European Trial Upholds Use of Erlotinib in EGFR-Mutant Lung Cancer

Article Type
Changed
Fri, 01/04/2019 - 11:42
Display Headline
European Trial Upholds Use of Erlotinib in EGFR-Mutant Lung Cancer

CHICAGO – Data from the prospective, phase-III EURTAC trial cement the need for personalized treatment of lung cancer patients but also leave clinicians in uncharted waters in terms of treatment options.

First-line erlotinib (Tarceva) improved the primary end point of progression-free survival from 5.2 months with standard platinum-based chemotherapy to 9.4 months in white patients who had advanced non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations in an interim analysis.

Study cochair Dr. Rafael Rosell, president of the Spanish Lung Cancer Group, reported a significant 63% reduction in the risk of progression (hazard ratio, 0.37; log-rank P less than .0001) in an updated analysis presented at the annual meeting of the American Society of Clinical Oncology.

Based on positive results in the earlier interim analysis, Genentech and partner OSI Pharmaceuticals announced in January that the trial had been halted and they were set to pursue a broader indication for erlotinib as first-line treatment in NSCLC with EGFR mutations.

Erlotinib, a tyrosine kinase inhibitor (TKI), is approved in the United States and Europe as a maintenance and second-line treatment for advanced or metastatic NSCLC with and without EGFR activating mutations. Genentech’s parent company, Roche, submitted a bid to the European Medicines Agency in June 2010 to expand the drug’s label.

Even though the proverbial cat had already been let out of the bag by the drug makers, EURTAC caused a stir at ASCO, where the full data were formally presented and the study was chosen as one of the Best of ASCO 2011.

Invited discussant Dr. Tony Mok of the Chinese University of Hong Kong called the data trustworthy and a true reflection of erlotinib’s efficacy in patients with EGFR mutations. He drew parallels between EURTAC and the OPTIMAL trial in which erlotinib proved potent among Asians with this genetically distinct form of lung cancer. EGFR mutations are present in about 10% of patients in the West and about 30% of Asians, and they are associated with an increased response to erlotinib and the TKI gefitinib (Iressa).

Dr. Mok said that there’s a good chance erlotinib will be approved as first-line therapy. The EURTAC data are on par with the IPASS trial that helped gain approval for gefitinib (Iressa) as first-line therapy for patients with EGFR mutations in more than 70 countries, except the United States, where gefitinib use is restricted and AstraZeneca has said it will not seek a new indication for the drug.

"Now we have two drugs," said Dr. Mok, principal investigator of IPASS. "What are we going to do when faced with an EGFR mutation? Is there a difference in terms of the effectiveness between the TKIs in patients with EGFR mutations? That is the million-dollar question or the billion-dollar question."

Dr. Mok pointed out that three other TKIs are in the pipeline for patients with EGFR mutations, including icotinib (Zhejang BetaPharma); afatinib (Boehringer Ingelheim), which binds EGFR and inhibits HER2; and the oral, once-daily PF-299804 (Pfizer). A poster presented at ASCO on the phase-III ICOGEN trial reported that icotinib provides similar overall efficacy and better tolerability than gefitinib in patients with NSCLC who progressed after one to two lines of chemotherapy; it also improved efficacy in a subset of EGFR-mutant patient.

The EURTAC trial randomly assigned 174 chemo-naive, stage IIIB/IV NSCLC patients with exon 19 deletions or L858R mutations to receive erlotinib 150 mg/day or platinum-based doublet chemotherapy every 3 weeks for four cycles. The doublet could include cisplatin 75 mg/m2 on day 1 plus docetaxel 75 mg/m2 on day 1; cisplatin 75 mg/m2 on day 1 plus gemcitabine 1,250 mg/m2 on days 1 and 8; carboplatin area under the curve (AUC) 6 on day 1 plus docetaxel 75 mg/m2 on day 1 or carboplatin AUC 5 on day 1 plus gemcitabine 1,000 mg/m2 on days 1 and 8.

The objective response rate was 58% for erlotinib vs. 15% for chemotherapy in the updated analysis, said Dr. Rosell, head of medical oncology at the Catalan Institute of Oncology in Barcelona. At the time of the interim analysis, two patients had a complete response to erlotinib and 40 had partial responses, with 8 additional partial responses reported in the updated analysis. No patient had a complete response with chemotherapy, eight patients had partial responses early on, and five more reported partial responses in the updated analysis.

The disease control rate in the interim analysis was 79% for erlotinib vs. 66% in the updated analysis.

Median overall survival was 18.8 months with chemotherapy and 22.9 months in the interim analysis (hazard ratio, 0.80; log rank P = 0.42). As of the Jan. 26, 2011 cutoff date for the updated analysis, 94 patients remain in overall survival follow-up, with a high level of known crossover, Dr. Rosell said. A subgroup analysis suggested that progression-free survival was better in patients with a performance status of 0, never-smokers, and those with an exon 19 deletion.

 

 

The majority of patients who relapsed on erlotinib were switched to chemotherapy. The tolerability of erlotinib was consistent with previous studies, he noted.

The Spanish Lung Cancer Group sponsored the trial. Dr. Rosell disclosed a consultant/advisory role with Roche. Two of his coauthors reported a similar role, with one also providing expert testimony for Roche. Dr. Mok disclosed relationships with several drug companies, including AstraZeneca, Roche, Boehringer Ingelheim, and Pfizer.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Legacy Keywords
EURTAC trial, lung cancer, First-line erlotinib, Tarceva, platinum-based chemotherapy, white patients, advanced non-small cell lung cancer, NSCLC, epidermal growth factor receptor mutations, EGFR, Dr. Rafael Rosell, Spanish Lung Cancer Group, American Society of Clinical Oncology.

Sections
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Data from the prospective, phase-III EURTAC trial cement the need for personalized treatment of lung cancer patients but also leave clinicians in uncharted waters in terms of treatment options.

First-line erlotinib (Tarceva) improved the primary end point of progression-free survival from 5.2 months with standard platinum-based chemotherapy to 9.4 months in white patients who had advanced non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations in an interim analysis.

Study cochair Dr. Rafael Rosell, president of the Spanish Lung Cancer Group, reported a significant 63% reduction in the risk of progression (hazard ratio, 0.37; log-rank P less than .0001) in an updated analysis presented at the annual meeting of the American Society of Clinical Oncology.

Based on positive results in the earlier interim analysis, Genentech and partner OSI Pharmaceuticals announced in January that the trial had been halted and they were set to pursue a broader indication for erlotinib as first-line treatment in NSCLC with EGFR mutations.

Erlotinib, a tyrosine kinase inhibitor (TKI), is approved in the United States and Europe as a maintenance and second-line treatment for advanced or metastatic NSCLC with and without EGFR activating mutations. Genentech’s parent company, Roche, submitted a bid to the European Medicines Agency in June 2010 to expand the drug’s label.

Even though the proverbial cat had already been let out of the bag by the drug makers, EURTAC caused a stir at ASCO, where the full data were formally presented and the study was chosen as one of the Best of ASCO 2011.

Invited discussant Dr. Tony Mok of the Chinese University of Hong Kong called the data trustworthy and a true reflection of erlotinib’s efficacy in patients with EGFR mutations. He drew parallels between EURTAC and the OPTIMAL trial in which erlotinib proved potent among Asians with this genetically distinct form of lung cancer. EGFR mutations are present in about 10% of patients in the West and about 30% of Asians, and they are associated with an increased response to erlotinib and the TKI gefitinib (Iressa).

Dr. Mok said that there’s a good chance erlotinib will be approved as first-line therapy. The EURTAC data are on par with the IPASS trial that helped gain approval for gefitinib (Iressa) as first-line therapy for patients with EGFR mutations in more than 70 countries, except the United States, where gefitinib use is restricted and AstraZeneca has said it will not seek a new indication for the drug.

"Now we have two drugs," said Dr. Mok, principal investigator of IPASS. "What are we going to do when faced with an EGFR mutation? Is there a difference in terms of the effectiveness between the TKIs in patients with EGFR mutations? That is the million-dollar question or the billion-dollar question."

Dr. Mok pointed out that three other TKIs are in the pipeline for patients with EGFR mutations, including icotinib (Zhejang BetaPharma); afatinib (Boehringer Ingelheim), which binds EGFR and inhibits HER2; and the oral, once-daily PF-299804 (Pfizer). A poster presented at ASCO on the phase-III ICOGEN trial reported that icotinib provides similar overall efficacy and better tolerability than gefitinib in patients with NSCLC who progressed after one to two lines of chemotherapy; it also improved efficacy in a subset of EGFR-mutant patient.

The EURTAC trial randomly assigned 174 chemo-naive, stage IIIB/IV NSCLC patients with exon 19 deletions or L858R mutations to receive erlotinib 150 mg/day or platinum-based doublet chemotherapy every 3 weeks for four cycles. The doublet could include cisplatin 75 mg/m2 on day 1 plus docetaxel 75 mg/m2 on day 1; cisplatin 75 mg/m2 on day 1 plus gemcitabine 1,250 mg/m2 on days 1 and 8; carboplatin area under the curve (AUC) 6 on day 1 plus docetaxel 75 mg/m2 on day 1 or carboplatin AUC 5 on day 1 plus gemcitabine 1,000 mg/m2 on days 1 and 8.

The objective response rate was 58% for erlotinib vs. 15% for chemotherapy in the updated analysis, said Dr. Rosell, head of medical oncology at the Catalan Institute of Oncology in Barcelona. At the time of the interim analysis, two patients had a complete response to erlotinib and 40 had partial responses, with 8 additional partial responses reported in the updated analysis. No patient had a complete response with chemotherapy, eight patients had partial responses early on, and five more reported partial responses in the updated analysis.

The disease control rate in the interim analysis was 79% for erlotinib vs. 66% in the updated analysis.

Median overall survival was 18.8 months with chemotherapy and 22.9 months in the interim analysis (hazard ratio, 0.80; log rank P = 0.42). As of the Jan. 26, 2011 cutoff date for the updated analysis, 94 patients remain in overall survival follow-up, with a high level of known crossover, Dr. Rosell said. A subgroup analysis suggested that progression-free survival was better in patients with a performance status of 0, never-smokers, and those with an exon 19 deletion.

 

 

The majority of patients who relapsed on erlotinib were switched to chemotherapy. The tolerability of erlotinib was consistent with previous studies, he noted.

The Spanish Lung Cancer Group sponsored the trial. Dr. Rosell disclosed a consultant/advisory role with Roche. Two of his coauthors reported a similar role, with one also providing expert testimony for Roche. Dr. Mok disclosed relationships with several drug companies, including AstraZeneca, Roche, Boehringer Ingelheim, and Pfizer.

CHICAGO – Data from the prospective, phase-III EURTAC trial cement the need for personalized treatment of lung cancer patients but also leave clinicians in uncharted waters in terms of treatment options.

First-line erlotinib (Tarceva) improved the primary end point of progression-free survival from 5.2 months with standard platinum-based chemotherapy to 9.4 months in white patients who had advanced non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) mutations in an interim analysis.

Study cochair Dr. Rafael Rosell, president of the Spanish Lung Cancer Group, reported a significant 63% reduction in the risk of progression (hazard ratio, 0.37; log-rank P less than .0001) in an updated analysis presented at the annual meeting of the American Society of Clinical Oncology.

Based on positive results in the earlier interim analysis, Genentech and partner OSI Pharmaceuticals announced in January that the trial had been halted and they were set to pursue a broader indication for erlotinib as first-line treatment in NSCLC with EGFR mutations.

Erlotinib, a tyrosine kinase inhibitor (TKI), is approved in the United States and Europe as a maintenance and second-line treatment for advanced or metastatic NSCLC with and without EGFR activating mutations. Genentech’s parent company, Roche, submitted a bid to the European Medicines Agency in June 2010 to expand the drug’s label.

Even though the proverbial cat had already been let out of the bag by the drug makers, EURTAC caused a stir at ASCO, where the full data were formally presented and the study was chosen as one of the Best of ASCO 2011.

Invited discussant Dr. Tony Mok of the Chinese University of Hong Kong called the data trustworthy and a true reflection of erlotinib’s efficacy in patients with EGFR mutations. He drew parallels between EURTAC and the OPTIMAL trial in which erlotinib proved potent among Asians with this genetically distinct form of lung cancer. EGFR mutations are present in about 10% of patients in the West and about 30% of Asians, and they are associated with an increased response to erlotinib and the TKI gefitinib (Iressa).

Dr. Mok said that there’s a good chance erlotinib will be approved as first-line therapy. The EURTAC data are on par with the IPASS trial that helped gain approval for gefitinib (Iressa) as first-line therapy for patients with EGFR mutations in more than 70 countries, except the United States, where gefitinib use is restricted and AstraZeneca has said it will not seek a new indication for the drug.

"Now we have two drugs," said Dr. Mok, principal investigator of IPASS. "What are we going to do when faced with an EGFR mutation? Is there a difference in terms of the effectiveness between the TKIs in patients with EGFR mutations? That is the million-dollar question or the billion-dollar question."

Dr. Mok pointed out that three other TKIs are in the pipeline for patients with EGFR mutations, including icotinib (Zhejang BetaPharma); afatinib (Boehringer Ingelheim), which binds EGFR and inhibits HER2; and the oral, once-daily PF-299804 (Pfizer). A poster presented at ASCO on the phase-III ICOGEN trial reported that icotinib provides similar overall efficacy and better tolerability than gefitinib in patients with NSCLC who progressed after one to two lines of chemotherapy; it also improved efficacy in a subset of EGFR-mutant patient.

The EURTAC trial randomly assigned 174 chemo-naive, stage IIIB/IV NSCLC patients with exon 19 deletions or L858R mutations to receive erlotinib 150 mg/day or platinum-based doublet chemotherapy every 3 weeks for four cycles. The doublet could include cisplatin 75 mg/m2 on day 1 plus docetaxel 75 mg/m2 on day 1; cisplatin 75 mg/m2 on day 1 plus gemcitabine 1,250 mg/m2 on days 1 and 8; carboplatin area under the curve (AUC) 6 on day 1 plus docetaxel 75 mg/m2 on day 1 or carboplatin AUC 5 on day 1 plus gemcitabine 1,000 mg/m2 on days 1 and 8.

The objective response rate was 58% for erlotinib vs. 15% for chemotherapy in the updated analysis, said Dr. Rosell, head of medical oncology at the Catalan Institute of Oncology in Barcelona. At the time of the interim analysis, two patients had a complete response to erlotinib and 40 had partial responses, with 8 additional partial responses reported in the updated analysis. No patient had a complete response with chemotherapy, eight patients had partial responses early on, and five more reported partial responses in the updated analysis.

The disease control rate in the interim analysis was 79% for erlotinib vs. 66% in the updated analysis.

Median overall survival was 18.8 months with chemotherapy and 22.9 months in the interim analysis (hazard ratio, 0.80; log rank P = 0.42). As of the Jan. 26, 2011 cutoff date for the updated analysis, 94 patients remain in overall survival follow-up, with a high level of known crossover, Dr. Rosell said. A subgroup analysis suggested that progression-free survival was better in patients with a performance status of 0, never-smokers, and those with an exon 19 deletion.

 

 

The majority of patients who relapsed on erlotinib were switched to chemotherapy. The tolerability of erlotinib was consistent with previous studies, he noted.

The Spanish Lung Cancer Group sponsored the trial. Dr. Rosell disclosed a consultant/advisory role with Roche. Two of his coauthors reported a similar role, with one also providing expert testimony for Roche. Dr. Mok disclosed relationships with several drug companies, including AstraZeneca, Roche, Boehringer Ingelheim, and Pfizer.

