years. We used 2 different gold standards for surveillance depending on the type of procedure performed to remove the lesion. A pathology report was the gold standard used for patients who underwent surgery. In patients who underwent endoscopic resection, we used the lack of recurrent disease, determined by normal endoscopic and endoscopic ultrasound examination, to signify complete endoscopic resection and therefore adequate staging as an early neoplasm.

Results

From January 2011 to August 2015, 65 rectal ultrasounds were performed. All EUS procedures were performed by 1 physician (C Ruben Tombazzi). All patients had previous endoscopic evaluation and tissue diagnoses. Twenty-eight patients were excluded: 18 had T3 or N1 disease, 2 had T2N0 but refused surgery, 2 had anal cancer, 3 patients with suspected cancer had benign nonneoplastic disease (2 radiation proctitis, 1 normal rectal wall), and 3 underwent EUS for benign tumors (1 ganglioneuroma and 2 lipomas).

Thirty-seven patients were included in the study, 3 of whom were staged as T2N0 and 34 as T1N0 or lower by EUS. All patients were men ranging in age from 43 to 73 years (mean, 59 years). All 37 patients underwent endoscopic or surgical resection of their early rectal neoplasm. The final pathologic evaluation of the specimens demonstrated 14 carcinoid tumors, 11 adenocarcinomas, 6 tubular adenomas with high-grade dysplasia, and 6 benign adenomas. The preoperative EUS staging was confirmed for all patients, with 100% sensitivity, specificity, and accuracy. None of the patients who underwent endoscopic or surgical transanal resection had recurrence, determined by normal endoscopic and endoscopic ultrasound appearance, during a mean of 32.6 months surveillance.

Discussion

EUS has long been a recognized method for T and N staging of RC.1,3-5,7,8 Our data confirm that, in experienced hands, EUS is highly accurate in the staging of early rectal cancers.

The impact of EUS on the management of RC was demonstrated in a Mayo Clinic prospective blinded study.1 In that cohort of 80 consecutive patients who had previously had a CT for staging, EUS altered patient management in about 30% of cases. The most common change precipatated by EUS was the indication for additional neoadjuvant treatment.

However, the results have not been as encouraging when ERUS is performed outside of strict research protocol. A multicenter, prospective, country-wide quality assurance study from > 300 German hospitals was designed to assess the diagnostic accuracy of EUS in RC.13 Of 29206 patients, 7096 underwent surgery, without neoadjuvant treatment, and were included in the final analysis. The correspondence of tumor invasion with histopathology was 64.7%, with understaging of 18% and overstaging of 17.3%.13 These numbers were better in hospitals with greater experience performing ERUS: 73% accuracy in the centers with a case load of > 30 cases per year compared with 63.2% accuracy for the centers with < 10 cases a year. Marusch and colleagues had previously demonstrated an EUS accuracy of 63.3% in a study of 1463 patients with RC in Germany.14 Another study based out of the UK had similar findings. Ashraf and colleagues performed a database analyses from 20 UK centers and identified 165 patients with RC who underwent ERUS and endoscopic microsurgery.15 Compared with histopathology, EUS had 57.1% sensitivity, 73% specificity, and 42.9% accuracy for T1 cancers; EUS accuracy was 50% for T2 and 58% for T3 tumors. The authors concluded that the general accuracy of EUS in determining stage was around 50%, the statistical equivalent of flipping a coin.

The low accuracy of EUS observed by German and British multicenter studies13-15 was attributed to the difference that may exist in clinical trials at specialized centers compared with wider use of EUS in a community setting. As seen by our data, the Memphis VAMC is not a high-volume center for the treatment of RC. However, all our EUS procedures were performed and interpreted by a single operator (C. Ruben Tombazzi) with 18 years of EUS experience. We cannot conclude that no patient was overstaged, as patients receiving a stage of T3N0 or T > N0 received neoadjuvant treatment and were not included. However, we can conclude that no patient was understaged. All patients deemed to be T1 to T2N0 included in our study received accurate staging. Our results are consistent with the high accuracy of EUS reported from other centers with experience in diagnosis and treatment of RC.1,3-5,17,18

Although EUS is accurate in differentiating T1 from T2 tumors, it cannot reliably differentiate T1 from T0 lesions. In one study, 57.6% of adenomas and 30.7% of carcinomas in situ were staged as T1 on EUS, while almost half of T1 cancers were interpreted as T0.17 This drawback is a well-known limitation of EUS; although, the misinterpretation does not affect treatment, as both T0 and T1 lesions can be treated successfully by local excision alone, which was the algorithm used for our patients. The choice of the specific procedure for local excision was left to the clinicians and included transanal endoscopic or surgical resections. At a mean follow-up of 32.6 months, none of the 37 patients who underwent endoscopic or surgical transanal resection had evidence of recurrent disease.

A limitation of EUS, or any other imaging modality, is differentiating tumor invasion from peritumoral inflammation. The inflammation can render images of tumor borders ill-defined and irregular, which hinders precise staging. However, the accurate identification of tumors with deep involvement of the submucosa (T1sm3) is of importance, because these tumors are more advanced than the superficial and intermediate T1 lesions (T1sm1 and T1sm2, respectively).

Patients with RC whose lesions are considered T1sm3 are at higher risk of harboring lymph node metastases.18 Nascimbeni and colleagues had shown that the invasion into the lower third of the submucosa (sm3) was an independent risk factor for lower cancer-free survival among patients with T1 RC.19We did not measure the distance of the tumor to muscular layer in our study, but we relied on EUS to predict the circumferential tumor margins and guide the surgical resection. Of the 11 patients with T1 rectal adenocarcinomas and the 6 patients with tubular adenoma with high-grade dysplasia, all treated by local excision, none developed a local or distant recurrence during follow-up.

Unlike rectal adenocarcinomas, the prognosis for carcinoid tumors correlates not only with the depth of invasion but also with the size of the tumor. The other adverse prognostic features include poor differentiation, high mitosis index, and lymphovascular invasion.20

EUS had been shown to be highly accurate in determining the precise carcinoid tumor size, depth of invasion, and lymph node metastases.20,21 In a study of 66 resected rectal carcinoid tumors by Ishii and colleagues, 57 lesions had a diameter of ≤ 10 mm and 9 lesions had a diameter of > 10 mm.21 All of the 57 carcinoid tumors with a diameter of ≤ 10 mm were confined to the submucosa. In contrast, 5 of the 9 lesions > 10 mm invaded the muscularis propria, 6 had a lymphovascular invasion, 4 were lymph node metastases, and 1 was a liver metastasis.

In our series, 4 of the 14 carcinoid tumors were > 10 mm but none were > 20 mm. None of the carcinoids with a diameter ≤ 10 mm invaded the muscularis propria. Of the 4 carcinoids > 10 mm, 1 was T2N0 and 3 were T1N0. All carcinoid tumors in our series were low grade and with low proliferation indexes, and all were treated successfully by local excision.

Conclusion

We believe our study shows that EUS can be highly accurate in staging rectal lesions, specifically lesions that are T1-T2N0, be they adenocarcinoma or carcinoid. Although we could not assess overstaging for lesions that were staged > T2 or > N0, we were able to determine no understaging for all of our patients. In experienced hands, EUS remains a highly accurate staging tool for early rectal carcinoma.

Colorectal cancer is the second most common cause of cancer death in the US, with one-third of all colorectal cancers occurring within the rectum. Each year, an estimated 40000 Americans are diagnosed with rectal cancer (RC).1,2 The prognosis and treatment of RC depends on both T and N stage at the time of diagnosis.3-5 According to the most recent National Comprehensive Cancer Network guidelines from May 2019, patients with T1 to T2N0 tumors should undergo transanal or transabdominal surgery upfront, whereas patients with T3 to T4N0 or any TN1 to 2 should start with neoadjuvant therapy for better locoregional control, followed by surgery.6 Therefore, the appropriate management of RC requires adequate staging.

Endoscopic ultrasound (EUS), magnetic resonance imaging (MRI), and computed tomography (CT) are the imaging techniques currently used to stage RC. In a meta-analysis of 90 articles published between 1985 and 2002 that compared the 3 radiologic modalities, Bipat and colleagues found that MRI and EUS had a similar sensitivity of 94%, whereas the specificity of EUS (86%) was significantly higher than that of MRI (69%) for muscularis propria invasion.7 CT was performed only in a limited number of trials because CT was considered inadequate to assess early T stage. For perirectal tissue invasion, the sensitivity of EUS was statistically higher than that of CT and MRI imaging: 90% compared with 79% and 82%, respectively. The specificity estimates for EUS, CT, and MRI were comparable: 75%, 78%, and 76%, respectively. The respective sensitivity and specificity of the 3 imaging modalities to evaluate lymph nodes were also comparable: EUS, 67% and 78%; CT, 55% and 74%; and MRI, 66% and 76%.

The role of EUS in the diagnosis and treatment of RC has long been validated.1,2-5 A meta-analysis of 42 studies involving 5039 patients found EUS to be highly accurate for differentiating various T stages.8 However, EUS cannot assess iliac and mesenteric lymph nodes or posterior tumor extension beyond endopelvic fascia in advanced RC. Notable heterogeneity was found among the studies in the meta-analyses with regard to the type of equipment used for staging, as well as the criteria used to assess the depth of penetration and nodal status. The recent introduction of phased-array coils and the development of T2-weighted fast spin sequences have improved the resolution of MRI. The MERCURY trial showed that extension of tumor to within 1 mm of the circumferential margin on high-resolution MRI correctly predicted margin involvement at the time of surgery in 92% of the patients.9 In the retrospective study by Balyasnikova and colleagues, MRI was found to correctly identify partial submucosal invasion and suitability for local excision in 89% of the cases.10

Therefore, both EUS and MRI are useful, more so than CT, in assessment of the depth of tumor invasion, nodal staging, and predicting the circumferential resection margin. The use of EUS, however, does not preclude the use of MRI, or vice versa. Rather, the 2 modalities can complement each other in staging and proper patient selection for treatment.11

Despite data supporting the value of EUS in staging RC, its use is limited by a high degree of operator dependence and a substantial learning curve,12-17 which may explain the low EUS accuracy observed in some reports.7,13,15 Given the presence of recognized alternatives such as MRI, we decided to reevaluate EUS accuracy for the staging of RC outside high-volume specialized centers and prospective clinical trials.

 

 

Methods

A retrospective chart review was performed that included all consecutive patients undergoing rectal ultrasound from January 2011 to August 2015 at the US Department of Veterans Affairs Medical Center (VAMC) in Memphis, Tennessee. Sixty-five patients with short-stocked or sessile lesions < 15 cm from anal margin staged T2N0M0 or lower by endorectal ultrasound (ERUS) were included. The patients with neoplasms staged in excess of T2 or N0 were excluded from the study because treatment protocol dictates immediate neoadjuvant treatment, the administration of which would affect subsequent histopathology.

For the 37 patients included in the final analysis, ERUS results were compared with surgical pathology to ascertain accuracy. The resections were performed endoscopically or surgically with a goal of obtaining clear margins. The choice of procedure depended on size, shape, location, and depth of invasion. All patients underwent clinical and endoscopic surveillance with flexible sigmoidoscopy/EUS every 3 to 6 months for the first 2 years. We used 2 different gold standards for surveillance depending on the type of procedure performed to remove the lesion. A pathology report was the gold standard used for patients who underwent surgery. In patients who underwent endoscopic resection, we used the lack of recurrent disease, determined by normal endoscopic and endoscopic ultrasound examination, to signify complete endoscopic resection and therefore adequate staging as an early neoplasm.

Results

From January 2011 to August 2015, 65 rectal ultrasounds were performed. All EUS procedures were performed by 1 physician (C Ruben Tombazzi). All patients had previous endoscopic evaluation and tissue diagnoses. Twenty-eight patients were excluded: 18 had T3 or N1 disease, 2 had T2N0 but refused surgery, 2 had anal cancer, 3 patients with suspected cancer had benign nonneoplastic disease (2 radiation proctitis, 1 normal rectal wall), and 3 underwent EUS for benign tumors (1 ganglioneuroma and 2 lipomas).

Thirty-seven patients were included in the study, 3 of whom were staged as T2N0 and 34 as T1N0 or lower by EUS. All patients were men ranging in age from 43 to 73 years (mean, 59 years). All 37 patients underwent endoscopic or surgical resection of their early rectal neoplasm. The final pathologic evaluation of the specimens demonstrated 14 carcinoid tumors, 11 adenocarcinomas, 6 tubular adenomas with high-grade dysplasia, and 6 benign adenomas. The preoperative EUS staging was confirmed for all patients, with 100% sensitivity, specificity, and accuracy. None of the patients who underwent endoscopic or surgical transanal resection had recurrence, determined by normal endoscopic and endoscopic ultrasound appearance, during a mean of 32.6 months surveillance.

Discussion

EUS has long been a recognized method for T and N staging of RC.1,3-5,7,8 Our data confirm that, in experienced hands, EUS is highly accurate in the staging of early rectal cancers.

The impact of EUS on the management of RC was demonstrated in a Mayo Clinic prospective blinded study.1 In that cohort of 80 consecutive patients who had previously had a CT for staging, EUS altered patient management in about 30% of cases. The most common change precipatated by EUS was the indication for additional neoadjuvant treatment.

However, the results have not been as encouraging when ERUS is performed outside of strict research protocol. A multicenter, prospective, country-wide quality assurance study from > 300 German hospitals was designed to assess the diagnostic accuracy of EUS in RC.13 Of 29206 patients, 7096 underwent surgery, without neoadjuvant treatment, and were included in the final analysis. The correspondence of tumor invasion with histopathology was 64.7%, with understaging of 18% and overstaging of 17.3%.13 These numbers were better in hospitals with greater experience performing ERUS: 73% accuracy in the centers with a case load of > 30 cases per year compared with 63.2% accuracy for the centers with < 10 cases a year. Marusch and colleagues had previously demonstrated an EUS accuracy of 63.3% in a study of 1463 patients with RC in Germany.14 Another study based out of the UK had similar findings. Ashraf and colleagues performed a database analyses from 20 UK centers and identified 165 patients with RC who underwent ERUS and endoscopic microsurgery.15 Compared with histopathology, EUS had 57.1% sensitivity, 73% specificity, and 42.9% accuracy for T1 cancers; EUS accuracy was 50% for T2 and 58% for T3 tumors. The authors concluded that the general accuracy of EUS in determining stage was around 50%, the statistical equivalent of flipping a coin.

The low accuracy of EUS observed by German and British multicenter studies13-15 was attributed to the difference that may exist in clinical trials at specialized centers compared with wider use of EUS in a community setting. As seen by our data, the Memphis VAMC is not a high-volume center for the treatment of RC. However, all our EUS procedures were performed and interpreted by a single operator (C. Ruben Tombazzi) with 18 years of EUS experience. We cannot conclude that no patient was overstaged, as patients receiving a stage of T3N0 or T > N0 received neoadjuvant treatment and were not included. However, we can conclude that no patient was understaged. All patients deemed to be T1 to T2N0 included in our study received accurate staging. Our results are consistent with the high accuracy of EUS reported from other centers with experience in diagnosis and treatment of RC.1,3-5,17,18

Although EUS is accurate in differentiating T1 from T2 tumors, it cannot reliably differentiate T1 from T0 lesions. In one study, 57.6% of adenomas and 30.7% of carcinomas in situ were staged as T1 on EUS, while almost half of T1 cancers were interpreted as T0.17 This drawback is a well-known limitation of EUS; although, the misinterpretation does not affect treatment, as both T0 and T1 lesions can be treated successfully by local excision alone, which was the algorithm used for our patients. The choice of the specific procedure for local excision was left to the clinicians and included transanal endoscopic or surgical resections. At a mean follow-up of 32.6 months, none of the 37 patients who underwent endoscopic or surgical transanal resection had evidence of recurrent disease.

A limitation of EUS, or any other imaging modality, is differentiating tumor invasion from peritumoral inflammation. The inflammation can render images of tumor borders ill-defined and irregular, which hinders precise staging. However, the accurate identification of tumors with deep involvement of the submucosa (T1sm3) is of importance, because these tumors are more advanced than the superficial and intermediate T1 lesions (T1sm1 and T1sm2, respectively).

Patients with RC whose lesions are considered T1sm3 are at higher risk of harboring lymph node metastases.18 Nascimbeni and colleagues had shown that the invasion into the lower third of the submucosa (sm3) was an independent risk factor for lower cancer-free survival among patients with T1 RC.19We did not measure the distance of the tumor to muscular layer in our study, but we relied on EUS to predict the circumferential tumor margins and guide the surgical resection. Of the 11 patients with T1 rectal adenocarcinomas and the 6 patients with tubular adenoma with high-grade dysplasia, all treated by local excision, none developed a local or distant recurrence during follow-up.

Unlike rectal adenocarcinomas, the prognosis for carcinoid tumors correlates not only with the depth of invasion but also with the size of the tumor. The other adverse prognostic features include poor differentiation, high mitosis index, and lymphovascular invasion.20

EUS had been shown to be highly accurate in determining the precise carcinoid tumor size, depth of invasion, and lymph node metastases.20,21 In a study of 66 resected rectal carcinoid tumors by Ishii and colleagues, 57 lesions had a diameter of ≤ 10 mm and 9 lesions had a diameter of > 10 mm.21 All of the 57 carcinoid tumors with a diameter of ≤ 10 mm were confined to the submucosa. In contrast, 5 of the 9 lesions > 10 mm invaded the muscularis propria, 6 had a lymphovascular invasion, 4 were lymph node metastases, and 1 was a liver metastasis.

In our series, 4 of the 14 carcinoid tumors were > 10 mm but none were > 20 mm. None of the carcinoids with a diameter ≤ 10 mm invaded the muscularis propria. Of the 4 carcinoids > 10 mm, 1 was T2N0 and 3 were T1N0. All carcinoid tumors in our series were low grade and with low proliferation indexes, and all were treated successfully by local excision.

Conclusion

We believe our study shows that EUS can be highly accurate in staging rectal lesions, specifically lesions that are T1-T2N0, be they adenocarcinoma or carcinoid. Although we could not assess overstaging for lesions that were staged > T2 or > N0, we were able to determine no understaging for all of our patients. In experienced hands, EUS remains a highly accurate staging tool for early rectal carcinoma.

References

1. Harewood GC, Wiersema MJ, Nelson H, et al. A prospective, blinded assessment of the impact of preoperative staging on the management of rectal cancer. Gastroenterology. 2002;123(1):24-32.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5-29.

3. Ahuja NK, Sauer BG, Wang AY, et al. Performance of endoscopic ultrasound in staging rectal adenocarcinoma appropriate for primary surgical resection. Clin Gastroenterol Hepatol. 2015;13:339-44.

4. Doornebosch PG, Bronkhorst PJ, Hop WC, Bode WA, Sing AK, de Graaf EJ. The role of endorectal ultrasound in therapeutic decision-making for local vs. transabdominal resection of rectal tumors. Dis Colon Rectum. 2008;51(1):38-42.

5. Santoro GA, Gizzi G, Pellegrini L, Battistella G, Di Falco G. The value of high-resolution three-dimensional endorectal ultrasonography in the management of submucosal invasive rectal tumors. Dis Colon Rectum. 2009;52(11):1837-1843.

6. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: rectal cancer, version 2.2019. https://www.nccn.org/professionals/physician_gls/pdf/rectal.pdf. Published May 15, 2019. Accessed July 19, 2019.

7. Bipat S, Glas AS, Slors FJ, Zwinderman AH, Bossuyt PM, Stoker J. Rectal cancer: local staging and assessment of lymph node involvement with endoluminal US, CT, and MR imaging—a meta-analysis. Radiology. 2004;232(3):773-783.