Publications
Publications
Topics
Article Type
Display Headline
European Trial Upholds Use of Erlotinib in EGFR-Mutant Lung Cancer
Display Headline
European Trial Upholds Use of Erlotinib in EGFR-Mutant Lung Cancer
Legacy Keywords
EURTAC trial, lung cancer, First-line erlotinib, Tarceva, platinum-based chemotherapy, white patients, advanced non-small cell lung cancer, NSCLC, epidermal growth factor receptor mutations, EGFR, Dr. Rafael Rosell, Spanish Lung Cancer Group, American Society of Clinical Oncology.

Legacy Keywords
EURTAC trial, lung cancer, First-line erlotinib, Tarceva, platinum-based chemotherapy, white patients, advanced non-small cell lung cancer, NSCLC, epidermal growth factor receptor mutations, EGFR, Dr. Rafael Rosell, Spanish Lung Cancer Group, American Society of Clinical Oncology.

Sections
Article Source

FFROM THE ANNUAL MEETING OF THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY

PURLs Copyright

Inside the Article

Vitals

Major Finding: Erlotinib resulted in a significant 63% reduction in the risk of progression, compared with standard chemotherapy (HR 0.37).

Data Source: Phase-III, prospective randomized EURTAC trial in 174 white patients with advanced non-small cell lung cancer and EGFR mutations.

Disclosures: The Spanish Lung Cancer Group sponsored the trial. Dr. Rosell disclosed a consultant/advisory role with Roche. Two of his coauthors reported a similar role, with one also providing expert testimony for Roche. Dr. Mok disclosed relationships with several drug companies, including AstraZeneca, Roche, Boehringer Ingelheim, and Pfizer.

Accurate Biomarker Testing Key to Experimental MetMAb in Lung Cancer

Article Type
Changed
Fri, 01/04/2019 - 11:42
Display Headline
Accurate Biomarker Testing Key to Experimental MetMAb in Lung Cancer

CHICAGO – Final efficacy results from the phase II OAM4458g trial confirm that the success of MetMAb in previously treated advanced lung cancer lies in accurate biomarker testing.

While some patients gained a striking survival advantage when given the investigational monoclonal antibody as second-or third-line therapy for non–small cell lung cancer (NSCLC), the study group as a whole did not and others actually did worse. The difference appears to be driven by expression of the c-Met receptor.

    Dr. David Spigel

MetMAb targets hepatocyte growth factor and its receptor, c-Met. Expression of c-Met is associated with a worse prognosis in many cancers, including NSCLC. Met activation by hepatocyte growth factor is also thought to decrease sensitivity to erlotinib (Tarceva). Hence the interest in combining MetMAb with erlotinib. In this trial patients received either a combination of the two drugs or erlotinib with a placebo.

In NSCLC patients whose tumors were classified as Met positive, the addition of MetMAb to erlotinib nearly doubled the median time that they were free of disease from 1.5 months to 2.9 months (hazard ratio, 0.53; log rank P = .04) and tripled median overall survival from 3.8 months to 12.6 months (HR, 0.37; log rank P = .002), Dr. David Spigel said at the annual meeting of the American Society of Clinical Oncology.

When MetMAb plus erlotinib was given to patients with Met-negative tumors, however, median progression-free survival was significantly lower at 1.4 months, compared with 2.7 months in the control arm given erlotinib plus placebo (HR, 1.82; P = .05).

Median overall survival was also shorter with the combination in the Met-negative group – 8.1 months vs. 15.3 months with erlotinib and placebo – although the difference did not reach statistical significance (HR, 1.78; P = .158), said Dr Spigel, director of lung cancer research at the Sarah Cannon Research Institute in Nashville, Tenn.

Invited discussant Dr. Tony Mok, with the Chinese University of Hong Kong, said there’s no doubt that MetMAb should move into phase III evaluation, but stressed the need for accurate biomarker testing in patients to determine Met status.

"This is the key to the success of this drug," he said. "Is this biomarker valid and trustworthy?"

The Met diagnostic test used in the phase II study was developed by Ventana Medical Systems, a tissue diagnostics company owned by Roche, the parent company of the study sponsor, Genentech. Met status was assessed after randomization and prior to unblinding, with 93% of all 137 patients having adequate tissue for evaluation of c-Met by immunohistochemistry. In all, 52% of patients with evaluable tissue were "Met diagnostic positive," defined by at least 50% of tumor cells with moderate or strong staining intensity, Dr. Spigel explained.

Patients were randomized to erlotinib 150 mg daily plus MetMAb 15 mg/kg IV every 3 weeks or the same dosing of erlotinib and placebo. Coprimary end points were progression-free survival in the Met diagnostic–positive and intention-to-treat populations.

In the latter, the combination of MetMAb and erlotinib failed to significantly improve median time to progression over erlotinib (2.2 months vs. 2.6 months; HR, = 1.09; P = .69) or overall survival (8.9 months vs. 7.4 months; HR, 0.80; P = .34), he said.

The researchers performed additional analyses in key subpopulations, suggesting that the benefit from MetMAb is not related to epidermal growth factor receptor mutation or fluorescence in situ hybridization (FISH) status.

Although the patient numbers were small, an overall survival advantage was observed with MetMAb for patients with high Met expression (at least 5 copies) by FISH (HR, 0.60; P = .35), and was maintained in FISH-negative/Met diagnostic–positive patients (HR, 0.37; P = .01), Dr. Spigel said. Patients who were Met diagnostic positive and did not have an epidermal growth factor receptor mutation also gained a survival advantage (HR, 0.42; P = .01).

"Outcomes in the diagnostic subpopulations highlight the importance of developing tools to identify patients who might best benefit from this treatment," he said, adding that immunohistochemistry appears to be more sensitive than FISH in determining benefit from combination MetMAb/erlotinib.

The study confirmed that Met expression by immunohistochemistry is associated with worse outcomes. An analysis of the 68 patients treated with erlotinib plus placebo confirmed that Met expression revealed that progression-free survival was worse among Met diagnostic–positive vs. Met diagnostic–negative patients (1.5 months vs. 2.7 months; HR, 1.71; P = .06), as was overall survival (3.8 months vs. 15.3 months; HR, 2.61; P = .004).

In response to audience questions, Dr. Spigel said it is unknown whether metastatic sites have different Met expression than primary tumor sites or why outcomes are worse in low Met tumors.

 

 

Dr. Spigel observed that no new safety concerns emerged in the trial, although patients treated with MetMAb had more peripheral edema that was largely low grade, reversible, and manageable.

A phase III study testing MetMAb plus erlotinib in Met diagnostic–positive patients is expected to start enrolling this year, he said.

Dr. Spigel disclosed a consultant/advisory role with Genentech, which sponsored the study. His coauthors disclosed financial relationships with several firms including employment with Genentech.

Meeting/Event
Author and Disclosure Information

Publications
Topics
Author and Disclosure Information

Author and Disclosure Information

Meeting/Event
Meeting/Event

CHICAGO – Final efficacy results from the phase II OAM4458g trial confirm that the success of MetMAb in previously treated advanced lung cancer lies in accurate biomarker testing.

While some patients gained a striking survival advantage when given the investigational monoclonal antibody as second-or third-line therapy for non–small cell lung cancer (NSCLC), the study group as a whole did not and others actually did worse. The difference appears to be driven by expression of the c-Met receptor.

    Dr. David Spigel

MetMAb targets hepatocyte growth factor and its receptor, c-Met. Expression of c-Met is associated with a worse prognosis in many cancers, including NSCLC. Met activation by hepatocyte growth factor is also thought to decrease sensitivity to erlotinib (Tarceva). Hence the interest in combining MetMAb with erlotinib. In this trial patients received either a combination of the two drugs or erlotinib with a placebo.

In NSCLC patients whose tumors were classified as Met positive, the addition of MetMAb to erlotinib nearly doubled the median time that they were free of disease from 1.5 months to 2.9 months (hazard ratio, 0.53; log rank P = .04) and tripled median overall survival from 3.8 months to 12.6 months (HR, 0.37; log rank P = .002), Dr. David Spigel said at the annual meeting of the American Society of Clinical Oncology.

When MetMAb plus erlotinib was given to patients with Met-negative tumors, however, median progression-free survival was significantly lower at 1.4 months, compared with 2.7 months in the control arm given erlotinib plus placebo (HR, 1.82; P = .05).

Median overall survival was also shorter with the combination in the Met-negative group – 8.1 months vs. 15.3 months with erlotinib and placebo – although the difference did not reach statistical significance (HR, 1.78; P = .158), said Dr Spigel, director of lung cancer research at the Sarah Cannon Research Institute in Nashville, Tenn.

Invited discussant Dr. Tony Mok, with the Chinese University of Hong Kong, said there’s no doubt that MetMAb should move into phase III evaluation, but stressed the need for accurate biomarker testing in patients to determine Met status.

"This is the key to the success of this drug," he said. "Is this biomarker valid and trustworthy?"

The Met diagnostic test used in the phase II study was developed by Ventana Medical Systems, a tissue diagnostics company owned by Roche, the parent company of the study sponsor, Genentech. Met status was assessed after randomization and prior to unblinding, with 93% of all 137 patients having adequate tissue for evaluation of c-Met by immunohistochemistry. In all, 52% of patients with evaluable tissue were "Met diagnostic positive," defined by at least 50% of tumor cells with moderate or strong staining intensity, Dr. Spigel explained.

Patients were randomized to erlotinib 150 mg daily plus MetMAb 15 mg/kg IV every 3 weeks or the same dosing of erlotinib and placebo. Coprimary end points were progression-free survival in the Met diagnostic–positive and intention-to-treat populations.

In the latter, the combination of MetMAb and erlotinib failed to significantly improve median time to progression over erlotinib (2.2 months vs. 2.6 months; HR, = 1.09; P = .69) or overall survival (8.9 months vs. 7.4 months; HR, 0.80; P = .34), he said.

The researchers performed additional analyses in key subpopulations, suggesting that the benefit from MetMAb is not related to epidermal growth factor receptor mutation or fluorescence in situ hybridization (FISH) status.

Although the patient numbers were small, an overall survival advantage was observed with MetMAb for patients with high Met expression (at least 5 copies) by FISH (HR, 0.60; P = .35), and was maintained in FISH-negative/Met diagnostic–positive patients (HR, 0.37; P = .01), Dr. Spigel said. Patients who were Met diagnostic positive and did not have an epidermal growth factor receptor mutation also gained a survival advantage (HR, 0.42; P = .01).

"Outcomes in the diagnostic subpopulations highlight the importance of developing tools to identify patients who might best benefit from this treatment," he said, adding that immunohistochemistry appears to be more sensitive than FISH in determining benefit from combination MetMAb/erlotinib.

The study confirmed that Met expression by immunohistochemistry is associated with worse outcomes. An analysis of the 68 patients treated with erlotinib plus placebo confirmed that Met expression revealed that progression-free survival was worse among Met diagnostic–positive vs. Met diagnostic–negative patients (1.5 months vs. 2.7 months; HR, 1.71; P = .06), as was overall survival (3.8 months vs. 15.3 months; HR, 2.61; P = .004).

In response to audience questions, Dr. Spigel said it is unknown whether metastatic sites have different Met expression than primary tumor sites or why outcomes are worse in low Met tumors.

 

 

Dr. Spigel observed that no new safety concerns emerged in the trial, although patients treated with MetMAb had more peripheral edema that was largely low grade, reversible, and manageable.

A phase III study testing MetMAb plus erlotinib in Met diagnostic–positive patients is expected to start enrolling this year, he said.

Dr. Spigel disclosed a consultant/advisory role with Genentech, which sponsored the study. His coauthors disclosed financial relationships with several firms including employment with Genentech.

CHICAGO – Final efficacy results from the phase II OAM4458g trial confirm that the success of MetMAb in previously treated advanced lung cancer lies in accurate biomarker testing.

While some patients gained a striking survival advantage when given the investigational monoclonal antibody as second-or third-line therapy for non–small cell lung cancer (NSCLC), the study group as a whole did not and others actually did worse. The difference appears to be driven by expression of the c-Met receptor.

    Dr. David Spigel

MetMAb targets hepatocyte growth factor and its receptor, c-Met. Expression of c-Met is associated with a worse prognosis in many cancers, including NSCLC. Met activation by hepatocyte growth factor is also thought to decrease sensitivity to erlotinib (Tarceva). Hence the interest in combining MetMAb with erlotinib. In this trial patients received either a combination of the two drugs or erlotinib with a placebo.

In NSCLC patients whose tumors were classified as Met positive, the addition of MetMAb to erlotinib nearly doubled the median time that they were free of disease from 1.5 months to 2.9 months (hazard ratio, 0.53; log rank P = .04) and tripled median overall survival from 3.8 months to 12.6 months (HR, 0.37; log rank P = .002), Dr. David Spigel said at the annual meeting of the American Society of Clinical Oncology.

When MetMAb plus erlotinib was given to patients with Met-negative tumors, however, median progression-free survival was significantly lower at 1.4 months, compared with 2.7 months in the control arm given erlotinib plus placebo (HR, 1.82; P = .05).

Median overall survival was also shorter with the combination in the Met-negative group – 8.1 months vs. 15.3 months with erlotinib and placebo – although the difference did not reach statistical significance (HR, 1.78; P = .158), said Dr Spigel, director of lung cancer research at the Sarah Cannon Research Institute in Nashville, Tenn.

Invited discussant Dr. Tony Mok, with the Chinese University of Hong Kong, said there’s no doubt that MetMAb should move into phase III evaluation, but stressed the need for accurate biomarker testing in patients to determine Met status.

"This is the key to the success of this drug," he said. "Is this biomarker valid and trustworthy?"

The Met diagnostic test used in the phase II study was developed by Ventana Medical Systems, a tissue diagnostics company owned by Roche, the parent company of the study sponsor, Genentech. Met status was assessed after randomization and prior to unblinding, with 93% of all 137 patients having adequate tissue for evaluation of c-Met by immunohistochemistry. In all, 52% of patients with evaluable tissue were "Met diagnostic positive," defined by at least 50% of tumor cells with moderate or strong staining intensity, Dr. Spigel explained.

Patients were randomized to erlotinib 150 mg daily plus MetMAb 15 mg/kg IV every 3 weeks or the same dosing of erlotinib and placebo. Coprimary end points were progression-free survival in the Met diagnostic–positive and intention-to-treat populations.

In the latter, the combination of MetMAb and erlotinib failed to significantly improve median time to progression over erlotinib (2.2 months vs. 2.6 months; HR, = 1.09; P = .69) or overall survival (8.9 months vs. 7.4 months; HR, 0.80; P = .34), he said.

The researchers performed additional analyses in key subpopulations, suggesting that the benefit from MetMAb is not related to epidermal growth factor receptor mutation or fluorescence in situ hybridization (FISH) status.

Although the patient numbers were small, an overall survival advantage was observed with MetMAb for patients with high Met expression (at least 5 copies) by FISH (HR, 0.60; P = .35), and was maintained in FISH-negative/Met diagnostic–positive patients (HR, 0.37; P = .01), Dr. Spigel said. Patients who were Met diagnostic positive and did not have an epidermal growth factor receptor mutation also gained a survival advantage (HR, 0.42; P = .01).

"Outcomes in the diagnostic subpopulations highlight the importance of developing tools to identify patients who might best benefit from this treatment," he said, adding that immunohistochemistry appears to be more sensitive than FISH in determining benefit from combination MetMAb/erlotinib.

The study confirmed that Met expression by immunohistochemistry is associated with worse outcomes. An analysis of the 68 patients treated with erlotinib plus placebo confirmed that Met expression revealed that progression-free survival was worse among Met diagnostic–positive vs. Met diagnostic–negative patients (1.5 months vs. 2.7 months; HR, 1.71; P = .06), as was overall survival (3.8 months vs. 15.3 months; HR, 2.61; P = .004).

In response to audience questions, Dr. Spigel said it is unknown whether metastatic sites have different Met expression than primary tumor sites or why outcomes are worse in low Met tumors.