8. Puli SR, Bechtold ML, Reddy JB, Choudhary A, Antillon MR, Brugge WR. How good is endoscopic ultrasound in differentiating various T stages of rectal cancer? Meta-analysis and systematic review. Ann Surg Oncol. 2009;16(2):254-265.

9. MERCURY Study Group. Diagnostic accuracy of preoperative magnetic resonance imaging in predicting curative resection of rectal cancer: prospective observational study. BMJ. 2006;333(7572):779.

10. Balyasnikova S, Read J, Wotherspoon A, et al. Diagnostic accuracy of high-resolution MRI as a method to predict potentially safe endoscopic and surgical planes in patient with early rectal cancer. BMJ Open Gastroenterol. 2017;4(1):e000151.

11. Frasson M, Garcia-Granero E, Roda D, et al. Preoperative chemoradiation may not always be needed for patients with T3 and T2N+ rectal cancer. Cancer. 2011;117(14):3118-3125.

12. Rafaelsen SR, Sørensen T, Jakobsen A, Bisgaard C, Lindebjerg J. Transrectal ultrasonography and magnetic resonance imaging in the staging of rectal cancer. Effect of experience. Scand J Gastroenterol. 2008;43(4):440-446.

13. Marusch F, Ptok H, Sahm M, et al. Endorectal ultrasound in rectal carcinoma – do the literature results really correspond to the realities of routine clinical care? Endoscopy. 2011;43(5):425-431.

14. Marusch F, Koch A, Schmidt U, et al. Routine use of transrectal ultrasound in rectal carcinoma: results of a prospective multicenter study. Endoscopy. 2002;34(5):385-390.

15. Ashraf S, Hompes R, Slater A, et al; Association of Coloproctology of Great Britain and Ireland Transanal Endoscopic Microsurgery (TEM) Collaboration. A critical appraisal of endorectal ultrasound and transanal endoscopic microsurgery and decision-making in early rectal cancer. Colorectal Dis. 2012;14(7):821-826.

16. Harewood GC. Assessment of clinical impact of endoscopic ultrasound on rectal cancer. Am J Gastroenterol. 2004;99(4):623-627.

17. Zorcolo L, Fantola G, Cabras F, Marongiu L, D’Alia G, Casula G. Preoperative staging of patients with rectal tumors suitable for transanal endoscopic microsurgery (TEM): comparison of endorectal ultrasound and histopathologic findings. Surg Endosc. 2009;23(6):1384-1389.

18. Akasu T, Kondo H, Moriya Y, et al. Endoscopic ultrasonography and treatment of early stage rectal cancer. World J Surg. 2000;24(9):1061-1068.

19. Nascimbeni R, Nivatvongs S, Larson DR, Burgart LJ. Long-term survival after local excision for T1 carcinoma of the rectum. Dis Colon Rectum. 2004;47(11):1773-1779.

20. Park CH, Cheon JH, Kim JO, et al. Criteria for decision making after endoscopic resection of well-differentiated rectal carcinoids with regard to potential lymphatic spread. Endoscopy. 2011;43(9):790-795.

21. Ishii N, Horiki N, Itoh T, et al. Endoscopic submucosal dissection and preoperative assessment with endoscopic ultrasonography for the treatment of rectal carcinoid tumors. Surg Endosc. 2010;24(6):1413-1419.

References

1. Harewood GC, Wiersema MJ, Nelson H, et al. A prospective, blinded assessment of the impact of preoperative staging on the management of rectal cancer. Gastroenterology. 2002;123(1):24-32.

2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 2015;65(1):5-29.

3. Ahuja NK, Sauer BG, Wang AY, et al. Performance of endoscopic ultrasound in staging rectal adenocarcinoma appropriate for primary surgical resection. Clin Gastroenterol Hepatol. 2015;13:339-44.

4. Doornebosch PG, Bronkhorst PJ, Hop WC, Bode WA, Sing AK, de Graaf EJ. The role of endorectal ultrasound in therapeutic decision-making for local vs. transabdominal resection of rectal tumors. Dis Colon Rectum. 2008;51(1):38-42.

5. Santoro GA, Gizzi G, Pellegrini L, Battistella G, Di Falco G. The value of high-resolution three-dimensional endorectal ultrasonography in the management of submucosal invasive rectal tumors. Dis Colon Rectum. 2009;52(11):1837-1843.

6. National Comprehensive Cancer Network. NCCN clinical practice guidelines in oncology: rectal cancer, version 2.2019. https://www.nccn.org/professionals/physician_gls/pdf/rectal.pdf. Published May 15, 2019. Accessed July 19, 2019.

7. Bipat S, Glas AS, Slors FJ, Zwinderman AH, Bossuyt PM, Stoker J. Rectal cancer: local staging and assessment of lymph node involvement with endoluminal US, CT, and MR imaging—a meta-analysis. Radiology. 2004;232(3):773-783.

8. Puli SR, Bechtold ML, Reddy JB, Choudhary A, Antillon MR, Brugge WR. How good is endoscopic ultrasound in differentiating various T stages of rectal cancer? Meta-analysis and systematic review. Ann Surg Oncol. 2009;16(2):254-265.

9. MERCURY Study Group. Diagnostic accuracy of preoperative magnetic resonance imaging in predicting curative resection of rectal cancer: prospective observational study. BMJ. 2006;333(7572):779.

10. Balyasnikova S, Read J, Wotherspoon A, et al. Diagnostic accuracy of high-resolution MRI as a method to predict potentially safe endoscopic and surgical planes in patient with early rectal cancer. BMJ Open Gastroenterol. 2017;4(1):e000151.

11. Frasson M, Garcia-Granero E, Roda D, et al. Preoperative chemoradiation may not always be needed for patients with T3 and T2N+ rectal cancer. Cancer. 2011;117(14):3118-3125.

12. Rafaelsen SR, Sørensen T, Jakobsen A, Bisgaard C, Lindebjerg J. Transrectal ultrasonography and magnetic resonance imaging in the staging of rectal cancer. Effect of experience. Scand J Gastroenterol. 2008;43(4):440-446.

13. Marusch F, Ptok H, Sahm M, et al. Endorectal ultrasound in rectal carcinoma – do the literature results really correspond to the realities of routine clinical care? Endoscopy. 2011;43(5):425-431.

14. Marusch F, Koch A, Schmidt U, et al. Routine use of transrectal ultrasound in rectal carcinoma: results of a prospective multicenter study. Endoscopy. 2002;34(5):385-390.

15. Ashraf S, Hompes R, Slater A, et al; Association of Coloproctology of Great Britain and Ireland Transanal Endoscopic Microsurgery (TEM) Collaboration. A critical appraisal of endorectal ultrasound and transanal endoscopic microsurgery and decision-making in early rectal cancer. Colorectal Dis. 2012;14(7):821-826.

16. Harewood GC. Assessment of clinical impact of endoscopic ultrasound on rectal cancer. Am J Gastroenterol. 2004;99(4):623-627.

17. Zorcolo L, Fantola G, Cabras F, Marongiu L, D’Alia G, Casula G. Preoperative staging of patients with rectal tumors suitable for transanal endoscopic microsurgery (TEM): comparison of endorectal ultrasound and histopathologic findings. Surg Endosc. 2009;23(6):1384-1389.

18. Akasu T, Kondo H, Moriya Y, et al. Endoscopic ultrasonography and treatment of early stage rectal cancer. World J Surg. 2000;24(9):1061-1068.

19. Nascimbeni R, Nivatvongs S, Larson DR, Burgart LJ. Long-term survival after local excision for T1 carcinoma of the rectum. Dis Colon Rectum. 2004;47(11):1773-1779.

20. Park CH, Cheon JH, Kim JO, et al. Criteria for decision making after endoscopic resection of well-differentiated rectal carcinoids with regard to potential lymphatic spread. Endoscopy. 2011;43(9):790-795.

21. Ishii N, Horiki N, Itoh T, et al. Endoscopic submucosal dissection and preoperative assessment with endoscopic ultrasonography for the treatment of rectal carcinoid tumors. Surg Endosc. 2010;24(6):1413-1419.

Issue
Federal Practitioner - 36(5)s
Issue
Federal Practitioner - 36(5)s
Page Number
S26-S29
Page Number
S26-S29
Publications
Publications
Topics
Article Type
Display Headline
Accuracy of Endoscopic Ultrasound in Staging of Early Rectal Cancer
Display Headline
Accuracy of Endoscopic Ultrasound in Staging of Early Rectal Cancer
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Portrayal of Federal Endoscopy Technology

Article Type
Changed
Wed, 08/14/2019 - 10:02

To the Editor: I was excited to see that in the latest issue of Federal Practitioner there is an article titled “Unrelated Death After Colorectal Cancer Screening: Implications for Improving Colonoscopy Referrals.”1 In fact, it made the cover! But your cover image showed what appears to be an ancient (an ancient artifact, perhaps)—did I mention ancient?—fiber-optic endoscope. Fiber-optic endoscopes haven’t been used in maybe 20 years. High-definition endoscopy is the standard of care. Before that it was standard definition. The cover image suggests that federal endoscopists may be using museum-quality colonoscopes, which I know is not the case. I just wanted to point out what I found to be humorous.

Thank you for opportunity to share my opinion.

CDR R. Daniel Lawson, MD, MC, USN
Head, Endoscopy
Naval Medical Center San Diego
Owner, Lawson GI LLC
Gastroenterologist

Response: Dr. Lawson, thank you for your concern. The image in question was selected by myself and the art director and not the authors of the article in question, purely for its recognizable and iconic nature. The image was in no way meant to portray the current state of the technology used at federal facilities. We regret that it may have confused or misled any readers about the current standard of endoscopy care. In the future we will retire such images to the museums where they belong.

Reid A. Paul, MA
Editor

References

1. Gawron A, Bielefeldt K. Unrelated death after colorectal cancer screening: implications for improving colonoscopy referrals. Fed Pract. 2019;36(6):262-270.