 

 

Dr. Spigel observed that no new safety concerns emerged in the trial, although patients treated with MetMAb had more peripheral edema that was largely low grade, reversible, and manageable.

A phase III study testing MetMAb plus erlotinib in Met diagnostic–positive patients is expected to start enrolling this year, he said.

Dr. Spigel disclosed a consultant/advisory role with Genentech, which sponsored the study. His coauthors disclosed financial relationships with several firms including employment with Genentech.

Publications
Publications
Topics
Article Type
Display Headline
Accurate Biomarker Testing Key to Experimental MetMAb in Lung Cancer
Display Headline
Accurate Biomarker Testing Key to Experimental MetMAb in Lung Cancer
Article Source

FROM THE ANNUAL MEETING OF THE AMERICAN SOCIETY OF CLINICAL ONCOLOGY

PURLs Copyright

Inside the Article

Vitals

Major Finding: Among Met-positive patients, median progression-free survival doubled from 1.5 months with erlotinib to 2.9 months with the addition of MetMAb (HR, 0.53; P = .04).

Data Source: Randomized phase II trial in 137 patients with advanced non–small cell lung cancer.

Disclosures: Dr. Spigel disclosed a consultant/advisory role with Genentech, which sponsored the study. His coauthors disclosed financial relationships with several firms including employment with Genentech.

Statin Cuts Recurrent Stroke Risk Similarly in Diabetic, Nondiabetic Patients

Article Type
Changed
Fri, 12/07/2018 - 14:03
Display Headline
Statin Cuts Recurrent Stroke Risk Similarly in Diabetic, Nondiabetic Patients

Statin therapy appears to reduce the risk of recurrent stroke among patients with diabetes or the metabolic syndrome to the same degree that it does in patients who have neither disorder, according to a planned post hoc analysis of data collected in the Stroke Prevention by Aggressive Reduction in Cholesterol Levels clinical trial.

However, since patients with diabetes start off with a much higher risk of recurrent stroke, their risk remains higher than that of nondiabetic patients even after statin therapy, Dr. Alfred Callahan of Vanderbilt University, Nashville, Tenn., and his associates reported online June 13 in Archives of Neurology.

Until now, no information has been available on the effect of statin treatment on secondary stroke prevention in patients with type 2 diabetes or the metabolic syndrome, the investigators noted.

The primary conclusion of the SPARCL clinical trial was that atorvastatin (Lipitor) reduced stroke risk in general. For this secondary analysis, Dr. Callahan and his colleagues assessed stroke risk in 794 adults who had type 2 diabetes, 642 who had the metabolic syndrome, and a reference group of 3,295 who had neither disorder.

The study subjects were ambulatory men and women with no known coronary heart disease who had had ischemic stroke, hemorrhagic stroke, or transient ischemic attack (TIA) 1-6 months before undergoing randomization in the SPARCL trial. They were treated at 205 medical centers in Africa, Australia, Europe, the Middle East, North America, and South America. The mean age was 63 years, and subjects were assessed every 6 months for a mean of 5 years.

Treatment with atorvastatin decreased LDL cholesterol levels to a similar degree across the three study groups, and lowered triglycerides by 11% in the group with diabetes, 20% in the group with metabolic syndrome, and 9% in the reference group.

Despite these treatment benefits, subjects with diabetes remained at increased risk of recurrent stroke (hazard ratio, 1.62), of major cardiovascular events (HR, 1.66), and of revascularization procedures (HR, 2.39), compared with the reference group. Subjects with metabolic syndrome were at increased risk of revascularization procedures (HR, 1.78) but not of other adverse cardiovascular outcomes.

At the conclusion of the study, the rate of recurrent stroke was 18% in patients with diabetes, 11% in those with metabolic syndrome, and 11% in the reference group.

"There was no evidence of a difference in treatment effect" among the three study groups, Dr. Callahan and his associates said (Arch. Neurol. 2011 June 13 [doi:10.1001/archneurol.2011.146]).

"These results should be viewed as exploratory" because the SPARCL trial was not powered to test for subgroup effects, they noted.

However, the findings agree with those of the Cholesterol Treatment Trialists’ collaboration, which also found that the effect of statins on stroke risk was similar between diabetic and nondiabetic patients, the researchers added.

Pfizer sponsored the study. Dr. Callahan reported receiving support from Pfizer, Sanofi-Aventis, and Bristol-Myers Squibb. His associates reported ties to numerous pharmaceutical and device companies.

Author and Disclosure Information

Publications
Topics
Legacy Keywords
Statin therapy, recurrent stroke, diabetes, metabolic syndrome, Stroke Prevention by Aggressive Reduction in Cholesterol Levels clinical trial, Dr. Alfred Callahan, Archives of Neurology, atorvastatin, Lipitor
Author and Disclosure Information

Author and Disclosure Information

Statin therapy appears to reduce the risk of recurrent stroke among patients with diabetes or the metabolic syndrome to the same degree that it does in patients who have neither disorder, according to a planned post hoc analysis of data collected in the Stroke Prevention by Aggressive Reduction in Cholesterol Levels clinical trial.

However, since patients with diabetes start off with a much higher risk of recurrent stroke, their risk remains higher than that of nondiabetic patients even after statin therapy, Dr. Alfred Callahan of Vanderbilt University, Nashville, Tenn., and his associates reported online June 13 in Archives of Neurology.

Until now, no information has been available on the effect of statin treatment on secondary stroke prevention in patients with type 2 diabetes or the metabolic syndrome, the investigators noted.

The primary conclusion of the SPARCL clinical trial was that atorvastatin (Lipitor) reduced stroke risk in general. For this secondary analysis, Dr. Callahan and his colleagues assessed stroke risk in 794 adults who had type 2 diabetes, 642 who had the metabolic syndrome, and a reference group of 3,295 who had neither disorder.

The study subjects were ambulatory men and women with no known coronary heart disease who had had ischemic stroke, hemorrhagic stroke, or transient ischemic attack (TIA) 1-6 months before undergoing randomization in the SPARCL trial. They were treated at 205 medical centers in Africa, Australia, Europe, the Middle East, North America, and South America. The mean age was 63 years, and subjects were assessed every 6 months for a mean of 5 years.

Treatment with atorvastatin decreased LDL cholesterol levels to a similar degree across the three study groups, and lowered triglycerides by 11% in the group with diabetes, 20% in the group with metabolic syndrome, and 9% in the reference group.

Despite these treatment benefits, subjects with diabetes remained at increased risk of recurrent stroke (hazard ratio, 1.62), of major cardiovascular events (HR, 1.66), and of revascularization procedures (HR, 2.39), compared with the reference group. Subjects with metabolic syndrome were at increased risk of revascularization procedures (HR, 1.78) but not of other adverse cardiovascular outcomes.

At the conclusion of the study, the rate of recurrent stroke was 18% in patients with diabetes, 11% in those with metabolic syndrome, and 11% in the reference group.

"There was no evidence of a difference in treatment effect" among the three study groups, Dr. Callahan and his associates said (Arch. Neurol. 2011 June 13 [doi:10.1001/archneurol.2011.146]).

"These results should be viewed as exploratory" because the SPARCL trial was not powered to test for subgroup effects, they noted.

However, the findings agree with those of the Cholesterol Treatment Trialists’ collaboration, which also found that the effect of statins on stroke risk was similar between diabetic and nondiabetic patients, the researchers added.

Pfizer sponsored the study. Dr. Callahan reported receiving support from Pfizer, Sanofi-Aventis, and Bristol-Myers Squibb. His associates reported ties to numerous pharmaceutical and device companies.

Statin therapy appears to reduce the risk of recurrent stroke among patients with diabetes or the metabolic syndrome to the same degree that it does in patients who have neither disorder, according to a planned post hoc analysis of data collected in the Stroke Prevention by Aggressive Reduction in Cholesterol Levels clinical trial.

However, since patients with diabetes start off with a much higher risk of recurrent stroke, their risk remains higher than that of nondiabetic patients even after statin therapy, Dr. Alfred Callahan of Vanderbilt University, Nashville, Tenn., and his associates reported online June 13 in Archives of Neurology.

Until now, no information has been available on the effect of statin treatment on secondary stroke prevention in patients with type 2 diabetes or the metabolic syndrome, the investigators noted.

The primary conclusion of the SPARCL clinical trial was that atorvastatin (Lipitor) reduced stroke risk in general. For this secondary analysis, Dr. Callahan and his colleagues assessed stroke risk in 794 adults who had type 2 diabetes, 642 who had the metabolic syndrome, and a reference group of 3,295 who had neither disorder.

The study subjects were ambulatory men and women with no known coronary heart disease who had had ischemic stroke, hemorrhagic stroke, or transient ischemic attack (TIA) 1-6 months before undergoing randomization in the SPARCL trial. They were treated at 205 medical centers in Africa, Australia, Europe, the Middle East, North America, and South America. The mean age was 63 years, and subjects were assessed every 6 months for a mean of 5 years.

Treatment with atorvastatin decreased LDL cholesterol levels to a similar degree across the three study groups, and lowered triglycerides by 11% in the group with diabetes, 20% in the group with metabolic syndrome, and 9% in the reference group.

Despite these treatment benefits, subjects with diabetes remained at increased risk of recurrent stroke (hazard ratio, 1.62), of major cardiovascular events (HR, 1.66), and of revascularization procedures (HR, 2.39), compared with the reference group. Subjects with metabolic syndrome were at increased risk of revascularization procedures (HR, 1.78) but not of other adverse cardiovascular outcomes.

At the conclusion of the study, the rate of recurrent stroke was 18% in patients with diabetes, 11% in those with metabolic syndrome, and 11% in the reference group.

"There was no evidence of a difference in treatment effect" among the three study groups, Dr. Callahan and his associates said (Arch. Neurol. 2011 June 13 [doi:10.1001/archneurol.2011.146]).

"These results should be viewed as exploratory" because the SPARCL trial was not powered to test for subgroup effects, they noted.

However, the findings agree with those of the Cholesterol Treatment Trialists’ collaboration, which also found that the effect of statins on stroke risk was similar between diabetic and nondiabetic patients, the researchers added.

Pfizer sponsored the study. Dr. Callahan reported receiving support from Pfizer, Sanofi-Aventis, and Bristol-Myers Squibb. His associates reported ties to numerous pharmaceutical and device companies.

Publications
Publications
Topics
Article Type
Display Headline
Statin Cuts Recurrent Stroke Risk Similarly in Diabetic, Nondiabetic Patients
Display Headline
Statin Cuts Recurrent Stroke Risk Similarly in Diabetic, Nondiabetic Patients
Legacy Keywords
Statin therapy, recurrent stroke, diabetes, metabolic syndrome, Stroke Prevention by Aggressive Reduction in Cholesterol Levels clinical trial, Dr. Alfred Callahan, Archives of Neurology, atorvastatin, Lipitor
Legacy Keywords
Statin therapy, recurrent stroke, diabetes, metabolic syndrome, Stroke Prevention by Aggressive Reduction in Cholesterol Levels clinical trial, Dr. Alfred Callahan, Archives of Neurology, atorvastatin, Lipitor
Article Source

FROM ARCHIVES OF NEUROLOGY

PURLs Copyright

Inside the Article

Vitals

Major Finding: The rate of recurrent stroke was 18% in patients with type 2 diabetes, 11% in patients with metabolic syndrome, and 11% in patients who had neither disorder.

Data Source: A planned secondary analysis of data from the international Stroke Prevention by Aggressive Reduction in Cholesterol Levels (SPARCL) trial involving 4,731 patients with recent stroke or transient ischemic attack who were followed for a mean of 5 years.

Disclosures: Pfizer sponsored the study. Dr. Callahan reported receiving support from Pfizer, Sanofi-Aventis, and Bristol-Myers Squibb. His associates reported ties to numerous pharmaceutical and device companies.

JAK inhibitor ruxolitinib improves treatment landscape in MF

Article Type
Changed
Sat, 06/11/2011 - 05:00
Display Headline
JAK inhibitor ruxolitinib improves treatment landscape in MF

Splenomegaly

CHICAGO—Two phase 3 studies demonstrate the effectiveness of the investigational Janus kinase (JAK) inhibitor INC424 (ruxolitinib) in treating patients with myelofibrosis (MF), according to presentations at the 2011 ASCO Annual Meeting.

Ruxolitinib has the potential to change the treatment landscape in MF, said Alessandro Vannucchi, MD, of the University of Florence, who reported the results of the COMFORT-II trial.

Srdan Verostovsek, MD, PhD, of the MD Anderson Cancer Center, presented results from COMFORT-I.

COMFORT-II

In the COMFORT-II trial, ruxolitinib produced a volumetric spleen size reduction of 35% or greater in 28.5% of MF patients at 48 weeks. None of the patients receiving best available therapy (BAT) experienced such a reduction, Dr Vannucchi said.

At week 24, 31.9% of ruxolitinib-treated patients had a 35% or greater volumetric spleen size reduction, compared to none of the BAT-treated patients. Ruxolitinib also showed a marked improvement in overall quality of life measures, functioning, and symptoms relative to the BAT arm.

This open-label, phase 3 study included 146 patients randomized to ruxolitinib starting at doses of 15 or 20 mg twice daily and 73 patients randomized to BAT, which was administered at doses and schedules determined by the investigator.

Two-thirds of the BAT patients received at least one medication, and one-third received no medication. The most commonly administered agents were hydroxyurea (47% of patients) and glucocorticoids (16% of patients).

All study participants had intermediate-2 or high-risk primary MF, post-polycythemia vera MF, or post-essential thrombocythemia MF.

The safety profile of ruxolitinib was consistent with previous studies, Dr Vannucchi said. The most common grade 3 or higher adverse events in the ruxolitinib arm were anemia and thrombocytopenia. In the BAT arm, the most common events were anemia, thrombocytopenia, pneumonia, and dyspnea.

COMFORT-I

The COMFORT-I trial enrolled 309 patients with intermediate-2 or high-risk primary MF, post-polycythemia vera MF, or post-essential thrombocythemia MF. They were randomly assigned to twice-daily oral ruxolitinib (155 patients) or placebo (154 patients).

Ruxolitinib was dosed at 15 mg or 20 mg, depending on the baseline platelet count. Patients in the placebo arm could cross over to the ruxolitinib arm upon disease progression, and nearly a quarter of patients did cross over (11% prior to week 24 and 13% after week 24).

The median follow-up was 32 weeks. A significantly higher proportion of patients on the ruxolitinib arm (41.9%) attained the primary endpoint of at least a 35% reduction in spleen volume after 24 weeks of therapy compared with placebo patients (0.7%), Dr Verstovsek reported.

Ruxolitinib was also more effective than placebo for improving symptom burden, as 45.9% of patients in the ruxolitinib had at least a 50% improvement in symptom burden, compared to 5.3% of placebo-treated patients.

Symptoms that significantly improved with ruxolitinib included abdominal discomfort, pain under the left ribs, early satiety, night sweats, itching, bone or muscle pain, and inactivity.

“Data from the COMFORT studies indicate that ruxolitinib has the potential to significantly improve the current treatment landscape for most patients with MF,” Dr Vannucchi said.

Publications
Topics

Splenomegaly

CHICAGO—Two phase 3 studies demonstrate the effectiveness of the investigational Janus kinase (JAK) inhibitor INC424 (ruxolitinib) in treating patients with myelofibrosis (MF), according to presentations at the 2011 ASCO Annual Meeting.

Ruxolitinib has the potential to change the treatment landscape in MF, said Alessandro Vannucchi, MD, of the University of Florence, who reported the results of the COMFORT-II trial.

Srdan Verostovsek, MD, PhD, of the MD Anderson Cancer Center, presented results from COMFORT-I.

COMFORT-II

In the COMFORT-II trial, ruxolitinib produced a volumetric spleen size reduction of 35% or greater in 28.5% of MF patients at 48 weeks. None of the patients receiving best available therapy (BAT) experienced such a reduction, Dr Vannucchi said.