Article PDF
Issue
Federal Practitioner - 36(8)a
Publications
Topics
Page Number
351
Sections
Article PDF
Article PDF
Related Articles

To the Editor: I was excited to see that in the latest issue of Federal Practitioner there is an article titled “Unrelated Death After Colorectal Cancer Screening: Implications for Improving Colonoscopy Referrals.”1 In fact, it made the cover! But your cover image showed what appears to be an ancient (an ancient artifact, perhaps)—did I mention ancient?—fiber-optic endoscope. Fiber-optic endoscopes haven’t been used in maybe 20 years. High-definition endoscopy is the standard of care. Before that it was standard definition. The cover image suggests that federal endoscopists may be using museum-quality colonoscopes, which I know is not the case. I just wanted to point out what I found to be humorous.

Thank you for opportunity to share my opinion.

CDR R. Daniel Lawson, MD, MC, USN
Head, Endoscopy
Naval Medical Center San Diego
Owner, Lawson GI LLC
Gastroenterologist

Response: Dr. Lawson, thank you for your concern. The image in question was selected by myself and the art director and not the authors of the article in question, purely for its recognizable and iconic nature. The image was in no way meant to portray the current state of the technology used at federal facilities. We regret that it may have confused or misled any readers about the current standard of endoscopy care. In the future we will retire such images to the museums where they belong.

Reid A. Paul, MA
Editor

To the Editor: I was excited to see that in the latest issue of Federal Practitioner there is an article titled “Unrelated Death After Colorectal Cancer Screening: Implications for Improving Colonoscopy Referrals.”1 In fact, it made the cover! But your cover image showed what appears to be an ancient (an ancient artifact, perhaps)—did I mention ancient?—fiber-optic endoscope. Fiber-optic endoscopes haven’t been used in maybe 20 years. High-definition endoscopy is the standard of care. Before that it was standard definition. The cover image suggests that federal endoscopists may be using museum-quality colonoscopes, which I know is not the case. I just wanted to point out what I found to be humorous.

Thank you for opportunity to share my opinion.

CDR R. Daniel Lawson, MD, MC, USN
Head, Endoscopy
Naval Medical Center San Diego
Owner, Lawson GI LLC
Gastroenterologist

Response: Dr. Lawson, thank you for your concern. The image in question was selected by myself and the art director and not the authors of the article in question, purely for its recognizable and iconic nature. The image was in no way meant to portray the current state of the technology used at federal facilities. We regret that it may have confused or misled any readers about the current standard of endoscopy care. In the future we will retire such images to the museums where they belong.

Reid A. Paul, MA
Editor

References

1. Gawron A, Bielefeldt K. Unrelated death after colorectal cancer screening: implications for improving colonoscopy referrals. Fed Pract. 2019;36(6):262-270.

References

1. Gawron A, Bielefeldt K. Unrelated death after colorectal cancer screening: implications for improving colonoscopy referrals. Fed Pract. 2019;36(6):262-270.

Issue
Federal Practitioner - 36(8)a
Issue
Federal Practitioner - 36(8)a
Page Number
351
Page Number
351
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Eyebrow Default
Letters
Use ProPublica
Hide sidebar & use full width
render the right sidebar.
Article PDF Media

Lynch syndrome screening shows low efficiency in elderly

Article Type
Changed
Wed, 05/26/2021 - 13:46

 

The need for universal screening for Lynch syndrome in elderly patients with newly diagnosed colorectal cancer (CRC) has been questioned, according to results from a retrospective cohort study.

In addition, discontinuing reflex CRC screening for Lynch syndrome in patients over age 80 years could be feasible because of very low efficiency.

“The universal strategy advocates screening all patients with newly diagnosed CRC for Lynch syndrome and has been shown to be the most sensitive method,” wrote Dan Li, MD, of Kaiser Permanente Northern California, Santa Clara, and colleagues. The findings were published in Annals of Internal Medicine.

The researchers studied 3,891 patients with newly diagnosed CRC who were screened for Lynch syndrome from 2011 to 2016. Data were collected from a population-based screening program at Kaiser Permanente Northern California.

“The system provides comprehensive medical care for more than 4 million members across 21 medical centers covering urban, suburban, and semirural areas,” Dr. Li and his colleagues wrote.

To compare universal and age-restricted screening, the team obtained surgical samples of all newly diagnosed CRC tumors and tested them for reflex mismatch repair protein expression using immunohistochemistry.

Subsequently, the age-restricted screening groups were divided into several age categories, ranging from age 50 to 85 years.

The diagnostic yield, defined as the “percentage of patients with pathogenic reflex mismatch repair gene variants among all patients with CRC screened with immunohistochemistry,” was measured and compared with the universal screening technique.

“We calculated the number of patients with CRC who needed to be screened in each age group to identify one case of Lynch syndrome by dividing the number of patients screened in each age group by the number of Lynch syndrome cases diagnosed in that group,” they explained.

After analysis, the researchers detected a total of 63 cases of Lynch syndrome (diagnostic yield, 1.62%) with universal screening, among which 5 (7.9%) were over age 70 years and 1 (1.6%) was over age 80 years.

When patients with CRC who were universally screened were used as the denominator, 58 cases (diagnostic yield, 1.49%) were detected in those with CRC diagnosed at or prior to age 70 years.

In addition, in patients diagnosed at or before age 75 and 80 years, 60 and 62 cases of Lynch syndrome (diagnostic yield, 1.54% and 1.59%) were detected, respectively.

“The incremental diagnostic yield decreased substantially after age 70 to 75 years,” they wrote.

With these findings, Dr. Li and his colleagues suggested that cessation of screening for Lynch syndrome post age 80 years may be acceptable, especially in resource-limited environments.

“Using age as the primary criterion is a simple method of selecting patients for Lynch syndrome screening in clinical practice,” they added.

In accordance with previous studies, a major reduction in Lynch syndrome incidence has been noted among elderly populations.

There remains a need for additional studies exploring the effects of diagnosing Lynch syndrome in elderly patients on family members.

The researchers acknowledged a key limitation of the study was that patients who did not finish germline analysis but were eligible for it were excluded from certain measurements. To reduce potential bias, the team conducted a sensitivity analysis, and the findings were negligible with respect to main results.

“Given the geographic variation in the reported prevalence of Lynch syndrome, the diagnostic efficiency of Lynch syndrome screening among elderly populations should be further investigated in other populations,” they concluded.

The study was funded by Kaiser Permanente Northern California. The authors reported financial affiliations with Bayer, Clinical Genomics, Covidien, Exact Sciences, Motus GI, Quorum, Universal DX, and the National Cancer Institute.

SOURCE: Li D et al. Ann Intern Med. 2019 Jun 11. doi: 10.7326/M18-3316.

Publications
Topics
Sections

 

The need for universal screening for Lynch syndrome in elderly patients with newly diagnosed colorectal cancer (CRC) has been questioned, according to results from a retrospective cohort study.

In addition, discontinuing reflex CRC screening for Lynch syndrome in patients over age 80 years could be feasible because of very low efficiency.

“The universal strategy advocates screening all patients with newly diagnosed CRC for Lynch syndrome and has been shown to be the most sensitive method,” wrote Dan Li, MD, of Kaiser Permanente Northern California, Santa Clara, and colleagues. The findings were published in Annals of Internal Medicine.

The researchers studied 3,891 patients with newly diagnosed CRC who were screened for Lynch syndrome from 2011 to 2016. Data were collected from a population-based screening program at Kaiser Permanente Northern California.

“The system provides comprehensive medical care for more than 4 million members across 21 medical centers covering urban, suburban, and semirural areas,” Dr. Li and his colleagues wrote.

To compare universal and age-restricted screening, the team obtained surgical samples of all newly diagnosed CRC tumors and tested them for reflex mismatch repair protein expression using immunohistochemistry.

Subsequently, the age-restricted screening groups were divided into several age categories, ranging from age 50 to 85 years.

The diagnostic yield, defined as the “percentage of patients with pathogenic reflex mismatch repair gene variants among all patients with CRC screened with immunohistochemistry,” was measured and compared with the universal screening technique.

“We calculated the number of patients with CRC who needed to be screened in each age group to identify one case of Lynch syndrome by dividing the number of patients screened in each age group by the number of Lynch syndrome cases diagnosed in that group,” they explained.

After analysis, the researchers detected a total of 63 cases of Lynch syndrome (diagnostic yield, 1.62%) with universal screening, among which 5 (7.9%) were over age 70 years and 1 (1.6%) was over age 80 years.

When patients with CRC who were universally screened were used as the denominator, 58 cases (diagnostic yield, 1.49%) were detected in those with CRC diagnosed at or prior to age 70 years.

In addition, in patients diagnosed at or before age 75 and 80 years, 60 and 62 cases of Lynch syndrome (diagnostic yield, 1.54% and 1.59%) were detected, respectively.

“The incremental diagnostic yield decreased substantially after age 70 to 75 years,” they wrote.

With these findings, Dr. Li and his colleagues suggested that cessation of screening for Lynch syndrome post age 80 years may be acceptable, especially in resource-limited environments.

“Using age as the primary criterion is a simple method of selecting patients for Lynch syndrome screening in clinical practice,” they added.

In accordance with previous studies, a major reduction in Lynch syndrome incidence has been noted among elderly populations.

There remains a need for additional studies exploring the effects of diagnosing Lynch syndrome in elderly patients on family members.

The researchers acknowledged a key limitation of the study was that patients who did not finish germline analysis but were eligible for it were excluded from certain measurements. To reduce potential bias, the team conducted a sensitivity analysis, and the findings were negligible with respect to main results.

“Given the geographic variation in the reported prevalence of Lynch syndrome, the diagnostic efficiency of Lynch syndrome screening among elderly populations should be further investigated in other populations,” they concluded.

The study was funded by Kaiser Permanente Northern California. The authors reported financial affiliations with Bayer, Clinical Genomics, Covidien, Exact Sciences, Motus GI, Quorum, Universal DX, and the National Cancer Institute.

SOURCE: Li D et al. Ann Intern Med. 2019 Jun 11. doi: 10.7326/M18-3316.

 

The need for universal screening for Lynch syndrome in elderly patients with newly diagnosed colorectal cancer (CRC) has been questioned, according to results from a retrospective cohort study.

In addition, discontinuing reflex CRC screening for Lynch syndrome in patients over age 80 years could be feasible because of very low efficiency.

“The universal strategy advocates screening all patients with newly diagnosed CRC for Lynch syndrome and has been shown to be the most sensitive method,” wrote Dan Li, MD, of Kaiser Permanente Northern California, Santa Clara, and colleagues. The findings were published in Annals of Internal Medicine.

The researchers studied 3,891 patients with newly diagnosed CRC who were screened for Lynch syndrome from 2011 to 2016. Data were collected from a population-based screening program at Kaiser Permanente Northern California.

“The system provides comprehensive medical care for more than 4 million members across 21 medical centers covering urban, suburban, and semirural areas,” Dr. Li and his colleagues wrote.

To compare universal and age-restricted screening, the team obtained surgical samples of all newly diagnosed CRC tumors and tested them for reflex mismatch repair protein expression using immunohistochemistry.

Subsequently, the age-restricted screening groups were divided into several age categories, ranging from age 50 to 85 years.

The diagnostic yield, defined as the “percentage of patients with pathogenic reflex mismatch repair gene variants among all patients with CRC screened with immunohistochemistry,” was measured and compared with the universal screening technique.

“We calculated the number of patients with CRC who needed to be screened in each age group to identify one case of Lynch syndrome by dividing the number of patients screened in each age group by the number of Lynch syndrome cases diagnosed in that group,” they explained.

After analysis, the researchers detected a total of 63 cases of Lynch syndrome (diagnostic yield, 1.62%) with universal screening, among which 5 (7.9%) were over age 70 years and 1 (1.6%) was over age 80 years.

When patients with CRC who were universally screened were used as the denominator, 58 cases (diagnostic yield, 1.49%) were detected in those with CRC diagnosed at or prior to age 70 years.

In addition, in patients diagnosed at or before age 75 and 80 years, 60 and 62 cases of Lynch syndrome (diagnostic yield, 1.54% and 1.59%) were detected, respectively.

“The incremental diagnostic yield decreased substantially after age 70 to 75 years,” they wrote.

With these findings, Dr. Li and his colleagues suggested that cessation of screening for Lynch syndrome post age 80 years may be acceptable, especially in resource-limited environments.

“Using age as the primary criterion is a simple method of selecting patients for Lynch syndrome screening in clinical practice,” they added.

In accordance with previous studies, a major reduction in Lynch syndrome incidence has been noted among elderly populations.

There remains a need for additional studies exploring the effects of diagnosing Lynch syndrome in elderly patients on family members.

The researchers acknowledged a key limitation of the study was that patients who did not finish germline analysis but were eligible for it were excluded from certain measurements. To reduce potential bias, the team conducted a sensitivity analysis, and the findings were negligible with respect to main results.

“Given the geographic variation in the reported prevalence of Lynch syndrome, the diagnostic efficiency of Lynch syndrome screening among elderly populations should be further investigated in other populations,” they concluded.

The study was funded by Kaiser Permanente Northern California. The authors reported financial affiliations with Bayer, Clinical Genomics, Covidien, Exact Sciences, Motus GI, Quorum, Universal DX, and the National Cancer Institute.

SOURCE: Li D et al. Ann Intern Med. 2019 Jun 11. doi: 10.7326/M18-3316.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM ANNALS OF INTERNAL MEDICINE

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Clopidogrel matches aspirin for reducing risk of colorectal cancer

Does clopidogrel reduce colorectal cancer risk?
Article Type
Changed
Wed, 05/26/2021 - 13:46

Clopidogrel appears to reduce the risk of colorectal cancer (CRC) as much as low-dose aspirin, based on a case-control study involving more than 15,000 cases.

Source: American Gastroenterological Association

Risk of CRC was reduced by 20%-30% when clopidogrel was given alone or in combination with aspirin, reported lead author Antonio Rodríguez-Miguel of Príncipe de Asturias University Hospital in Madrid and colleagues. This finding adds support to the hypothesis that low-dose aspirin is chemoprotective primarily because of its antiplatelet properties, they noted.

“The mechanism of action of low-dose aspirin to explain its protective effect is subject to debate,” the investigators wrote in Clinical Gastroenterology and Hepatology. “Although aspirin is a nonsteroidal anti-inflammatory drug (NSAID) and these drugs are known to prevent CRC through the inhibition of cyclooxygenase (COX)-2 in epithelial and stromal cells in the large bowel, at low doses (75-300 mg/d) aspirin has only transient effects on this isozyme, while permanently inactivating platelet COX-1 and suppressing thromboxane A2 production. The apparent lack of dose-dependence of the chemoprotective effect of aspirin, as well as the potential role of locally activated platelets in upregulating COX-2 expression in adjacent nucleated cells of the intestinal mucosa, have led [to] the postulation that low-dose aspirin could exert its chemoprotective effect via its antiplatelet action.”

Although previous studies have explored the chemoprotective potential of other antiplatelet agents, such as clopidogrel, the resultant body of evidence remains small. In 2017, for example, Avi Leader, MD, and colleagues reported that the chemoprotective effect of dual-antiplatelet therapy (DAPT) with clopidogrel and aspirin was superior to aspirin monotherapy, based on an additional 8% risk reduction. The present study aimed to build on such findings with evaluation of a Mediterranean cohort, which could reduce confounding lifestyle factors, owing to a lower rate of cardiovascular morbidity than other populations.

The nested, case-control study involved 15,491 cases of CRC and 60,000 controls who were randomly selected and frequency matched by sex, age, and year of indexing. Data were drawn from Base de datos para la Investigación Farmacoepidemiológica en Atención Primaria (BIFAP), a Spanish medical record database with more than 7 million patients. Records of patients involved in the present study were screened for prescription of three antiplatelet agents: low-dose aspirin, clopidogrel, and triflusal. Additional categorization identified current users, recent users, past users, and nonusers. The effects of clopidogrel and aspirin were evaluated separately, as monotherapies, and together, as DAPT.

Demographically, the mean age of the entire study population was 68.6 years, with a slight male predominance (59%). Median follow-up was similar between cases and controls, at approximately 3 years, ranging from about 1.5 to 6 years. Cases showed higher rates of gout, alcohol abuse, acute digestive diseases, and peripheral artery disease, whereas controls were more likely to have histories involving stroke, acute myocardial infarction, chronic digestive diseases, and constipation.

Controls were more likely to be current aspirin users than patients diagnosed with CRC (12.8% vs. 12.2%), giving an associated adjusted odds ratio (AOR) of 0.83. Risk reduction became statistically apparent after 180 days of aspirin usage, with an AOR of 0.79, and more prominent in the 1- to 3-year range, with an AOR of 0.73. This chemoprotective effect faded rapidly with discontinuation.

Current clopidogrel usage led to a comparable level of risk reduction, with an AOR of 0.80. It wasn’t until a year of continuous clopidogrel monotherapy that risk reduction became statistically significant, with an AOR of 0.65, which dropped to 0.57 between years 1 and 3.

Turning to a matched comparison of aspirin or clopidogrel monotherapy versus DAPT, the investigators found similar rates of chemoprotection. Current aspirin usage of any duration offered an adjusted risk reduction of 17%, compared with 25% for clopidogrel, and 29% for DAPT. Beyond 1 year of continuous and current usage, the superiority of DAPT was called into question, as clopidogrel monotherapy offered the greatest risk reduction, at 37%, compared with 22% for aspirin, and 22% for DAPT. Risk analyses involving triflusal lacked statistical significance.

“The results of the present study are compatible with a chemoprotective effect of clopidogrel against CRC, equivalent in magnitude to the one observed for low-dose aspirin,” the investigators wrote. “This finding indirectly supports the hypothesis that the chemoprotective effect of low-dose aspirin is mediated mostly through the permanent inactivation of platelet COX-1.”

The investigators pointed out that the chemoprotective effects of antiplatelet therapy begin to appear early in treatment, independently from lifestyle factors, but risk reduction depends on current usage. Although short-term usage of either aspirin or clopidogrel was associated with an increased risk of CRC, the investigators suggested that this was more likely a perceived risk rather than an actual one. “In our view, this observation could be explained in part by a detection bias, owing to an increased risk of GI bleeding induced by antiplatelet agents that could lead to a greater number of colonoscopies, and, as a result, an early cancer diagnosis,” they wrote.

The study was funded by the Fundación Instituto Teófilo Hernando. Dr. García-Rodríguez disclosed a relationship with CEIFE, which has received funding from Bayer and AstraZeneca.

SOURCE: Rodríguez-Miguel et al. Clin Gastrenterol Hepatol. 2018 Dec 20. doi: 10.1016/j.cgh.2018.12.012.

Body

The role of aspirin in reducing the risk of colorectal cancer is well established, although the mechanisms of actions are not entirely clear. One possible mechanism is through inhibition of the cyclooxygenase-1 (COX-1) pathway. The authors investigated the role of aspirin but also clopidogrel, another antiplatelet drug that works through inhibition of the COX-1 pathway in reducing the risk of CRC in a case-control study from Spain. CRC cases were randomly matched with cancer-free controls, and the use of aspirin and clopidogrel as a risk factor for CRC was studied. Not surprisingly, aspirin use was associated with reduced risk of CRC by 17%, However, what’s new is that the use of clopidogrel was associated with reduced risk of CRC by 20% also but use of dual therapy (aspirin plus clopidogrel) did not confer additional benefit. The results did not differ by patient age or sex. The caveat is that history of CRC screening or colonoscopy was not known for cases or controls, and many other confounders, such as diet, exercise, and other lifestyle and medication history that may account for the differences could not be easily teased apart. If confirmed by others, these data suggest an additional beneficial effect of antiplatelet agent clopidogrel in reducing risk of CRC, if taken for more than 1 year. The study opens the door to exploring mechanisms by which antiplatelet agents may reduce risk of CRC, and the potential role of other antiplatelet agents in reducing risk of CRC.