At week 24, 31.9% of ruxolitinib-treated patients had a 35% or greater volumetric spleen size reduction, compared to none of the BAT-treated patients. Ruxolitinib also showed a marked improvement in overall quality of life measures, functioning, and symptoms relative to the BAT arm.

This open-label, phase 3 study included 146 patients randomized to ruxolitinib starting at doses of 15 or 20 mg twice daily and 73 patients randomized to BAT, which was administered at doses and schedules determined by the investigator.

Two-thirds of the BAT patients received at least one medication, and one-third received no medication. The most commonly administered agents were hydroxyurea (47% of patients) and glucocorticoids (16% of patients).

All study participants had intermediate-2 or high-risk primary MF, post-polycythemia vera MF, or post-essential thrombocythemia MF.

The safety profile of ruxolitinib was consistent with previous studies, Dr Vannucchi said. The most common grade 3 or higher adverse events in the ruxolitinib arm were anemia and thrombocytopenia. In the BAT arm, the most common events were anemia, thrombocytopenia, pneumonia, and dyspnea.

COMFORT-I

The COMFORT-I trial enrolled 309 patients with intermediate-2 or high-risk primary MF, post-polycythemia vera MF, or post-essential thrombocythemia MF. They were randomly assigned to twice-daily oral ruxolitinib (155 patients) or placebo (154 patients).

Ruxolitinib was dosed at 15 mg or 20 mg, depending on the baseline platelet count. Patients in the placebo arm could cross over to the ruxolitinib arm upon disease progression, and nearly a quarter of patients did cross over (11% prior to week 24 and 13% after week 24).

The median follow-up was 32 weeks. A significantly higher proportion of patients on the ruxolitinib arm (41.9%) attained the primary endpoint of at least a 35% reduction in spleen volume after 24 weeks of therapy compared with placebo patients (0.7%), Dr Verstovsek reported.

Ruxolitinib was also more effective than placebo for improving symptom burden, as 45.9% of patients in the ruxolitinib had at least a 50% improvement in symptom burden, compared to 5.3% of placebo-treated patients.

Symptoms that significantly improved with ruxolitinib included abdominal discomfort, pain under the left ribs, early satiety, night sweats, itching, bone or muscle pain, and inactivity.

“Data from the COMFORT studies indicate that ruxolitinib has the potential to significantly improve the current treatment landscape for most patients with MF,” Dr Vannucchi said.

Splenomegaly

CHICAGO—Two phase 3 studies demonstrate the effectiveness of the investigational Janus kinase (JAK) inhibitor INC424 (ruxolitinib) in treating patients with myelofibrosis (MF), according to presentations at the 2011 ASCO Annual Meeting.

Ruxolitinib has the potential to change the treatment landscape in MF, said Alessandro Vannucchi, MD, of the University of Florence, who reported the results of the COMFORT-II trial.

Srdan Verostovsek, MD, PhD, of the MD Anderson Cancer Center, presented results from COMFORT-I.

COMFORT-II

In the COMFORT-II trial, ruxolitinib produced a volumetric spleen size reduction of 35% or greater in 28.5% of MF patients at 48 weeks. None of the patients receiving best available therapy (BAT) experienced such a reduction, Dr Vannucchi said.

At week 24, 31.9% of ruxolitinib-treated patients had a 35% or greater volumetric spleen size reduction, compared to none of the BAT-treated patients. Ruxolitinib also showed a marked improvement in overall quality of life measures, functioning, and symptoms relative to the BAT arm.

This open-label, phase 3 study included 146 patients randomized to ruxolitinib starting at doses of 15 or 20 mg twice daily and 73 patients randomized to BAT, which was administered at doses and schedules determined by the investigator.

Two-thirds of the BAT patients received at least one medication, and one-third received no medication. The most commonly administered agents were hydroxyurea (47% of patients) and glucocorticoids (16% of patients).

All study participants had intermediate-2 or high-risk primary MF, post-polycythemia vera MF, or post-essential thrombocythemia MF.

The safety profile of ruxolitinib was consistent with previous studies, Dr Vannucchi said. The most common grade 3 or higher adverse events in the ruxolitinib arm were anemia and thrombocytopenia. In the BAT arm, the most common events were anemia, thrombocytopenia, pneumonia, and dyspnea.

COMFORT-I

The COMFORT-I trial enrolled 309 patients with intermediate-2 or high-risk primary MF, post-polycythemia vera MF, or post-essential thrombocythemia MF. They were randomly assigned to twice-daily oral ruxolitinib (155 patients) or placebo (154 patients).

Ruxolitinib was dosed at 15 mg or 20 mg, depending on the baseline platelet count. Patients in the placebo arm could cross over to the ruxolitinib arm upon disease progression, and nearly a quarter of patients did cross over (11% prior to week 24 and 13% after week 24).

The median follow-up was 32 weeks. A significantly higher proportion of patients on the ruxolitinib arm (41.9%) attained the primary endpoint of at least a 35% reduction in spleen volume after 24 weeks of therapy compared with placebo patients (0.7%), Dr Verstovsek reported.

Ruxolitinib was also more effective than placebo for improving symptom burden, as 45.9% of patients in the ruxolitinib had at least a 50% improvement in symptom burden, compared to 5.3% of placebo-treated patients.

Symptoms that significantly improved with ruxolitinib included abdominal discomfort, pain under the left ribs, early satiety, night sweats, itching, bone or muscle pain, and inactivity.

“Data from the COMFORT studies indicate that ruxolitinib has the potential to significantly improve the current treatment landscape for most patients with MF,” Dr Vannucchi said.

Publications
Publications
Topics
Article Type
Display Headline
JAK inhibitor ruxolitinib improves treatment landscape in MF
Display Headline
JAK inhibitor ruxolitinib improves treatment landscape in MF
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

ONLINE EXCLUSIVE: Listen to experts discuss new anticoagulants

Article Type
Changed
Fri, 09/14/2018 - 12:27
Display Headline
ONLINE EXCLUSIVE: Listen to experts discuss new anticoagulants

Click here to listen to Dr. Merli

 

Audio / Podcast
Issue
The Hospitalist - 2011(06)
Publications
Sections
Audio / Podcast
Audio / Podcast

Click here to listen to Dr. Merli

 

Click here to listen to Dr. Merli

 

Issue
The Hospitalist - 2011(06)
Issue
The Hospitalist - 2011(06)
Publications
Publications
Article Type
Display Headline
ONLINE EXCLUSIVE: Listen to experts discuss new anticoagulants
Display Headline
ONLINE EXCLUSIVE: Listen to experts discuss new anticoagulants
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)

Treatment of Diabetes in Emergency Dept.

Article Type
Changed
Mon, 05/22/2017 - 21:44
Display Headline
Treatment of inpatient hyperglycemia beginning in the emergency department: A randomized trial using insulins aspart and detemir compared with usual care

Current consensus guidelines from the American Diabetes Association and the American Association of Clinical Endocrinologists recommend the use of insulin‐based treatment protocols for most hospitalized patients with hyperglycemia.1 For noncritically ill patients, it is recommended to target a fasting blood glucose (BG) < 140 mg/dL and a random BG < 180‐200 mg/dL, without excess hypoglycemia. Prior studies recommended using a basal‐bolus insulin protocol that specifies starting doses and parameters for dose adjustment, applied by well‐educated teams of physicians and nurses.27 We have shown that insulin detemir given as a once‐daily basal injection coupled with rapid‐acting insulin aspart with meals is an effective regimen for managing hyperglycemia in hospitalized patients with type 2 diabetes.7 We and others have shown that once‐daily basal insulin mealtime rapid‐acting insulin is significantly more effective than sliding‐scale regular insulin in the hospital setting.6, 8

The majority of patients admitted to general medical units are first evaluated in the emergency department (ED), and significant hyperglycemia is not uncommon in ED patients. However, protocols for the treatment of hyperglycemia in the ED have not been widely implemented. Ginde et al studied 160 ED patients with a history of diabetes and BG > 200 mg/dL and found that although 73% were admitted to the hospital, only 31% were treated with insulin, and only 18% had a diagnosis of diabetes charted.9 A recent survey of 152 residents and attendings in 3 academic EDs found that only 32% would give insulin for a BG > 200 mg/dL, 59% for a BG > 250 mg/dL, and 91% for a BG > 300 mg/dL to ED patients with known diabetes.10 We completed a preliminary study of a novel protocol for the administration of subcutaneous insulin aspart in the ED at Rush University in 2008.11 We found that the mean BG was significantly lowered during an ED stay, from 333 to 158 mg/dL, and that the protocol was easily adopted by ED staff, with a low rate of hypoglycemia. Historically, only 35% of hyperglycemic patients with diabetes received insulin in our ED.11 Reasons for limited ED management of hyperglycemia may include presence of more critical issues, time and resource restriction, unfamiliarity with glycemic targets, and concerns regarding hypoglycemia.9, 10

In the current study we focused on 3 questions:

  • Could we further reduce the risk of hypoglycemia by modifying our original Rush ED insulin protocol? We reduced insulin aspart from 0.1 to 0.05 units/kg for BG 200‐299 mg/dL, from 0.15 to 0.1 units/kg for BG 300‐399 mg/dL, and from 0.2 to 0.15 units/kg for BG 400 mg/dL.

  • Could we couple our ED insulin aspart protocol with prompt initiation of a detemir‐aspart protocol in those patients who were subsequently admitted to general medical units from the ED?

  • Would the hospital length of stay, mean BG, and incidence of hypoglycemia be improved by the use of 2 back‐to‐back subcutaneous insulin protocols in a randomized clinical trial compared with the usual care provided in the ED and general medical inpatient units?

 

Research Design and Methods

From May 2008 through June 2009, patients presenting to the Rush University Medical Center ED with a history of type 2 diabetes and an initial point‐of‐care BG 200 mg/dL were randomized to an intervention group (INT) or to a usual care group (UC) after giving informed consent. Inclusion criteria for the study were: ages 18‐80 years, history of type 2 diabetes for at least 3 months, and prior therapy with dietary management, oral agents, or insulin. Patients were excluded if subsequently found to have diabetic ketoacidosis, hyperosmolar nonketotic syndrome, or critical illness requiring intensive care unit admission/direct surgical intervention. Other exclusion criteria included a positive pregnancy test or an inability to give informed consent secondary to acute drug or alcohol intoxication or active mental illness. Patients with clinically significant liver disease, with ALT or AST > 3 times the upper limit of normal, or with a history of end‐stage renal disease requiring dialysis were also excluded because of their increased risk of hypoglycemia, as they have required a more conservative insulin regimen. Similarly, we excluded patients with a history of type 1 diabetes because our aspart algorithm for the ED had only been tested in a type 2 diabetes population, and we did not want to disrupt the insulin regimen of type 1 diabetes patients, usually glargine‐based or an insulin pump.

The study consisted of 2 phases. Patients randomized to INT or UC in phase 1 stayed in their respective groups through phase 2. After informed consent was obtained by the study staff, implementation of the protocol was carried out by the ED staff. ED nurses were trained in the study protocol. During phase 1, INT patients received subcutaneous aspart every 2 hours while in the ED if BG was > 200 mg/dL. Aspart dosing per protocol was limited to 2 doses. Uncommonly, when a third dose of aspart was needed, physician input was requested. Aspart dosing was weight‐ and BG level based (0.05 units/kg for BG 200‐299 mg/dL, 0.1 units/kg for BG 300‐399 mg/dL, or 0.15 units/kg for BG 400mg/dL; see Supporting Appendix Fig. 1). Regardless of BG level, the ED aspart protocol was discontinued on patient discharge home or admission to the hospital. UC patients received treatment for hyperglycemia at the discretion of their ED physicians. INT subjects who required hospital admission were transitioned to basal‐bolus insulin therapy with detemir and aspart, receiving their first dose of detemir in the ED. Detemir dosing was weight‐based (0.3 units/kg) if the patient was not on home insulin or based on the patient's home dose of insulin (same dose for detemir, unit‐for‐unit conversion from glargine to detemir, or 80% of total NPH dose). If a patient received basal insulin prior to arrival, the first dose of detemir was held until 12 hours after the last dose of NPH or until 20 hours after the last dose of glargine or detemir. We found from our preliminary study that inadvertent overlaps in long‐acting insulin were one cause of hypoglycemia. We compared differences between groups in the final ED BG level, frequency of hypoglycemia, and ED length of stay (LOS).

Patients subsequently admitted to the hospital entered phase 2. During phase 2, INT patients had detemir and premeal aspart titrated by study staff using a predefined protocol (see Supporting Appendix Fig. 2). Detemir was given once daily, 24 hours after the initial ED dose. UC patients had their diabetes managed by medical house staff teams. House staff members have been educated on the Rush inpatient insulin protocol on which the INT protocol was based. The Rush inpatient diabetes protocol is implemented via a single computerized order set in which all patients should receive mealtime insulin using aspart and a basal insulin (glargine or detemir or NPH). We compared differences between groups in the mean admission BG level, mean daily BG level, mean BG level before each meal, hospital LOS, and frequency of hypoglycemic events. Moderate hypoglycemia was defined as a BG between 50 and 69 mg/dL, and severe hypoglycemia was defined as a BG < 50 mg/dL. We also compared the frequency of BG < 60 mg/dL.

The Rush University Medical Center institutional review board approved the study. Statistical analysis was done using SPSS version 11.0. The Student t test was used to determine any significant difference in BG means between the INT and UC groups. The Fisher's exact test or the chi‐square test was used to determine any difference in proportions of hypoglycemic events between INT and UC patients.

Results

Phase 1: Emergency Department

A total of 176 patients were randomized: 87 to the INT group and 89 to the UC group. Baseline characteristics were similar between groups (Table 1). Mean initial ED BG was similar: 300 70 mg/dL for INT patients and 307 82 mg/dL for UC patients. During phase 1, all INT patients were treated with aspart every 2 hours if BG > 200 mg/dL until discharge home or admission to the hospital. They received an initial mean insulin dose of 0.08 0.04 units/kg of subscutaneous aspart. Twenty‐five percent of INT patients received a second dose of aspart, and 3% received a third dose. For UC patients, only 55% received insulin therapy. Aspart was used for all UC patients who received insulin. Of those in the UC group who received insulin, 11% received a second dose for persistent hyperglycemia; none received a third dose. The mean initial ED BG for UC patients who received insulin was 358 73 mg/dL, and they received an initial mean dose of 0.11 0.05 units/kg. UC patients who did not receive insulin had a lower mean initial ED BG, 241 35 mg/dL. The mean final ED BG was 217 71 mg/dL for the INT group and 257 89 mg/dL for the UC group (P < .01; Fig. 1). The mean ED LOS was 30 minutes longer in the INT group (P = .06; Table 1). Sixty INT patients (69%) and 61 UC patients (69%) were admitted to the hospital. Fifty‐six percent of INT patients received the first dose of detemir based on their home insulin dose, and 44% received a weight‐based dose per protocol.

Figure 1
Blood glucose trend during ED phase (mean ± SEM).
Patient Characteristics and Biochemical Parameters
 Intervention (n = 87)Usual Care (n = 89)Significance
  • Data are in means standard deviation or percentages.