Aasma Shaukat, MD, MPH, GI section chief Minneapolis VAMC and professor of medicine, University of Minnesota, Minneapolis. She has no conflicts of interest. 
 

Publications
Topics
Sections
Body

The role of aspirin in reducing the risk of colorectal cancer is well established, although the mechanisms of actions are not entirely clear. One possible mechanism is through inhibition of the cyclooxygenase-1 (COX-1) pathway. The authors investigated the role of aspirin but also clopidogrel, another antiplatelet drug that works through inhibition of the COX-1 pathway in reducing the risk of CRC in a case-control study from Spain. CRC cases were randomly matched with cancer-free controls, and the use of aspirin and clopidogrel as a risk factor for CRC was studied. Not surprisingly, aspirin use was associated with reduced risk of CRC by 17%, However, what’s new is that the use of clopidogrel was associated with reduced risk of CRC by 20% also but use of dual therapy (aspirin plus clopidogrel) did not confer additional benefit. The results did not differ by patient age or sex. The caveat is that history of CRC screening or colonoscopy was not known for cases or controls, and many other confounders, such as diet, exercise, and other lifestyle and medication history that may account for the differences could not be easily teased apart. If confirmed by others, these data suggest an additional beneficial effect of antiplatelet agent clopidogrel in reducing risk of CRC, if taken for more than 1 year. The study opens the door to exploring mechanisms by which antiplatelet agents may reduce risk of CRC, and the potential role of other antiplatelet agents in reducing risk of CRC.

Aasma Shaukat, MD, MPH, GI section chief Minneapolis VAMC and professor of medicine, University of Minnesota, Minneapolis. She has no conflicts of interest. 
 

Body

The role of aspirin in reducing the risk of colorectal cancer is well established, although the mechanisms of actions are not entirely clear. One possible mechanism is through inhibition of the cyclooxygenase-1 (COX-1) pathway. The authors investigated the role of aspirin but also clopidogrel, another antiplatelet drug that works through inhibition of the COX-1 pathway in reducing the risk of CRC in a case-control study from Spain. CRC cases were randomly matched with cancer-free controls, and the use of aspirin and clopidogrel as a risk factor for CRC was studied. Not surprisingly, aspirin use was associated with reduced risk of CRC by 17%, However, what’s new is that the use of clopidogrel was associated with reduced risk of CRC by 20% also but use of dual therapy (aspirin plus clopidogrel) did not confer additional benefit. The results did not differ by patient age or sex. The caveat is that history of CRC screening or colonoscopy was not known for cases or controls, and many other confounders, such as diet, exercise, and other lifestyle and medication history that may account for the differences could not be easily teased apart. If confirmed by others, these data suggest an additional beneficial effect of antiplatelet agent clopidogrel in reducing risk of CRC, if taken for more than 1 year. The study opens the door to exploring mechanisms by which antiplatelet agents may reduce risk of CRC, and the potential role of other antiplatelet agents in reducing risk of CRC.

Aasma Shaukat, MD, MPH, GI section chief Minneapolis VAMC and professor of medicine, University of Minnesota, Minneapolis. She has no conflicts of interest. 
 

Title
Does clopidogrel reduce colorectal cancer risk?
Does clopidogrel reduce colorectal cancer risk?

Clopidogrel appears to reduce the risk of colorectal cancer (CRC) as much as low-dose aspirin, based on a case-control study involving more than 15,000 cases.

Source: American Gastroenterological Association

Risk of CRC was reduced by 20%-30% when clopidogrel was given alone or in combination with aspirin, reported lead author Antonio Rodríguez-Miguel of Príncipe de Asturias University Hospital in Madrid and colleagues. This finding adds support to the hypothesis that low-dose aspirin is chemoprotective primarily because of its antiplatelet properties, they noted.

“The mechanism of action of low-dose aspirin to explain its protective effect is subject to debate,” the investigators wrote in Clinical Gastroenterology and Hepatology. “Although aspirin is a nonsteroidal anti-inflammatory drug (NSAID) and these drugs are known to prevent CRC through the inhibition of cyclooxygenase (COX)-2 in epithelial and stromal cells in the large bowel, at low doses (75-300 mg/d) aspirin has only transient effects on this isozyme, while permanently inactivating platelet COX-1 and suppressing thromboxane A2 production. The apparent lack of dose-dependence of the chemoprotective effect of aspirin, as well as the potential role of locally activated platelets in upregulating COX-2 expression in adjacent nucleated cells of the intestinal mucosa, have led [to] the postulation that low-dose aspirin could exert its chemoprotective effect via its antiplatelet action.”

Although previous studies have explored the chemoprotective potential of other antiplatelet agents, such as clopidogrel, the resultant body of evidence remains small. In 2017, for example, Avi Leader, MD, and colleagues reported that the chemoprotective effect of dual-antiplatelet therapy (DAPT) with clopidogrel and aspirin was superior to aspirin monotherapy, based on an additional 8% risk reduction. The present study aimed to build on such findings with evaluation of a Mediterranean cohort, which could reduce confounding lifestyle factors, owing to a lower rate of cardiovascular morbidity than other populations.

The nested, case-control study involved 15,491 cases of CRC and 60,000 controls who were randomly selected and frequency matched by sex, age, and year of indexing. Data were drawn from Base de datos para la Investigación Farmacoepidemiológica en Atención Primaria (BIFAP), a Spanish medical record database with more than 7 million patients. Records of patients involved in the present study were screened for prescription of three antiplatelet agents: low-dose aspirin, clopidogrel, and triflusal. Additional categorization identified current users, recent users, past users, and nonusers. The effects of clopidogrel and aspirin were evaluated separately, as monotherapies, and together, as DAPT.

Demographically, the mean age of the entire study population was 68.6 years, with a slight male predominance (59%). Median follow-up was similar between cases and controls, at approximately 3 years, ranging from about 1.5 to 6 years. Cases showed higher rates of gout, alcohol abuse, acute digestive diseases, and peripheral artery disease, whereas controls were more likely to have histories involving stroke, acute myocardial infarction, chronic digestive diseases, and constipation.

Controls were more likely to be current aspirin users than patients diagnosed with CRC (12.8% vs. 12.2%), giving an associated adjusted odds ratio (AOR) of 0.83. Risk reduction became statistically apparent after 180 days of aspirin usage, with an AOR of 0.79, and more prominent in the 1- to 3-year range, with an AOR of 0.73. This chemoprotective effect faded rapidly with discontinuation.

Current clopidogrel usage led to a comparable level of risk reduction, with an AOR of 0.80. It wasn’t until a year of continuous clopidogrel monotherapy that risk reduction became statistically significant, with an AOR of 0.65, which dropped to 0.57 between years 1 and 3.

Turning to a matched comparison of aspirin or clopidogrel monotherapy versus DAPT, the investigators found similar rates of chemoprotection. Current aspirin usage of any duration offered an adjusted risk reduction of 17%, compared with 25% for clopidogrel, and 29% for DAPT. Beyond 1 year of continuous and current usage, the superiority of DAPT was called into question, as clopidogrel monotherapy offered the greatest risk reduction, at 37%, compared with 22% for aspirin, and 22% for DAPT. Risk analyses involving triflusal lacked statistical significance.

“The results of the present study are compatible with a chemoprotective effect of clopidogrel against CRC, equivalent in magnitude to the one observed for low-dose aspirin,” the investigators wrote. “This finding indirectly supports the hypothesis that the chemoprotective effect of low-dose aspirin is mediated mostly through the permanent inactivation of platelet COX-1.”

The investigators pointed out that the chemoprotective effects of antiplatelet therapy begin to appear early in treatment, independently from lifestyle factors, but risk reduction depends on current usage. Although short-term usage of either aspirin or clopidogrel was associated with an increased risk of CRC, the investigators suggested that this was more likely a perceived risk rather than an actual one. “In our view, this observation could be explained in part by a detection bias, owing to an increased risk of GI bleeding induced by antiplatelet agents that could lead to a greater number of colonoscopies, and, as a result, an early cancer diagnosis,” they wrote.

The study was funded by the Fundación Instituto Teófilo Hernando. Dr. García-Rodríguez disclosed a relationship with CEIFE, which has received funding from Bayer and AstraZeneca.

SOURCE: Rodríguez-Miguel et al. Clin Gastrenterol Hepatol. 2018 Dec 20. doi: 10.1016/j.cgh.2018.12.012.

Clopidogrel appears to reduce the risk of colorectal cancer (CRC) as much as low-dose aspirin, based on a case-control study involving more than 15,000 cases.

Source: American Gastroenterological Association

Risk of CRC was reduced by 20%-30% when clopidogrel was given alone or in combination with aspirin, reported lead author Antonio Rodríguez-Miguel of Príncipe de Asturias University Hospital in Madrid and colleagues. This finding adds support to the hypothesis that low-dose aspirin is chemoprotective primarily because of its antiplatelet properties, they noted.

“The mechanism of action of low-dose aspirin to explain its protective effect is subject to debate,” the investigators wrote in Clinical Gastroenterology and Hepatology. “Although aspirin is a nonsteroidal anti-inflammatory drug (NSAID) and these drugs are known to prevent CRC through the inhibition of cyclooxygenase (COX)-2 in epithelial and stromal cells in the large bowel, at low doses (75-300 mg/d) aspirin has only transient effects on this isozyme, while permanently inactivating platelet COX-1 and suppressing thromboxane A2 production. The apparent lack of dose-dependence of the chemoprotective effect of aspirin, as well as the potential role of locally activated platelets in upregulating COX-2 expression in adjacent nucleated cells of the intestinal mucosa, have led [to] the postulation that low-dose aspirin could exert its chemoprotective effect via its antiplatelet action.”

Although previous studies have explored the chemoprotective potential of other antiplatelet agents, such as clopidogrel, the resultant body of evidence remains small. In 2017, for example, Avi Leader, MD, and colleagues reported that the chemoprotective effect of dual-antiplatelet therapy (DAPT) with clopidogrel and aspirin was superior to aspirin monotherapy, based on an additional 8% risk reduction. The present study aimed to build on such findings with evaluation of a Mediterranean cohort, which could reduce confounding lifestyle factors, owing to a lower rate of cardiovascular morbidity than other populations.

The nested, case-control study involved 15,491 cases of CRC and 60,000 controls who were randomly selected and frequency matched by sex, age, and year of indexing. Data were drawn from Base de datos para la Investigación Farmacoepidemiológica en Atención Primaria (BIFAP), a Spanish medical record database with more than 7 million patients. Records of patients involved in the present study were screened for prescription of three antiplatelet agents: low-dose aspirin, clopidogrel, and triflusal. Additional categorization identified current users, recent users, past users, and nonusers. The effects of clopidogrel and aspirin were evaluated separately, as monotherapies, and together, as DAPT.

Demographically, the mean age of the entire study population was 68.6 years, with a slight male predominance (59%). Median follow-up was similar between cases and controls, at approximately 3 years, ranging from about 1.5 to 6 years. Cases showed higher rates of gout, alcohol abuse, acute digestive diseases, and peripheral artery disease, whereas controls were more likely to have histories involving stroke, acute myocardial infarction, chronic digestive diseases, and constipation.

Controls were more likely to be current aspirin users than patients diagnosed with CRC (12.8% vs. 12.2%), giving an associated adjusted odds ratio (AOR) of 0.83. Risk reduction became statistically apparent after 180 days of aspirin usage, with an AOR of 0.79, and more prominent in the 1- to 3-year range, with an AOR of 0.73. This chemoprotective effect faded rapidly with discontinuation.

Current clopidogrel usage led to a comparable level of risk reduction, with an AOR of 0.80. It wasn’t until a year of continuous clopidogrel monotherapy that risk reduction became statistically significant, with an AOR of 0.65, which dropped to 0.57 between years 1 and 3.

Turning to a matched comparison of aspirin or clopidogrel monotherapy versus DAPT, the investigators found similar rates of chemoprotection. Current aspirin usage of any duration offered an adjusted risk reduction of 17%, compared with 25% for clopidogrel, and 29% for DAPT. Beyond 1 year of continuous and current usage, the superiority of DAPT was called into question, as clopidogrel monotherapy offered the greatest risk reduction, at 37%, compared with 22% for aspirin, and 22% for DAPT. Risk analyses involving triflusal lacked statistical significance.

“The results of the present study are compatible with a chemoprotective effect of clopidogrel against CRC, equivalent in magnitude to the one observed for low-dose aspirin,” the investigators wrote. “This finding indirectly supports the hypothesis that the chemoprotective effect of low-dose aspirin is mediated mostly through the permanent inactivation of platelet COX-1.”

The investigators pointed out that the chemoprotective effects of antiplatelet therapy begin to appear early in treatment, independently from lifestyle factors, but risk reduction depends on current usage. Although short-term usage of either aspirin or clopidogrel was associated with an increased risk of CRC, the investigators suggested that this was more likely a perceived risk rather than an actual one. “In our view, this observation could be explained in part by a detection bias, owing to an increased risk of GI bleeding induced by antiplatelet agents that could lead to a greater number of colonoscopies, and, as a result, an early cancer diagnosis,” they wrote.

The study was funded by the Fundación Instituto Teófilo Hernando. Dr. García-Rodríguez disclosed a relationship with CEIFE, which has received funding from Bayer and AstraZeneca.

SOURCE: Rodríguez-Miguel et al. Clin Gastrenterol Hepatol. 2018 Dec 20. doi: 10.1016/j.cgh.2018.12.012.

Publications
Publications
Topics
Article Type
Click for Credit Status
Ready
Sections
Article Source

FROM CLINICAL GASTROENTEROLOGY AND HEPATOLOGY

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Vitals

 

Key clinical point: Clopidogrel usage appears to reduce the risk of colorectal cancer as much as low-dose aspirin.

Major finding: Current clopidogrel usage was associated with a 20% reduced risk of colorectal cancer (adjusted odds ratio, 0.8).

Study details: A nested case-control study involving 15,491 cases of colorectal cancer and 60,000 controls.

Disclosures: The study was funded by the Fundación Instituto Teófilo Hernando. Dr. García-Rodríguez disclosed a relationship with CEIFE, which has received funding from Bayer and AstraZeneca.

Source: Rodríguez-Miguel A et al. Clin Gastrenterol Hepatol. 2018 Dec 20. doi: 10.1016/j.cgh.2018.12.012.

Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.

Colorectal cancer diagnoses still moving up in younger adults, with no sign of plateau

Benefit of lower screening age ‘unknown’ without further study
Article Type
Changed
Wed, 05/26/2021 - 13:46

 

Colorectal cancer (CRC) incidence continues to rise in younger adults, with no signs of plateauing, according to investigators who recently conducted an analysis of a large US cancer registry.

Adults aged 50 years and younger accounted for about 12% of colorectal cancer diagnoses in 2015, up from 10% in 2004, and significantly more of those younger patients had advanced disease at diagnosis as compared to older adults, according to the analysis of the National Cancer Database (NCDB) by Boone Goodgame, MD, of the University of Texas at Austin, and colleagues.

“These results may provide support for adjusting CRC screening guidelines to identify patients before the age of 50 years,” said Dr. Goodgame and study coauthors in Cancer.

Only 5.8% of colorectal cancer cases were diagnosed in individuals younger than 45 years, suggesting that age may be an “appropriate target” for the screening age, the authors said in their report, alluding to the 2018 qualified recommendation from the American Cancer Society to begin screening at age 45.

However, a member of the U.S. Preventive Services Task Force – which continues to recommend screening of asymptomatic adults starting at 50 years – said in an editorial that it remains “unknown” whether the harms of screening for sporadic cases of colorectal cancer in younger individuals would outweigh the benefits.

The study by Dr. Goodgame and colleagues included a total of 1,185,763 colorectal cancer cases in the NCDB during 2004-2015, of which 89% were diagnosed at the age of 50 or older, and 11% were diagnosed in younger individuals.

The proportion of colorectal cancer cases diagnosed in people aged 50 years and younger increased from 10.0% to 12.2% during the study period (P less than .0001), with comparable increases for both rectal and colon primary tumors, according to the the journal article.

Younger patients were more likely to have stage III and stage IV disease than older patients were, according to the investigators. Stage III disease was reported for 28.1% and 23.1% of younger individuals and those over 50 years, respectively, while stage IV disease was reported for 23.5% and 16.9% (P less than .0001 for both comparisons).

Race and sex differences were seen in proportions of patients younger than 50 years with colorectal cancer, further analysis of the NCDB data showed.

Among men, only non-Hispanic whites had a significant increase in colorectal cancer diagnoses under the age of 50 years over the study period, while in women, significant increases were seen in Hispanic and non-Hispanic whites, according to the report.

It’s unclear exactly what’s behind the increase in colorectal cancer diagnosis, the authors acknowledged in their report, citing a host of potentially explanatory factors, such as access to health care, lifestyle factors such as obesity, or increased antibiotic use.

Some say the increase could simply be from the more liberal use of colonoscopy, resulting in a lead time bias, the authors noted.

“However, a change in the lead-time bias should also increase the proportion of earlier stage disease in younger adults, and we did not see this in our study,” they said in the report. “Therefore, increasing colonoscopy use does not appear to be a sufficient explanation for this association.”

In any case, more studies are needed to better determine the risks, benefits, and costs of screening individuals younger than 50 years for colorectal cancer, they concluded, saying that their data should be included in an “ongoing discussion” of screening guidelines.

Dr. Goodgame and coauthors made no conflict of interest disclosures related to the study, which was supported by the National Cancer Institute and the Cancer Prevention and Research Institute of Texas.

SOURCE: Virostko J et al. Cancer. 2019 Jul 22. doi: 10.1002/cncr.32347.

Body

 

This study of the U.S. National Cancer Data Base by Virostko et al. shows that the proportion of colorectal cancer cases diagnosed before the age of 50 years continues to increase. According to investigators, those findings may provide support for adjusting screening guidelines downward.

While those findings are “provocative,” colorectal cancer remains a very rare condition in younger individuals, in whom heritable risks play a larger role than in older individuals, according to Chyke A. Doubeni, MD, MPH, a member of the U.S. Preventive Services Task Force.

“Because the number of colorectal cancer cases from inherited causes is much higher in younger individuals, it is unknown whether screening for sporadic cases in a group with such low disease rates would result in a favorable balance of harms and benefits,” he wrote in an editorial accompanying the report.

To determine whether changing the screening age for individuals not at risk is the most appropriate public health response, multiple hypotheses need to be tested as to why colorectal cancer incidence is increasing in younger people, according to Dr. Doubeni.

“A core principle of population-based screening is to do no more harm around the time of screening than the potential future health benefits,” he said.

While evidence is needed to show that routine screening is as effective in preventing colorectal cancer deaths in younger individuals as it is in older individuals, it’s unlikely that empirical studies could be designed to adequately assess that endpoint, according to Dr. Doubeni.

“At a population level, a decrease in the average age at diagnosis or in the proportion of colorectal cancers in people 50 years and older and a shift to an earlier stage at diagnosis may all suggest that current preventive interventions are effective,” he said in the editorial.
 

Dr. Doubeni is a member of the U.S. Preventive Services Task Force. He reported no specific funding support related to his editorial, which appears in the journal Cancer .

Publications
Topics
Sections
Body

 

This study of the U.S. National Cancer Data Base by Virostko et al. shows that the proportion of colorectal cancer cases diagnosed before the age of 50 years continues to increase. According to investigators, those findings may provide support for adjusting screening guidelines downward.

While those findings are “provocative,” colorectal cancer remains a very rare condition in younger individuals, in whom heritable risks play a larger role than in older individuals, according to Chyke A. Doubeni, MD, MPH, a member of the U.S. Preventive Services Task Force.