Age (y)55 1355 13 
Sex (% male)4839 
BMI (kg/m2)34 933 9 
Ethnicity (%)
African American5866 
Hispanic2419
White1511
Other34
Duration of diabetes (y)13 912 10 
HA1C10.4 2.29.8 2.6 
Insulin treatment at home (%)5662 
Presenting complaint/diagnosis (%)
Cardiac2023 
Gastrointestinal3023
Hyperglycemia1318
Infection911
Initial ED blood glucose (mg/dL)300 70307 82 
Final ED blood glucose (mg/dL)217 71256 89P < .01
ED length of stay (h)5.4 1.74.9 1.9P = .06
Patients treated with insulin in ED (%)10054 
Initial dose of SQ aspart (units)7.9 4.29.5 4 
ED patients admitted (%)6969 
Admission blood glucose (mg/dL)184 70224 93P < .01
Treatment of hyperglycemia in hospital (%)
Detemir aspart insulin1007 
Glargine aspart insulin036
NPH aspart insulin034
Oral agents08
None015
Hospital length of stay (days)2.7 2.03.1 1.9P = .58

Phase 2: Inpatient Setting

In phase 2, mean admission BG was significantly lower in the INT group (184 70 mg/dL) than in the UC group (223 93 mg/dL), P < .01, as a result of aspart given in the ED. The day 1 mean fasting BG for INT patients was 148 54 mg/dL, significantly lower than the day 1 mean fasting BG for UC patients: 212 81 mg/dL (P < .01). The mean fasting BG for the entire hospitalization was significantly lower for INT patients, 135 48 mg/dL, than for UC patients, 185 72 mg/dL (P < .01). During phase 2, all INT patients had detemir and aspart titrated daily per protocol. Treatment of UC patients was as follows: 78.5% with insulin, 8.2% with oral agents, and 11.5% did not receive medication for hyperglycemia. Of those in the UC group who received insulin, 36.0% were treated with lantus/aspart or detemir/aspart, 34.4% with NPH/aspart, 6.5% with lantus or detemir alone, and 1.6% with aspart alone. Overall, 76.9% of UC received basal insulin, and 70.4% received nutritional insulin. Only 47% of UC patients had insulin adjusted on a daily basis despite persistent hyperglycemia. Significant differences were also seen between INT and UC patients in mean prelunch and predinner BG levels, but not in mean bedtime BG level (Fig. 2). Mean daily BG levels for the initial 5 days of inpatient stay were significantly lower in the INT group (P < .01), except for day 5, when only 21 patients remained in the study (Fig. 3). Patient‐day weighted mean glucose was 163 39 mg/dL for INT patients versus 202 39 mg/dL for UC patients (P < .01). On admission, day 1 mean insulin total daily dose (TDD) was 0.65 0.26 units/kg for INT patients and 0.52 0.29 units/kg for UC patients. The final mean TDD was 0.75 0.35 units/kg for INT patients and 0.61 0.38 units/kg for UC patients. Mean hospital LOS was 9.6 hours shorter for INT patients (2.7 2 days) than for UC patients (3.1 1.9 days), P = .58.

Figure 2
Blood glucose by time of day (mean ± SEM).
Figure 3
Blood glucose by hospital day (mean ± SEM).

Patient Safety: Frequency of Hypoglycemia

The frequency of hypoglycemia is shown in Table 2. During the ED phase, 3 UC patients (3.4%) had a BG < 50 mg/dL, and 2 INT patients (2.3%) had a BG of 67 mg/dL.

Frequency of Hypoglycemia
Blood GlucoseEmergency Department (Number of Episodes)SignificanceInpatient Phase (Patient Stays)Significance
Usual CareInterventionUsual CareIntervention
< 50 mg/dL30P = .5061P = .11
< 60 mg/dL30P = .5078P = .98
5069 mg/dL02P = .23612P = .20

During the hospital phase, INT patients had 4.3% of patient‐days and UC had 4.5% of patient‐days with any BG < 70 mg/dL. During 12 patient‐stays (20%) in the INT group there was an episode of moderate hypoglycemia, and during 1 patient‐stay (1.7%) there was an episode of severe hypoglycemia. During 6 patient‐stays (9.8%) in the UC group there was an episode of moderate hypoglycemia, and during 6 patient‐stays (9.8%) there was an episode of severe hypoglycemia (Table 2). The odds ratio (OR) for moderate hypoglycemia in the INT group compared with the UC group was 1.93 (95% CI, 0.7‐5.29), but for severe hypoglycemia the OR was 0.15 (95% CI, 0.018‐1.33). Moderate and severe hypoglycemic events in the UC group were split evenly between patients treated with glargine/detemir‐aspart and those treated with NPH‐aspart.

Discussion

This is the first randomized trial comparing the Rush Emergency Department Hyperglycemia Intervention (REDHI) protocol with usual care for the treatment of hyperglycemia in the ED. We believe this may be the first trial to initiate subcutaneous basal insulin therapy in the ED at the time of hospital admission. Initiation of our protocol for type 2 diabetic patients with BG > 200 mg/dL resulted in lower final ED and admission BGs compared with those in the UC group. Although a higher mean initial ED BG of 358 73 mg/dL was required to prompt initiation of insulin therapy for UC patients, 3 experienced severe hypoglycemia. By following the REDHI protocol, ED nurses avoided BG < 50 mg/dL in INT patients. Our first version of the REDHI protocol dosed more insulin than our current version, and we saw excess hypoglycemia.11 With a reduced dosing formula, there was less lowering of BG, but we eliminated all BG < 60 mg/dL. There was a trend toward an increased ED LOS in INT patients compared with UC patients. This may be because of delays in the administration of insulin or the requirement for a final BG check prior to discharge from the ED for INT patients. However, we did not receive feedback from ED nursing that either factor was a significant issue.

During phase 2, we observed improved glycemic control in INT, likely due to two factors: early initiation of basal insulin and protocol driven daily titration of both basal and mealtime insulin. We achieved a mean fasting BG of 148 54 mg/dL in INT the morning after the ED dose of detemir. BG levels in both groups continued to improve each day, but since the admission BG for INT was lower, this group maintained significantly lower BG levels throughout most of the hospitalization. There were also significant differences between groups at different times of day. Basal doses for INT patients were adjusted daily per fasting BG. Scheduled mealtime doses were based on the basal dose; each mealtime aspart dose was a third of the basal detemir dose. Therefore, patients who required larger doses for certain meals may have received less aspart than needed. This may explain why fasting BG control was better than BG control later in the day.

Current Rush guidelines recommend the same insulin doses as those that our intervention used, but patients in the UC group were less likely to have insulin titrated daily. Cook et al found that clinical inertia, or failure of health care providers to initiate or intensify therapy when indicated, is a common problem among medical residents treating inpatients with insulin.15 Reasons for clinical inertia may include unawareness of inpatient glycemic targets, lack of training or confidence in titrating insulin, and concerns regarding hypoglycemia. Our study shows that this is still an operative issue, even after residents have participated in multiple small‐group educational sessions. Details of the Rush inpatient insulin protocol are also on pocket cards distributed to residents. However, fewer than half of UC patients had insulin adjusted appropriately for persistent hyperglycemia. This may be one explanation for the improved control seen in the INT group and underscores the importance of daily dose titrations based on a uniform protocol.

During phase 2, despite improved glycemic control in INT, there was no significant difference in rates of hypoglycemia between the groups. The number of patient‐stays with moderate hypoglycemia was more in the INT group than in the UC group, 12 versus 6, respectively, but not statistically different (P = .20). There was a trend toward fewer patient‐stays with severe hypoglycemia in the INT group than in the UC group, 1 versus 6, respectively (P = .11).

Other studies have described improved inpatient glycemic control without excess hypoglycemia. In the RABBIT 2 trial, institution of a glargine‐glulisine insulin protocol, TTD of 0.4‐5 units/kg, among insulin‐naive inpatients resulted in a mean fasting BG of 147 36 mg/dL and a mean hospital BG of 166 32 mg/dL, with 3% of patient‐stays having a BG < 60 mg/dL.6 In a second trial, detemir‐aspart was compared with NPH‐aspart TTD of 0.4‐5 units/kg.7 Both groups achieved a similar mean fasting BG of 146 mg/dL and a mean hospital BG of 157 mg/dL. However, the rate of hypoglycemia was higher: 29% of patient‐stays overall. In our study, we achieved a mean fasting BG of 135 48 mg/dL and a mean hospital BG of 163 40 mg/dL in the INT group, using a mean initial TTD of 0.65 0.23 units/kg. The frequency of hypoglycemia in this trial, 22% of INT patient‐stays and 20% of UC patient‐stays, was less than that in Umpierrez et al,7 despite a lower mean fasting BG in our current trial. Maynard et al found 16% of patient‐stays and 3% of patient‐days had an episode of BG < 60 mg/dL in a trial of glarginerapid‐acting insulin (0.4‐5 units/kg TTD).12 Schnipper et al found that 6.1% of patient‐days had an episode of BG < 60 mg/dL using either glargine or NPH and a rapid‐acting insulin, TTD 0.6 units/kg.13 Our hypoglycemia rates were higher; however, we defined hypoglycemia as BG < 70 mg/dL, as suggested by the ADA workgroup.14 If we use a cutoff of < 60 mg/dL for hypoglycemia, it occurred in 13.3% of patient‐stays and 4.3% of patient‐days in the INT group, comparable to that in previous studies.

Our study has several limitations. First, this was a single‐center study, and our ED protocol should be tested in other ED settings, both academic and community. Second, although there were trends toward lower rates of severe hypoglycemia in the INT group, the study was underpowered to detect possible significant differences. Third, although ED nurses implemented the study protocol, study staff closely monitored nurses to ensure adherence. Therefore, it is difficult to speculate on protocol adherence under normal circumstances. Successful implementation requires ongoing nursing and medical staff education. A fourth limitation is the absence of patients with type 1 diabetes.

In conclusion we demonstrated that weight‐based subcutaneous aspart insulin therapy begun in the ED, coupled with prompt initiation of a detemir‐aspart insulin protocol, results in rapid correction of hyperglycemia and improved inpatient glycemic control without increasing hypoglycemia. Diabetes is a common comorbidity in patients presenting to the ED that is not uniformly addressed. These patients may present with uncontrolled hyperglycemia or diabetes‐related infections, and prompt, efficacious glucose control is important. The nurse‐driven Rush ED hyperglycemia protocol ensures that hyperglycemia is safely addressed, allowing the ED physician to address more critical issues. By initiating basal insulin in the ED, our protocol allows for a prompt and smooth transition to a basal‐bolus insulin regimen for the inpatient setting.

Files
References
  1. Moghissi ES,Korytkowski MT,DiNardo M, et al.American Association of Clinical Endocrinologists and American Diabetes Association Consensus Statement on Inpatient Glycemic Control.Diabetes Care.2009;32:11191131.
  2. Clement S,Braithwaite SS,Magee MF, et al.Management of diabetes and hyperglycemia in hospitals.Diabetes Care.2004;27:553591.
  3. Baldwin D,Villanueva G.,McNutt R.,Bhatnagar S.Eliminating inpatient sliding scale insulin: a re‐education project with medical house staff.Diabetes Care.2008;28:10081011.
  4. Schmeltz LR,DeSantis AJ,Thiyagarajan V, et al.Reduction of surgical mortality and morbidity in diabetic patients undergoing cardiac surgery with a combined intravenous and subcutaneous insulin glucose management strategy.Diabetes Care.2007;30:823828.
  5. Yeldandi R,Lurie A,Baldwin D.Comparison of once daily glargine insulin with twice‐daily NPH/Regular insulin for control of hyperglycemia in inpatients after cardiovascular surgery.Diabetes Technol Ther.2006;8:609616.
  6. Umpierrez GE,Smiley D,Zisman A, et al.Randomized study of basal‐bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial).Diabetes Care.2007;30:21812186.
  7. Umpierrez GE,Hor T,Smiley D, et al.Comparison of inpatient insulin regimens with detemir plus aspart versus neutral protamine Hagedorn plus regular in medical patients with type 2 diabetes.J Clin Endocrinol Metab.2009;94:564569.
  8. Datta S,Qaadir A,Villanueva G,Baldwin D.Once daily insulin glargine vs. six hourly sliding scale regular insulin for control of hyperglycemia after bariatric surgery: a randomized clinical trial.Endocr Pract.2007;13:225231.
  9. Ginde AA,Savaser DJ,Camargo CA.Limited communication and management of emergency department hyperglycemia in hospitalized patients.J Hosp Med.2009;4:4449.
  10. Ginde AA,Delaney KE,Pallin DJ,Camargo CA.Multicenter survey of emergency physician management and referral for hyperglycemia.J Emerg Med.2010;38:264272.
  11. Munoz C,Villanueva G,Fogg L,Johnson T,Baldwin D.Impact of a nurse‐driven subcutaneous insulin protocol: Rush Emergency Department Hyperglycemia Intervention (REDHI).J Emerg Med.2008 [Epub ahead of print].
  12. Maynard G,Lee J,Phillips G,Fink E,Renvall M.Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: effect of structured subcutaneous insulin orders and an insulin management algorithm.J Hosp Med.2009;4:315.
  13. Schnipper JL,Ndumele CD,Liang CL,Pendergrass ML.Effects of a subcutaneous insulin protocol, clinical education, and computerized order set on the quality of inpatient management of hyperglycemia: results of a clinical trial.J Hosp Med.2009;4:1627.
  14. ADA Workgroup on Hypoglycemia.Defining and reporting hypoglycemia in diabetes.Diabetes Care.2005;28:12451249.
  15. Cook CB,Castro JC,Schmidt RE et al.Diabetes care in hospitalized noncritically ill patients: More evidence for clinical inertia and negative therapeutic momentum.J Hosp Med.2007;2:203211.
Article PDF
Issue
Journal of Hospital Medicine - 6(5)
Page Number
279-284
Legacy Keywords
diabetes mellitus, hyperglycemia management, resident education
Sections
Files
Files
Article PDF
Article PDF

Current consensus guidelines from the American Diabetes Association and the American Association of Clinical Endocrinologists recommend the use of insulin‐based treatment protocols for most hospitalized patients with hyperglycemia.1 For noncritically ill patients, it is recommended to target a fasting blood glucose (BG) < 140 mg/dL and a random BG < 180‐200 mg/dL, without excess hypoglycemia. Prior studies recommended using a basal‐bolus insulin protocol that specifies starting doses and parameters for dose adjustment, applied by well‐educated teams of physicians and nurses.27 We have shown that insulin detemir given as a once‐daily basal injection coupled with rapid‐acting insulin aspart with meals is an effective regimen for managing hyperglycemia in hospitalized patients with type 2 diabetes.7 We and others have shown that once‐daily basal insulin mealtime rapid‐acting insulin is significantly more effective than sliding‐scale regular insulin in the hospital setting.6, 8

The majority of patients admitted to general medical units are first evaluated in the emergency department (ED), and significant hyperglycemia is not uncommon in ED patients. However, protocols for the treatment of hyperglycemia in the ED have not been widely implemented. Ginde et al studied 160 ED patients with a history of diabetes and BG > 200 mg/dL and found that although 73% were admitted to the hospital, only 31% were treated with insulin, and only 18% had a diagnosis of diabetes charted.9 A recent survey of 152 residents and attendings in 3 academic EDs found that only 32% would give insulin for a BG > 200 mg/dL, 59% for a BG > 250 mg/dL, and 91% for a BG > 300 mg/dL to ED patients with known diabetes.10 We completed a preliminary study of a novel protocol for the administration of subcutaneous insulin aspart in the ED at Rush University in 2008.11 We found that the mean BG was significantly lowered during an ED stay, from 333 to 158 mg/dL, and that the protocol was easily adopted by ED staff, with a low rate of hypoglycemia. Historically, only 35% of hyperglycemic patients with diabetes received insulin in our ED.11 Reasons for limited ED management of hyperglycemia may include presence of more critical issues, time and resource restriction, unfamiliarity with glycemic targets, and concerns regarding hypoglycemia.9, 10

In the current study we focused on 3 questions:

  • Could we further reduce the risk of hypoglycemia by modifying our original Rush ED insulin protocol? We reduced insulin aspart from 0.1 to 0.05 units/kg for BG 200‐299 mg/dL, from 0.15 to 0.1 units/kg for BG 300‐399 mg/dL, and from 0.2 to 0.15 units/kg for BG 400 mg/dL.

  • Could we couple our ED insulin aspart protocol with prompt initiation of a detemir‐aspart protocol in those patients who were subsequently admitted to general medical units from the ED?