“Because the number of colorectal cancer cases from inherited causes is much higher in younger individuals, it is unknown whether screening for sporadic cases in a group with such low disease rates would result in a favorable balance of harms and benefits,” he wrote in an editorial accompanying the report.

To determine whether changing the screening age for individuals not at risk is the most appropriate public health response, multiple hypotheses need to be tested as to why colorectal cancer incidence is increasing in younger people, according to Dr. Doubeni.

“A core principle of population-based screening is to do no more harm around the time of screening than the potential future health benefits,” he said.

While evidence is needed to show that routine screening is as effective in preventing colorectal cancer deaths in younger individuals as it is in older individuals, it’s unlikely that empirical studies could be designed to adequately assess that endpoint, according to Dr. Doubeni.

“At a population level, a decrease in the average age at diagnosis or in the proportion of colorectal cancers in people 50 years and older and a shift to an earlier stage at diagnosis may all suggest that current preventive interventions are effective,” he said in the editorial.
 

Dr. Doubeni is a member of the U.S. Preventive Services Task Force. He reported no specific funding support related to his editorial, which appears in the journal Cancer .

Body

 

This study of the U.S. National Cancer Data Base by Virostko et al. shows that the proportion of colorectal cancer cases diagnosed before the age of 50 years continues to increase. According to investigators, those findings may provide support for adjusting screening guidelines downward.

While those findings are “provocative,” colorectal cancer remains a very rare condition in younger individuals, in whom heritable risks play a larger role than in older individuals, according to Chyke A. Doubeni, MD, MPH, a member of the U.S. Preventive Services Task Force.

“Because the number of colorectal cancer cases from inherited causes is much higher in younger individuals, it is unknown whether screening for sporadic cases in a group with such low disease rates would result in a favorable balance of harms and benefits,” he wrote in an editorial accompanying the report.

To determine whether changing the screening age for individuals not at risk is the most appropriate public health response, multiple hypotheses need to be tested as to why colorectal cancer incidence is increasing in younger people, according to Dr. Doubeni.

“A core principle of population-based screening is to do no more harm around the time of screening than the potential future health benefits,” he said.

While evidence is needed to show that routine screening is as effective in preventing colorectal cancer deaths in younger individuals as it is in older individuals, it’s unlikely that empirical studies could be designed to adequately assess that endpoint, according to Dr. Doubeni.

“At a population level, a decrease in the average age at diagnosis or in the proportion of colorectal cancers in people 50 years and older and a shift to an earlier stage at diagnosis may all suggest that current preventive interventions are effective,” he said in the editorial.
 

Dr. Doubeni is a member of the U.S. Preventive Services Task Force. He reported no specific funding support related to his editorial, which appears in the journal Cancer .

Title
Benefit of lower screening age ‘unknown’ without further study
Benefit of lower screening age ‘unknown’ without further study

 

Colorectal cancer (CRC) incidence continues to rise in younger adults, with no signs of plateauing, according to investigators who recently conducted an analysis of a large US cancer registry.

Adults aged 50 years and younger accounted for about 12% of colorectal cancer diagnoses in 2015, up from 10% in 2004, and significantly more of those younger patients had advanced disease at diagnosis as compared to older adults, according to the analysis of the National Cancer Database (NCDB) by Boone Goodgame, MD, of the University of Texas at Austin, and colleagues.

“These results may provide support for adjusting CRC screening guidelines to identify patients before the age of 50 years,” said Dr. Goodgame and study coauthors in Cancer.

Only 5.8% of colorectal cancer cases were diagnosed in individuals younger than 45 years, suggesting that age may be an “appropriate target” for the screening age, the authors said in their report, alluding to the 2018 qualified recommendation from the American Cancer Society to begin screening at age 45.

However, a member of the U.S. Preventive Services Task Force – which continues to recommend screening of asymptomatic adults starting at 50 years – said in an editorial that it remains “unknown” whether the harms of screening for sporadic cases of colorectal cancer in younger individuals would outweigh the benefits.

The study by Dr. Goodgame and colleagues included a total of 1,185,763 colorectal cancer cases in the NCDB during 2004-2015, of which 89% were diagnosed at the age of 50 or older, and 11% were diagnosed in younger individuals.

The proportion of colorectal cancer cases diagnosed in people aged 50 years and younger increased from 10.0% to 12.2% during the study period (P less than .0001), with comparable increases for both rectal and colon primary tumors, according to the the journal article.

Younger patients were more likely to have stage III and stage IV disease than older patients were, according to the investigators. Stage III disease was reported for 28.1% and 23.1% of younger individuals and those over 50 years, respectively, while stage IV disease was reported for 23.5% and 16.9% (P less than .0001 for both comparisons).

Race and sex differences were seen in proportions of patients younger than 50 years with colorectal cancer, further analysis of the NCDB data showed.

Among men, only non-Hispanic whites had a significant increase in colorectal cancer diagnoses under the age of 50 years over the study period, while in women, significant increases were seen in Hispanic and non-Hispanic whites, according to the report.

It’s unclear exactly what’s behind the increase in colorectal cancer diagnosis, the authors acknowledged in their report, citing a host of potentially explanatory factors, such as access to health care, lifestyle factors such as obesity, or increased antibiotic use.

Some say the increase could simply be from the more liberal use of colonoscopy, resulting in a lead time bias, the authors noted.

“However, a change in the lead-time bias should also increase the proportion of earlier stage disease in younger adults, and we did not see this in our study,” they said in the report. “Therefore, increasing colonoscopy use does not appear to be a sufficient explanation for this association.”

In any case, more studies are needed to better determine the risks, benefits, and costs of screening individuals younger than 50 years for colorectal cancer, they concluded, saying that their data should be included in an “ongoing discussion” of screening guidelines.

Dr. Goodgame and coauthors made no conflict of interest disclosures related to the study, which was supported by the National Cancer Institute and the Cancer Prevention and Research Institute of Texas.

SOURCE: Virostko J et al. Cancer. 2019 Jul 22. doi: 10.1002/cncr.32347.

 

Colorectal cancer (CRC) incidence continues to rise in younger adults, with no signs of plateauing, according to investigators who recently conducted an analysis of a large US cancer registry.

Adults aged 50 years and younger accounted for about 12% of colorectal cancer diagnoses in 2015, up from 10% in 2004, and significantly more of those younger patients had advanced disease at diagnosis as compared to older adults, according to the analysis of the National Cancer Database (NCDB) by Boone Goodgame, MD, of the University of Texas at Austin, and colleagues.

“These results may provide support for adjusting CRC screening guidelines to identify patients before the age of 50 years,” said Dr. Goodgame and study coauthors in Cancer.

Only 5.8% of colorectal cancer cases were diagnosed in individuals younger than 45 years, suggesting that age may be an “appropriate target” for the screening age, the authors said in their report, alluding to the 2018 qualified recommendation from the American Cancer Society to begin screening at age 45.

However, a member of the U.S. Preventive Services Task Force – which continues to recommend screening of asymptomatic adults starting at 50 years – said in an editorial that it remains “unknown” whether the harms of screening for sporadic cases of colorectal cancer in younger individuals would outweigh the benefits.

The study by Dr. Goodgame and colleagues included a total of 1,185,763 colorectal cancer cases in the NCDB during 2004-2015, of which 89% were diagnosed at the age of 50 or older, and 11% were diagnosed in younger individuals.

The proportion of colorectal cancer cases diagnosed in people aged 50 years and younger increased from 10.0% to 12.2% during the study period (P less than .0001), with comparable increases for both rectal and colon primary tumors, according to the the journal article.

Younger patients were more likely to have stage III and stage IV disease than older patients were, according to the investigators. Stage III disease was reported for 28.1% and 23.1% of younger individuals and those over 50 years, respectively, while stage IV disease was reported for 23.5% and 16.9% (P less than .0001 for both comparisons).

Race and sex differences were seen in proportions of patients younger than 50 years with colorectal cancer, further analysis of the NCDB data showed.

Among men, only non-Hispanic whites had a significant increase in colorectal cancer diagnoses under the age of 50 years over the study period, while in women, significant increases were seen in Hispanic and non-Hispanic whites, according to the report.

It’s unclear exactly what’s behind the increase in colorectal cancer diagnosis, the authors acknowledged in their report, citing a host of potentially explanatory factors, such as access to health care, lifestyle factors such as obesity, or increased antibiotic use.

Some say the increase could simply be from the more liberal use of colonoscopy, resulting in a lead time bias, the authors noted.

“However, a change in the lead-time bias should also increase the proportion of earlier stage disease in younger adults, and we did not see this in our study,” they said in the report. “Therefore, increasing colonoscopy use does not appear to be a sufficient explanation for this association.”

In any case, more studies are needed to better determine the risks, benefits, and costs of screening individuals younger than 50 years for colorectal cancer, they concluded, saying that their data should be included in an “ongoing discussion” of screening guidelines.

Dr. Goodgame and coauthors made no conflict of interest disclosures related to the study, which was supported by the National Cancer Institute and the Cancer Prevention and Research Institute of Texas.

SOURCE: Virostko J et al. Cancer. 2019 Jul 22. doi: 10.1002/cncr.32347.

Publications
Publications
Topics
Article Type
Click for Credit Status
Active
Sections
Article Source

FROM CANCER

Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
CME ID
205005
Vitals

 

Key clinical point: Colorectal cancer incidence continues to rise in younger adults, with no signs of plateauing.

Major finding: Adults aged 50 years and younger accounted for about 12% of colorectal cancer diagnoses in 2015, up from 10% in 2004. Significantly more younger patients had advanced disease at diagnosis, compared with older adults.

Study details: Analysis of the National Cancer Data Base including 1,185,763 colorectal cancer cases diagnosed during 2004-2015.

Disclosures: Authors made no conflict of interest disclosures related to the study, which was supported by the National Cancer Institute and the Cancer Prevention and Research Institute of Texas.

Source: Virostko J et al. Cancer. 2019 Jul 22. doi: 10.1002/cncr.32347.

Disqus Comments
Default
Use ProPublica
Hide sidebar & use full width
render the right sidebar.