  • Would the hospital length of stay, mean BG, and incidence of hypoglycemia be improved by the use of 2 back‐to‐back subcutaneous insulin protocols in a randomized clinical trial compared with the usual care provided in the ED and general medical inpatient units?

 

Research Design and Methods

From May 2008 through June 2009, patients presenting to the Rush University Medical Center ED with a history of type 2 diabetes and an initial point‐of‐care BG 200 mg/dL were randomized to an intervention group (INT) or to a usual care group (UC) after giving informed consent. Inclusion criteria for the study were: ages 18‐80 years, history of type 2 diabetes for at least 3 months, and prior therapy with dietary management, oral agents, or insulin. Patients were excluded if subsequently found to have diabetic ketoacidosis, hyperosmolar nonketotic syndrome, or critical illness requiring intensive care unit admission/direct surgical intervention. Other exclusion criteria included a positive pregnancy test or an inability to give informed consent secondary to acute drug or alcohol intoxication or active mental illness. Patients with clinically significant liver disease, with ALT or AST > 3 times the upper limit of normal, or with a history of end‐stage renal disease requiring dialysis were also excluded because of their increased risk of hypoglycemia, as they have required a more conservative insulin regimen. Similarly, we excluded patients with a history of type 1 diabetes because our aspart algorithm for the ED had only been tested in a type 2 diabetes population, and we did not want to disrupt the insulin regimen of type 1 diabetes patients, usually glargine‐based or an insulin pump.

The study consisted of 2 phases. Patients randomized to INT or UC in phase 1 stayed in their respective groups through phase 2. After informed consent was obtained by the study staff, implementation of the protocol was carried out by the ED staff. ED nurses were trained in the study protocol. During phase 1, INT patients received subcutaneous aspart every 2 hours while in the ED if BG was > 200 mg/dL. Aspart dosing per protocol was limited to 2 doses. Uncommonly, when a third dose of aspart was needed, physician input was requested. Aspart dosing was weight‐ and BG level based (0.05 units/kg for BG 200‐299 mg/dL, 0.1 units/kg for BG 300‐399 mg/dL, or 0.15 units/kg for BG 400mg/dL; see Supporting Appendix Fig. 1). Regardless of BG level, the ED aspart protocol was discontinued on patient discharge home or admission to the hospital. UC patients received treatment for hyperglycemia at the discretion of their ED physicians. INT subjects who required hospital admission were transitioned to basal‐bolus insulin therapy with detemir and aspart, receiving their first dose of detemir in the ED. Detemir dosing was weight‐based (0.3 units/kg) if the patient was not on home insulin or based on the patient's home dose of insulin (same dose for detemir, unit‐for‐unit conversion from glargine to detemir, or 80% of total NPH dose). If a patient received basal insulin prior to arrival, the first dose of detemir was held until 12 hours after the last dose of NPH or until 20 hours after the last dose of glargine or detemir. We found from our preliminary study that inadvertent overlaps in long‐acting insulin were one cause of hypoglycemia. We compared differences between groups in the final ED BG level, frequency of hypoglycemia, and ED length of stay (LOS).

Patients subsequently admitted to the hospital entered phase 2. During phase 2, INT patients had detemir and premeal aspart titrated by study staff using a predefined protocol (see Supporting Appendix Fig. 2). Detemir was given once daily, 24 hours after the initial ED dose. UC patients had their diabetes managed by medical house staff teams. House staff members have been educated on the Rush inpatient insulin protocol on which the INT protocol was based. The Rush inpatient diabetes protocol is implemented via a single computerized order set in which all patients should receive mealtime insulin using aspart and a basal insulin (glargine or detemir or NPH). We compared differences between groups in the mean admission BG level, mean daily BG level, mean BG level before each meal, hospital LOS, and frequency of hypoglycemic events. Moderate hypoglycemia was defined as a BG between 50 and 69 mg/dL, and severe hypoglycemia was defined as a BG < 50 mg/dL. We also compared the frequency of BG < 60 mg/dL.

The Rush University Medical Center institutional review board approved the study. Statistical analysis was done using SPSS version 11.0. The Student t test was used to determine any significant difference in BG means between the INT and UC groups. The Fisher's exact test or the chi‐square test was used to determine any difference in proportions of hypoglycemic events between INT and UC patients.

Results

Phase 1: Emergency Department

A total of 176 patients were randomized: 87 to the INT group and 89 to the UC group. Baseline characteristics were similar between groups (Table 1). Mean initial ED BG was similar: 300 70 mg/dL for INT patients and 307 82 mg/dL for UC patients. During phase 1, all INT patients were treated with aspart every 2 hours if BG > 200 mg/dL until discharge home or admission to the hospital. They received an initial mean insulin dose of 0.08 0.04 units/kg of subscutaneous aspart. Twenty‐five percent of INT patients received a second dose of aspart, and 3% received a third dose. For UC patients, only 55% received insulin therapy. Aspart was used for all UC patients who received insulin. Of those in the UC group who received insulin, 11% received a second dose for persistent hyperglycemia; none received a third dose. The mean initial ED BG for UC patients who received insulin was 358 73 mg/dL, and they received an initial mean dose of 0.11 0.05 units/kg. UC patients who did not receive insulin had a lower mean initial ED BG, 241 35 mg/dL. The mean final ED BG was 217 71 mg/dL for the INT group and 257 89 mg/dL for the UC group (P < .01; Fig. 1). The mean ED LOS was 30 minutes longer in the INT group (P = .06; Table 1). Sixty INT patients (69%) and 61 UC patients (69%) were admitted to the hospital. Fifty‐six percent of INT patients received the first dose of detemir based on their home insulin dose, and 44% received a weight‐based dose per protocol.

Figure 1
Blood glucose trend during ED phase (mean ± SEM).
Patient Characteristics and Biochemical Parameters
 Intervention (n = 87)Usual Care (n = 89)Significance
  • Data are in means standard deviation or percentages.

Age (y)55 1355 13 
Sex (% male)4839 
BMI (kg/m2)34 933 9 
Ethnicity (%)
African American5866 
Hispanic2419
White1511
Other34
Duration of diabetes (y)13 912 10 
HA1C10.4 2.29.8 2.6 
Insulin treatment at home (%)5662 
Presenting complaint/diagnosis (%)
Cardiac2023 
Gastrointestinal3023
Hyperglycemia1318
Infection911
Initial ED blood glucose (mg/dL)300 70307 82 
Final ED blood glucose (mg/dL)217 71256 89P < .01
ED length of stay (h)5.4 1.74.9 1.9P = .06
Patients treated with insulin in ED (%)10054 
Initial dose of SQ aspart (units)7.9 4.29.5 4 
ED patients admitted (%)6969 
Admission blood glucose (mg/dL)184 70224 93P < .01
Treatment of hyperglycemia in hospital (%)
Detemir aspart insulin1007 
Glargine aspart insulin036
NPH aspart insulin034
Oral agents08
None015
Hospital length of stay (days)2.7 2.03.1 1.9P = .58

Phase 2: Inpatient Setting

In phase 2, mean admission BG was significantly lower in the INT group (184 70 mg/dL) than in the UC group (223 93 mg/dL), P < .01, as a result of aspart given in the ED. The day 1 mean fasting BG for INT patients was 148 54 mg/dL, significantly lower than the day 1 mean fasting BG for UC patients: 212 81 mg/dL (P < .01). The mean fasting BG for the entire hospitalization was significantly lower for INT patients, 135 48 mg/dL, than for UC patients, 185 72 mg/dL (P < .01). During phase 2, all INT patients had detemir and aspart titrated daily per protocol. Treatment of UC patients was as follows: 78.5% with insulin, 8.2% with oral agents, and 11.5% did not receive medication for hyperglycemia. Of those in the UC group who received insulin, 36.0% were treated with lantus/aspart or detemir/aspart, 34.4% with NPH/aspart, 6.5% with lantus or detemir alone, and 1.6% with aspart alone. Overall, 76.9% of UC received basal insulin, and 70.4% received nutritional insulin. Only 47% of UC patients had insulin adjusted on a daily basis despite persistent hyperglycemia. Significant differences were also seen between INT and UC patients in mean prelunch and predinner BG levels, but not in mean bedtime BG level (Fig. 2). Mean daily BG levels for the initial 5 days of inpatient stay were significantly lower in the INT group (P < .01), except for day 5, when only 21 patients remained in the study (Fig. 3). Patient‐day weighted mean glucose was 163 39 mg/dL for INT patients versus 202 39 mg/dL for UC patients (P < .01). On admission, day 1 mean insulin total daily dose (TDD) was 0.65 0.26 units/kg for INT patients and 0.52 0.29 units/kg for UC patients. The final mean TDD was 0.75 0.35 units/kg for INT patients and 0.61 0.38 units/kg for UC patients. Mean hospital LOS was 9.6 hours shorter for INT patients (2.7 2 days) than for UC patients (3.1 1.9 days), P = .58.

Figure 2
Blood glucose by time of day (mean ± SEM).
Figure 3
Blood glucose by hospital day (mean ± SEM).

Patient Safety: Frequency of Hypoglycemia

The frequency of hypoglycemia is shown in Table 2. During the ED phase, 3 UC patients (3.4%) had a BG < 50 mg/dL, and 2 INT patients (2.3%) had a BG of 67 mg/dL.

Frequency of Hypoglycemia
Blood GlucoseEmergency Department (Number of Episodes)SignificanceInpatient Phase (Patient Stays)Significance
Usual CareInterventionUsual CareIntervention
< 50 mg/dL30P = .5061P = .11
< 60 mg/dL30P = .5078P = .98
5069 mg/dL02P = .23612P = .20

During the hospital phase, INT patients had 4.3% of patient‐days and UC had 4.5% of patient‐days with any BG < 70 mg/dL. During 12 patient‐stays (20%) in the INT group there was an episode of moderate hypoglycemia, and during 1 patient‐stay (1.7%) there was an episode of severe hypoglycemia. During 6 patient‐stays (9.8%) in the UC group there was an episode of moderate hypoglycemia, and during 6 patient‐stays (9.8%) there was an episode of severe hypoglycemia (Table 2). The odds ratio (OR) for moderate hypoglycemia in the INT group compared with the UC group was 1.93 (95% CI, 0.7‐5.29), but for severe hypoglycemia the OR was 0.15 (95% CI, 0.018‐1.33). Moderate and severe hypoglycemic events in the UC group were split evenly between patients treated with glargine/detemir‐aspart and those treated with NPH‐aspart.

Discussion

This is the first randomized trial comparing the Rush Emergency Department Hyperglycemia Intervention (REDHI) protocol with usual care for the treatment of hyperglycemia in the ED. We believe this may be the first trial to initiate subcutaneous basal insulin therapy in the ED at the time of hospital admission. Initiation of our protocol for type 2 diabetic patients with BG > 200 mg/dL resulted in lower final ED and admission BGs compared with those in the UC group. Although a higher mean initial ED BG of 358 73 mg/dL was required to prompt initiation of insulin therapy for UC patients, 3 experienced severe hypoglycemia. By following the REDHI protocol, ED nurses avoided BG < 50 mg/dL in INT patients. Our first version of the REDHI protocol dosed more insulin than our current version, and we saw excess hypoglycemia.11 With a reduced dosing formula, there was less lowering of BG, but we eliminated all BG < 60 mg/dL. There was a trend toward an increased ED LOS in INT patients compared with UC patients. This may be because of delays in the administration of insulin or the requirement for a final BG check prior to discharge from the ED for INT patients. However, we did not receive feedback from ED nursing that either factor was a significant issue.

During phase 2, we observed improved glycemic control in INT, likely due to two factors: early initiation of basal insulin and protocol driven daily titration of both basal and mealtime insulin. We achieved a mean fasting BG of 148 54 mg/dL in INT the morning after the ED dose of detemir. BG levels in both groups continued to improve each day, but since the admission BG for INT was lower, this group maintained significantly lower BG levels throughout most of the hospitalization. There were also significant differences between groups at different times of day. Basal doses for INT patients were adjusted daily per fasting BG. Scheduled mealtime doses were based on the basal dose; each mealtime aspart dose was a third of the basal detemir dose. Therefore, patients who required larger doses for certain meals may have received less aspart than needed. This may explain why fasting BG control was better than BG control later in the day.

Current Rush guidelines recommend the same insulin doses as those that our intervention used, but patients in the UC group were less likely to have insulin titrated daily. Cook et al found that clinical inertia, or failure of health care providers to initiate or intensify therapy when indicated, is a common problem among medical residents treating inpatients with insulin.15 Reasons for clinical inertia may include unawareness of inpatient glycemic targets, lack of training or confidence in titrating insulin, and concerns regarding hypoglycemia. Our study shows that this is still an operative issue, even after residents have participated in multiple small‐group educational sessions. Details of the Rush inpatient insulin protocol are also on pocket cards distributed to residents. However, fewer than half of UC patients had insulin adjusted appropriately for persistent hyperglycemia. This may be one explanation for the improved control seen in the INT group and underscores the importance of daily dose titrations based on a uniform protocol.

During phase 2, despite improved glycemic control in INT, there was no significant difference in rates of hypoglycemia between the groups. The number of patient‐stays with moderate hypoglycemia was more in the INT group than in the UC group, 12 versus 6, respectively, but not statistically different (P = .20). There was a trend toward fewer patient‐stays with severe hypoglycemia in the INT group than in the UC group, 1 versus 6, respectively (P = .11).

Other studies have described improved inpatient glycemic control without excess hypoglycemia. In the RABBIT 2 trial, institution of a glargine‐glulisine insulin protocol, TTD of 0.4‐5 units/kg, among insulin‐naive inpatients resulted in a mean fasting BG of 147 36 mg/dL and a mean hospital BG of 166 32 mg/dL, with 3% of patient‐stays having a BG < 60 mg/dL.6 In a second trial, detemir‐aspart was compared with NPH‐aspart TTD of 0.4‐5 units/kg.7 Both groups achieved a similar mean fasting BG of 146 mg/dL and a mean hospital BG of 157 mg/dL. However, the rate of hypoglycemia was higher: 29% of patient‐stays overall. In our study, we achieved a mean fasting BG of 135 48 mg/dL and a mean hospital BG of 163 40 mg/dL in the INT group, using a mean initial TTD of 0.65 0.23 units/kg. The frequency of hypoglycemia in this trial, 22% of INT patient‐stays and 20% of UC patient‐stays, was less than that in Umpierrez et al,7 despite a lower mean fasting BG in our current trial. Maynard et al found 16% of patient‐stays and 3% of patient‐days had an episode of BG < 60 mg/dL in a trial of glarginerapid‐acting insulin (0.4‐5 units/kg TTD).12 Schnipper et al found that 6.1% of patient‐days had an episode of BG < 60 mg/dL using either glargine or NPH and a rapid‐acting insulin, TTD 0.6 units/kg.13 Our hypoglycemia rates were higher; however, we defined hypoglycemia as BG < 70 mg/dL, as suggested by the ADA workgroup.14 If we use a cutoff of < 60 mg/dL for hypoglycemia, it occurred in 13.3% of patient‐stays and 4.3% of patient‐days in the INT group, comparable to that in previous studies.

Our study has several limitations. First, this was a single‐center study, and our ED protocol should be tested in other ED settings, both academic and community. Second, although there were trends toward lower rates of severe hypoglycemia in the INT group, the study was underpowered to detect possible significant differences. Third, although ED nurses implemented the study protocol, study staff closely monitored nurses to ensure adherence. Therefore, it is difficult to speculate on protocol adherence under normal circumstances. Successful implementation requires ongoing nursing and medical staff education. A fourth limitation is the absence of patients with type 1 diabetes.

In conclusion we demonstrated that weight‐based subcutaneous aspart insulin therapy begun in the ED, coupled with prompt initiation of a detemir‐aspart insulin protocol, results in rapid correction of hyperglycemia and improved inpatient glycemic control without increasing hypoglycemia. Diabetes is a common comorbidity in patients presenting to the ED that is not uniformly addressed. These patients may present with uncontrolled hyperglycemia or diabetes‐related infections, and prompt, efficacious glucose control is important. The nurse‐driven Rush ED hyperglycemia protocol ensures that hyperglycemia is safely addressed, allowing the ED physician to address more critical issues. By initiating basal insulin in the ED, our protocol allows for a prompt and smooth transition to a basal‐bolus insulin regimen for the inpatient setting.

Current consensus guidelines from the American Diabetes Association and the American Association of Clinical Endocrinologists recommend the use of insulin‐based treatment protocols for most hospitalized patients with hyperglycemia.1 For noncritically ill patients, it is recommended to target a fasting blood glucose (BG) < 140 mg/dL and a random BG < 180‐200 mg/dL, without excess hypoglycemia. Prior studies recommended using a basal‐bolus insulin protocol that specifies starting doses and parameters for dose adjustment, applied by well‐educated teams of physicians and nurses.27 We have shown that insulin detemir given as a once‐daily basal injection coupled with rapid‐acting insulin aspart with meals is an effective regimen for managing hyperglycemia in hospitalized patients with type 2 diabetes.7 We and others have shown that once‐daily basal insulin mealtime rapid‐acting insulin is significantly more effective than sliding‐scale regular insulin in the hospital setting.6, 8

The majority of patients admitted to general medical units are first evaluated in the emergency department (ED), and significant hyperglycemia is not uncommon in ED patients. However, protocols for the treatment of hyperglycemia in the ED have not been widely implemented. Ginde et al studied 160 ED patients with a history of diabetes and BG > 200 mg/dL and found that although 73% were admitted to the hospital, only 31% were treated with insulin, and only 18% had a diagnosis of diabetes charted.9 A recent survey of 152 residents and attendings in 3 academic EDs found that only 32% would give insulin for a BG > 200 mg/dL, 59% for a BG > 250 mg/dL, and 91% for a BG > 300 mg/dL to ED patients with known diabetes.10 We completed a preliminary study of a novel protocol for the administration of subcutaneous insulin aspart in the ED at Rush University in 2008.11 We found that the mean BG was significantly lowered during an ED stay, from 333 to 158 mg/dL, and that the protocol was easily adopted by ED staff, with a low rate of hypoglycemia. Historically, only 35% of hyperglycemic patients with diabetes received insulin in our ED.11 Reasons for limited ED management of hyperglycemia may include presence of more critical issues, time and resource restriction, unfamiliarity with glycemic targets, and concerns regarding hypoglycemia.9, 10

In the current study we focused on 3 questions:

  • Could we further reduce the risk of hypoglycemia by modifying our original Rush ED insulin protocol? We reduced insulin aspart from 0.1 to 0.05 units/kg for BG 200‐299 mg/dL, from 0.15 to 0.1 units/kg for BG 300‐399 mg/dL, and from 0.2 to 0.15 units/kg for BG 400 mg/dL.

  • Could we couple our ED insulin aspart protocol with prompt initiation of a detemir‐aspart protocol in those patients who were subsequently admitted to general medical units from the ED?

  • Would the hospital length of stay, mean BG, and incidence of hypoglycemia be improved by the use of 2 back‐to‐back subcutaneous insulin protocols in a randomized clinical trial compared with the usual care provided in the ED and general medical inpatient units?

 

Research Design and Methods

From May 2008 through June 2009, patients presenting to the Rush University Medical Center ED with a history of type 2 diabetes and an initial point‐of‐care BG 200 mg/dL were randomized to an intervention group (INT) or to a usual care group (UC) after giving informed consent. Inclusion criteria for the study were: ages 18‐80 years, history of type 2 diabetes for at least 3 months, and prior therapy with dietary management, oral agents, or insulin. Patients were excluded if subsequently found to have diabetic ketoacidosis, hyperosmolar nonketotic syndrome, or critical illness requiring intensive care unit admission/direct surgical intervention. Other exclusion criteria included a positive pregnancy test or an inability to give informed consent secondary to acute drug or alcohol intoxication or active mental illness. Patients with clinically significant liver disease, with ALT or AST > 3 times the upper limit of normal, or with a history of end‐stage renal disease requiring dialysis were also excluded because of their increased risk of hypoglycemia, as they have required a more conservative insulin regimen. Similarly, we excluded patients with a history of type 1 diabetes because our aspart algorithm for the ED had only been tested in a type 2 diabetes population, and we did not want to disrupt the insulin regimen of type 1 diabetes patients, usually glargine‐based or an insulin pump.

The study consisted of 2 phases. Patients randomized to INT or UC in phase 1 stayed in their respective groups through phase 2. After informed consent was obtained by the study staff, implementation of the protocol was carried out by the ED staff. ED nurses were trained in the study protocol. During phase 1, INT patients received subcutaneous aspart every 2 hours while in the ED if BG was > 200 mg/dL. Aspart dosing per protocol was limited to 2 doses. Uncommonly, when a third dose of aspart was needed, physician input was requested. Aspart dosing was weight‐ and BG level based (0.05 units/kg for BG 200‐299 mg/dL, 0.1 units/kg for BG 300‐399 mg/dL, or 0.15 units/kg for BG 400mg/dL; see Supporting Appendix Fig. 1). Regardless of BG level, the ED aspart protocol was discontinued on patient discharge home or admission to the hospital. UC patients received treatment for hyperglycemia at the discretion of their ED physicians. INT subjects who required hospital admission were transitioned to basal‐bolus insulin therapy with detemir and aspart, receiving their first dose of detemir in the ED. Detemir dosing was weight‐based (0.3 units/kg) if the patient was not on home insulin or based on the patient's home dose of insulin (same dose for detemir, unit‐for‐unit conversion from glargine to detemir, or 80% of total NPH dose). If a patient received basal insulin prior to arrival, the first dose of detemir was held until 12 hours after the last dose of NPH or until 20 hours after the last dose of glargine or detemir. We found from our preliminary study that inadvertent overlaps in long‐acting insulin were one cause of hypoglycemia. We compared differences between groups in the final ED BG level, frequency of hypoglycemia, and ED length of stay (LOS).

Patients subsequently admitted to the hospital entered phase 2. During phase 2, INT patients had detemir and premeal aspart titrated by study staff using a predefined protocol (see Supporting Appendix Fig. 2). Detemir was given once daily, 24 hours after the initial ED dose. UC patients had their diabetes managed by medical house staff teams. House staff members have been educated on the Rush inpatient insulin protocol on which the INT protocol was based. The Rush inpatient diabetes protocol is implemented via a single computerized order set in which all patients should receive mealtime insulin using aspart and a basal insulin (glargine or detemir or NPH). We compared differences between groups in the mean admission BG level, mean daily BG level, mean BG level before each meal, hospital LOS, and frequency of hypoglycemic events. Moderate hypoglycemia was defined as a BG between 50 and 69 mg/dL, and severe hypoglycemia was defined as a BG < 50 mg/dL. We also compared the frequency of BG < 60 mg/dL.

The Rush University Medical Center institutional review board approved the study. Statistical analysis was done using SPSS version 11.0. The Student t test was used to determine any significant difference in BG means between the INT and UC groups. The Fisher's exact test or the chi‐square test was used to determine any difference in proportions of hypoglycemic events between INT and UC patients.

Results

Phase 1: Emergency Department

A total of 176 patients were randomized: 87 to the INT group and 89 to the UC group. Baseline characteristics were similar between groups (Table 1). Mean initial ED BG was similar: 300 70 mg/dL for INT patients and 307 82 mg/dL for UC patients. During phase 1, all INT patients were treated with aspart every 2 hours if BG > 200 mg/dL until discharge home or admission to the hospital. They received an initial mean insulin dose of 0.08 0.04 units/kg of subscutaneous aspart. Twenty‐five percent of INT patients received a second dose of aspart, and 3% received a third dose. For UC patients, only 55% received insulin therapy. Aspart was used for all UC patients who received insulin. Of those in the UC group who received insulin, 11% received a second dose for persistent hyperglycemia; none received a third dose. The mean initial ED BG for UC patients who received insulin was 358 73 mg/dL, and they received an initial mean dose of 0.11 0.05 units/kg. UC patients who did not receive insulin had a lower mean initial ED BG, 241 35 mg/dL. The mean final ED BG was 217 71 mg/dL for the INT group and 257 89 mg/dL for the UC group (P < .01; Fig. 1). The mean ED LOS was 30 minutes longer in the INT group (P = .06; Table 1). Sixty INT patients (69%) and 61 UC patients (69%) were admitted to the hospital. Fifty‐six percent of INT patients received the first dose of detemir based on their home insulin dose, and 44% received a weight‐based dose per protocol.

Figure 1
Blood glucose trend during ED phase (mean ± SEM).
Patient Characteristics and Biochemical Parameters
 Intervention (n = 87)Usual Care (n = 89)Significance
  • Data are in means standard deviation or percentages.

Age (y)55 1355 13 
Sex (% male)4839 
BMI (kg/m2)34 933 9 
Ethnicity (%)
African American5866 
Hispanic2419
White1511
Other34
Duration of diabetes (y)13 912 10 
HA1C10.4 2.29.8 2.6 
Insulin treatment at home (%)5662 
Presenting complaint/diagnosis (%)
Cardiac2023 
Gastrointestinal3023
Hyperglycemia1318
Infection911
Initial ED blood glucose (mg/dL)300 70307 82 
Final ED blood glucose (mg/dL)217 71256 89P < .01
ED length of stay (h)5.4 1.74.9 1.9P = .06
Patients treated with insulin in ED (%)10054 
Initial dose of SQ aspart (units)7.9 4.29.5 4 
ED patients admitted (%)6969 
Admission blood glucose (mg/dL)184 70224 93P < .01
Treatment of hyperglycemia in hospital (%)
Detemir aspart insulin1007 
Glargine aspart insulin036
NPH aspart insulin034
Oral agents08
None015
Hospital length of stay (days)2.7 2.03.1 1.9P = .58

Phase 2: Inpatient Setting

In phase 2, mean admission BG was significantly lower in the INT group (184 70 mg/dL) than in the UC group (223 93 mg/dL), P < .01, as a result of aspart given in the ED. The day 1 mean fasting BG for INT patients was 148 54 mg/dL, significantly lower than the day 1 mean fasting BG for UC patients: 212 81 mg/dL (P < .01). The mean fasting BG for the entire hospitalization was significantly lower for INT patients, 135 48 mg/dL, than for UC patients, 185 72 mg/dL (P < .01). During phase 2, all INT patients had detemir and aspart titrated daily per protocol. Treatment of UC patients was as follows: 78.5% with insulin, 8.2% with oral agents, and 11.5% did not receive medication for hyperglycemia. Of those in the UC group who received insulin, 36.0% were treated with lantus/aspart or detemir/aspart, 34.4% with NPH/aspart, 6.5% with lantus or detemir alone, and 1.6% with aspart alone. Overall, 76.9% of UC received basal insulin, and 70.4% received nutritional insulin. Only 47% of UC patients had insulin adjusted on a daily basis despite persistent hyperglycemia. Significant differences were also seen between INT and UC patients in mean prelunch and predinner BG levels, but not in mean bedtime BG level (Fig. 2). Mean daily BG levels for the initial 5 days of inpatient stay were significantly lower in the INT group (P < .01), except for day 5, when only 21 patients remained in the study (Fig. 3). Patient‐day weighted mean glucose was 163 39 mg/dL for INT patients versus 202 39 mg/dL for UC patients (P < .01). On admission, day 1 mean insulin total daily dose (TDD) was 0.65 0.26 units/kg for INT patients and 0.52 0.29 units/kg for UC patients. The final mean TDD was 0.75 0.35 units/kg for INT patients and 0.61 0.38 units/kg for UC patients. Mean hospital LOS was 9.6 hours shorter for INT patients (2.7 2 days) than for UC patients (3.1 1.9 days), P = .58.

Figure 2
Blood glucose by time of day (mean ± SEM).
Figure 3
Blood glucose by hospital day (mean ± SEM).

Patient Safety: Frequency of Hypoglycemia

The frequency of hypoglycemia is shown in Table 2. During the ED phase, 3 UC patients (3.4%) had a BG < 50 mg/dL, and 2 INT patients (2.3%) had a BG of 67 mg/dL.

Frequency of Hypoglycemia
Blood GlucoseEmergency Department (Number of Episodes)SignificanceInpatient Phase (Patient Stays)Significance
Usual CareInterventionUsual CareIntervention
< 50 mg/dL30P = .5061P = .11
< 60 mg/dL30P = .5078P = .98
5069 mg/dL02P = .23612P = .20

During the hospital phase, INT patients had 4.3% of patient‐days and UC had 4.5% of patient‐days with any BG < 70 mg/dL. During 12 patient‐stays (20%) in the INT group there was an episode of moderate hypoglycemia, and during 1 patient‐stay (1.7%) there was an episode of severe hypoglycemia. During 6 patient‐stays (9.8%) in the UC group there was an episode of moderate hypoglycemia, and during 6 patient‐stays (9.8%) there was an episode of severe hypoglycemia (Table 2). The odds ratio (OR) for moderate hypoglycemia in the INT group compared with the UC group was 1.93 (95% CI, 0.7‐5.29), but for severe hypoglycemia the OR was 0.15 (95% CI, 0.018‐1.33). Moderate and severe hypoglycemic events in the UC group were split evenly between patients treated with glargine/detemir‐aspart and those treated with NPH‐aspart.

Discussion

This is the first randomized trial comparing the Rush Emergency Department Hyperglycemia Intervention (REDHI) protocol with usual care for the treatment of hyperglycemia in the ED. We believe this may be the first trial to initiate subcutaneous basal insulin therapy in the ED at the time of hospital admission. Initiation of our protocol for type 2 diabetic patients with BG > 200 mg/dL resulted in lower final ED and admission BGs compared with those in the UC group. Although a higher mean initial ED BG of 358 73 mg/dL was required to prompt initiation of insulin therapy for UC patients, 3 experienced severe hypoglycemia. By following the REDHI protocol, ED nurses avoided BG < 50 mg/dL in INT patients. Our first version of the REDHI protocol dosed more insulin than our current version, and we saw excess hypoglycemia.11 With a reduced dosing formula, there was less lowering of BG, but we eliminated all BG < 60 mg/dL. There was a trend toward an increased ED LOS in INT patients compared with UC patients. This may be because of delays in the administration of insulin or the requirement for a final BG check prior to discharge from the ED for INT patients. However, we did not receive feedback from ED nursing that either factor was a significant issue.

During phase 2, we observed improved glycemic control in INT, likely due to two factors: early initiation of basal insulin and protocol driven daily titration of both basal and mealtime insulin. We achieved a mean fasting BG of 148 54 mg/dL in INT the morning after the ED dose of detemir. BG levels in both groups continued to improve each day, but since the admission BG for INT was lower, this group maintained significantly lower BG levels throughout most of the hospitalization. There were also significant differences between groups at different times of day. Basal doses for INT patients were adjusted daily per fasting BG. Scheduled mealtime doses were based on the basal dose; each mealtime aspart dose was a third of the basal detemir dose. Therefore, patients who required larger doses for certain meals may have received less aspart than needed. This may explain why fasting BG control was better than BG control later in the day.

Current Rush guidelines recommend the same insulin doses as those that our intervention used, but patients in the UC group were less likely to have insulin titrated daily. Cook et al found that clinical inertia, or failure of health care providers to initiate or intensify therapy when indicated, is a common problem among medical residents treating inpatients with insulin.15 Reasons for clinical inertia may include unawareness of inpatient glycemic targets, lack of training or confidence in titrating insulin, and concerns regarding hypoglycemia. Our study shows that this is still an operative issue, even after residents have participated in multiple small‐group educational sessions. Details of the Rush inpatient insulin protocol are also on pocket cards distributed to residents. However, fewer than half of UC patients had insulin adjusted appropriately for persistent hyperglycemia. This may be one explanation for the improved control seen in the INT group and underscores the importance of daily dose titrations based on a uniform protocol.

During phase 2, despite improved glycemic control in INT, there was no significant difference in rates of hypoglycemia between the groups. The number of patient‐stays with moderate hypoglycemia was more in the INT group than in the UC group, 12 versus 6, respectively, but not statistically different (P = .20). There was a trend toward fewer patient‐stays with severe hypoglycemia in the INT group than in the UC group, 1 versus 6, respectively (P = .11).

Other studies have described improved inpatient glycemic control without excess hypoglycemia. In the RABBIT 2 trial, institution of a glargine‐glulisine insulin protocol, TTD of 0.4‐5 units/kg, among insulin‐naive inpatients resulted in a mean fasting BG of 147 36 mg/dL and a mean hospital BG of 166 32 mg/dL, with 3% of patient‐stays having a BG < 60 mg/dL.6 In a second trial, detemir‐aspart was compared with NPH‐aspart TTD of 0.4‐5 units/kg.7 Both groups achieved a similar mean fasting BG of 146 mg/dL and a mean hospital BG of 157 mg/dL. However, the rate of hypoglycemia was higher: 29% of patient‐stays overall. In our study, we achieved a mean fasting BG of 135 48 mg/dL and a mean hospital BG of 163 40 mg/dL in the INT group, using a mean initial TTD of 0.65 0.23 units/kg. The frequency of hypoglycemia in this trial, 22% of INT patient‐stays and 20% of UC patient‐stays, was less than that in Umpierrez et al,7 despite a lower mean fasting BG in our current trial. Maynard et al found 16% of patient‐stays and 3% of patient‐days had an episode of BG < 60 mg/dL in a trial of glarginerapid‐acting insulin (0.4‐5 units/kg TTD).12 Schnipper et al found that 6.1% of patient‐days had an episode of BG < 60 mg/dL using either glargine or NPH and a rapid‐acting insulin, TTD 0.6 units/kg.13 Our hypoglycemia rates were higher; however, we defined hypoglycemia as BG < 70 mg/dL, as suggested by the ADA workgroup.14 If we use a cutoff of < 60 mg/dL for hypoglycemia, it occurred in 13.3% of patient‐stays and 4.3% of patient‐days in the INT group, comparable to that in previous studies.

Our study has several limitations. First, this was a single‐center study, and our ED protocol should be tested in other ED settings, both academic and community. Second, although there were trends toward lower rates of severe hypoglycemia in the INT group, the study was underpowered to detect possible significant differences. Third, although ED nurses implemented the study protocol, study staff closely monitored nurses to ensure adherence. Therefore, it is difficult to speculate on protocol adherence under normal circumstances. Successful implementation requires ongoing nursing and medical staff education. A fourth limitation is the absence of patients with type 1 diabetes.

In conclusion we demonstrated that weight‐based subcutaneous aspart insulin therapy begun in the ED, coupled with prompt initiation of a detemir‐aspart insulin protocol, results in rapid correction of hyperglycemia and improved inpatient glycemic control without increasing hypoglycemia. Diabetes is a common comorbidity in patients presenting to the ED that is not uniformly addressed. These patients may present with uncontrolled hyperglycemia or diabetes‐related infections, and prompt, efficacious glucose control is important. The nurse‐driven Rush ED hyperglycemia protocol ensures that hyperglycemia is safely addressed, allowing the ED physician to address more critical issues. By initiating basal insulin in the ED, our protocol allows for a prompt and smooth transition to a basal‐bolus insulin regimen for the inpatient setting.

References
  1. Moghissi ES,Korytkowski MT,DiNardo M, et al.American Association of Clinical Endocrinologists and American Diabetes Association Consensus Statement on Inpatient Glycemic Control.Diabetes Care.2009;32:11191131.
  2. Clement S,Braithwaite SS,Magee MF, et al.Management of diabetes and hyperglycemia in hospitals.Diabetes Care.2004;27:553591.
  3. Baldwin D,Villanueva G.,McNutt R.,Bhatnagar S.Eliminating inpatient sliding scale insulin: a re‐education project with medical house staff.Diabetes Care.2008;28:10081011.
  4. Schmeltz LR,DeSantis AJ,Thiyagarajan V, et al.Reduction of surgical mortality and morbidity in diabetic patients undergoing cardiac surgery with a combined intravenous and subcutaneous insulin glucose management strategy.Diabetes Care.2007;30:823828.
  5. Yeldandi R,Lurie A,Baldwin D.Comparison of once daily glargine insulin with twice‐daily NPH/Regular insulin for control of hyperglycemia in inpatients after cardiovascular surgery.Diabetes Technol Ther.2006;8:609616.
  6. Umpierrez GE,Smiley D,Zisman A, et al.Randomized study of basal‐bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial).Diabetes Care.2007;30:21812186.
  7. Umpierrez GE,Hor T,Smiley D, et al.Comparison of inpatient insulin regimens with detemir plus aspart versus neutral protamine Hagedorn plus regular in medical patients with type 2 diabetes.J Clin Endocrinol Metab.2009;94:564569.
  8. Datta S,Qaadir A,Villanueva G,Baldwin D.Once daily insulin glargine vs. six hourly sliding scale regular insulin for control of hyperglycemia after bariatric surgery: a randomized clinical trial.Endocr Pract.2007;13:225231.
  9. Ginde AA,Savaser DJ,Camargo CA.Limited communication and management of emergency department hyperglycemia in hospitalized patients.J Hosp Med.2009;4:4449.
  10. Ginde AA,Delaney KE,Pallin DJ,Camargo CA.Multicenter survey of emergency physician management and referral for hyperglycemia.J Emerg Med.2010;38:264272.
  11. Munoz C,Villanueva G,Fogg L,Johnson T,Baldwin D.Impact of a nurse‐driven subcutaneous insulin protocol: Rush Emergency Department Hyperglycemia Intervention (REDHI).J Emerg Med.2008 [Epub ahead of print].
  12. Maynard G,Lee J,Phillips G,Fink E,Renvall M.Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: effect of structured subcutaneous insulin orders and an insulin management algorithm.J Hosp Med.2009;4:315.
  13. Schnipper JL,Ndumele CD,Liang CL,Pendergrass ML.Effects of a subcutaneous insulin protocol, clinical education, and computerized order set on the quality of inpatient management of hyperglycemia: results of a clinical trial.J Hosp Med.2009;4:1627.
  14. ADA Workgroup on Hypoglycemia.Defining and reporting hypoglycemia in diabetes.Diabetes Care.2005;28:12451249.
  15. Cook CB,Castro JC,Schmidt RE et al.Diabetes care in hospitalized noncritically ill patients: More evidence for clinical inertia and negative therapeutic momentum.J Hosp Med.2007;2:203211.
References
  1. Moghissi ES,Korytkowski MT,DiNardo M, et al.American Association of Clinical Endocrinologists and American Diabetes Association Consensus Statement on Inpatient Glycemic Control.Diabetes Care.2009;32:11191131.
  2. Clement S,Braithwaite SS,Magee MF, et al.Management of diabetes and hyperglycemia in hospitals.Diabetes Care.2004;27:553591.
  3. Baldwin D,Villanueva G.,McNutt R.,Bhatnagar S.Eliminating inpatient sliding scale insulin: a re‐education project with medical house staff.Diabetes Care.2008;28:10081011.
  4. Schmeltz LR,DeSantis AJ,Thiyagarajan V, et al.Reduction of surgical mortality and morbidity in diabetic patients undergoing cardiac surgery with a combined intravenous and subcutaneous insulin glucose management strategy.Diabetes Care.2007;30:823828.
  5. Yeldandi R,Lurie A,Baldwin D.Comparison of once daily glargine insulin with twice‐daily NPH/Regular insulin for control of hyperglycemia in inpatients after cardiovascular surgery.Diabetes Technol Ther.2006;8:609616.
  6. Umpierrez GE,Smiley D,Zisman A, et al.Randomized study of basal‐bolus insulin therapy in the inpatient management of patients with type 2 diabetes (RABBIT 2 trial).Diabetes Care.2007;30:21812186.
  7. Umpierrez GE,Hor T,Smiley D, et al.Comparison of inpatient insulin regimens with detemir plus aspart versus neutral protamine Hagedorn plus regular in medical patients with type 2 diabetes.J Clin Endocrinol Metab.2009;94:564569.
  8. Datta S,Qaadir A,Villanueva G,Baldwin D.Once daily insulin glargine vs. six hourly sliding scale regular insulin for control of hyperglycemia after bariatric surgery: a randomized clinical trial.Endocr Pract.2007;13:225231.
  9. Ginde AA,Savaser DJ,Camargo CA.Limited communication and management of emergency department hyperglycemia in hospitalized patients.J Hosp Med.2009;4:4449.
  10. Ginde AA,Delaney KE,Pallin DJ,Camargo CA.Multicenter survey of emergency physician management and referral for hyperglycemia.J Emerg Med.2010;38:264272.
  11. Munoz C,Villanueva G,Fogg L,Johnson T,Baldwin D.Impact of a nurse‐driven subcutaneous insulin protocol: Rush Emergency Department Hyperglycemia Intervention (REDHI).J Emerg Med.2008 [Epub ahead of print].
  12. Maynard G,Lee J,Phillips G,Fink E,Renvall M.Improved inpatient use of basal insulin, reduced hypoglycemia, and improved glycemic control: effect of structured subcutaneous insulin orders and an insulin management algorithm.J Hosp Med.2009;4:315.
  13. Schnipper JL,Ndumele CD,Liang CL,Pendergrass ML.Effects of a subcutaneous insulin protocol, clinical education, and computerized order set on the quality of inpatient management of hyperglycemia: results of a clinical trial.J Hosp Med.2009;4:1627.
  14. ADA Workgroup on Hypoglycemia.Defining and reporting hypoglycemia in diabetes.Diabetes Care.2005;28:12451249.
  15. Cook CB,Castro JC,Schmidt RE et al.Diabetes care in hospitalized noncritically ill patients: More evidence for clinical inertia and negative therapeutic momentum.J Hosp Med.2007;2:203211.
Issue
Journal of Hospital Medicine - 6(5)
Issue
Journal of Hospital Medicine - 6(5)
Page Number
279-284
Page Number
279-284
Article Type
Display Headline
Treatment of inpatient hyperglycemia beginning in the emergency department: A randomized trial using insulins aspart and detemir compared with usual care
Display Headline
Treatment of inpatient hyperglycemia beginning in the emergency department: A randomized trial using insulins aspart and detemir compared with usual care
Legacy Keywords
diabetes mellitus, hyperglycemia management, resident education
Legacy Keywords
diabetes mellitus, hyperglycemia management, resident education
Sections
Article Source

Copyright © 2010 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
1725 W. Harrison St., Suite 250, Chicago, IL 60612
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media
Media Files

ECG Score as Embolism Predictor

Article Type
Changed
Mon, 05/22/2017 - 21:43
Display Headline
Electrocardiogram score predicts severity of pulmonary embolism in hemodynamically stable patients

The clinical outcome of patients with pulmonary embolism (PE) has a wide spectrum, ranging from a benign course, with early resolution of symptoms, to death. Prognostic stratification of patients with PE is therefore essential to guide therapy. Currently, this stratification is primarily based on blood pressure on admission. Hemodynamic instability or shock carries a bad prognosis, and is associated with a 3‐fold to 7‐fold increase in mortality.1 Thrombolytic therapy is warranted in these cases.2 There is a subset of patients that, despite normal blood pressure on presentation, can subsequently suffer hemodynamic deterioration and death.3

Echocardiography has been proposed as a useful tool to identify a subset of patients at a higher risk. It has been suggested that the finding of right ventricle dysfunction (RVD) in echocardiography predicts a worse prognosis, even in patients with normal blood pressure on presentation.4 Also, computed tomography pulmonary angiography (CTPA) is increasingly used as the main test to diagnose PE. This technique can also assess the degree of pulmonary arterial obstruction and the presence of signs suggestive of RVD. It has been suggested that CTPA findings can predict outcome in PE.5

Electrocardiography (ECG) has a widespread use and is routinely carried out and interpreted in the emergency room. Some ECG findings in patients with PE are suggestive of right ventricular strain and might therefore be used to prioritize cases that require closer monitoring.6

This study was designed with the objectives of assessing the correlation of standard ECG findings with echocardiography and CTPA signs of RVD, with the anatomic severity of the pulmonary vascular obstruction, and with the degree of hypoxemia, in hemodynamically stable patients with PE. Secondary objectives were determining if ECG could accurately predict RVD, and determining the interobserver agreement for the ECG score.

Patients and Methods

Patients and Study Design

We recently carried out a prospective, descriptive, single‐center follow‐up study to assess the prevalence of RVD and pulmonary hypertension (PH) in PE patients who presented with hemodynamic stability.7 We herein analyze ECG findings in these cases. The present, noninterventional study was approved by our institution's review board. The population studied included consecutive patients presenting with PE to our emergency room. Inclusion criteria were age >18 years and PE confirmed by CTPA. Exclusion criteria were high‐risk PE (presence of 2 consecutive systolic blood pressure measurements <90 mm Hg, measured >15 minutes apart or requiring inotropic support); comorbidity predictive of a 6‐month mortality >50% (eg, metastatic cancer or end‐stage respiratory or heart disease); creatinine clearance <35 mL/minute; or allergy to ionidated contrast media. All the patients gave informed consent to participate in the study.

Protocol

Initial assessment included clinical history, physical examination, ECG, arterial blood gas analysis, echocardiography, CTPA, and lower‐limb venous ultrasonography. All included individuals were treated with anticoagulation, and fibrinolytic therapy was not used.

ECG

A standard 12‐lead ECG was promptly obtained for all patients in the emergency room. We used the scoring system described by Daniel et al.8 (Table 1). This score is based on 4 signs that were previously found to be associated with high‐risk PE: tachycardia, incomplete or complete right bundle branch block, T wave inversion, and S1Q3T3 pattern. Two investigators trained in the interpretation of ECG results scored the ECG independently, and were blinded to patient clinical status and to echocardiography and CTPA findings.

ECG Scoring Method Used in the Study
CharacteristicsScore
  • If present check maximum only.

Tachycardia (>100 beats/min)2
Incomplete right bundle‐branch block2
Complete right bundle‐branch block3
T‐wave inversion in lead V1 through V44
T wave inversion in lead V1*
<1 mm0
12 mm1
>2 mm2
T wave inversion in lead V2*
<1 mm1
12 mm2
>2 mm3
T wave inversion in lead V3*
<1 mm1
12 mm2
>2 mm3
S wave in lead 10
Q wave in lead 31
Inverted T wave in lead 31
If all S1Q3T3, add2
Total score (maximum: 21)

Echocardiography

The study was done as soon as possible after admission using a SONOS 5500 (Hewlett Packard, Palo Alto, CA) ultrasound imaging system with a S4 transducer. In all cases M‐mode, 2‐dimensional, Doppler (continuous and pulsed wave), and color Doppler was performed with the patient in the left lateral position. The structure of the mitral, aortic, tricuspid, and pulmonary valves (different grades of regurgitation and stenosis were assessed), and the systolic and diastolic function of the left ventricle (LV) were systematically assessed.

We assessed if patients had either RVD or isolated PH. Individuals with at least 1 of the following findings were diagnosed as having RVD: right ventricle (RV) hypokinesis (asymmetrical or delayed contraction, usually in the RV base), paradoxical septal systolic motion or right ventricular dilatation (end‐diastolic diameter > 30 mm or right to left ventricular end‐diastolic diameter ratio 1 in the apical 4‐chamber view); PH was defined as pulmonary arterial systolic pressure (PASP) > 40 mm Hg.9 PASP was derived from the right atrioventricular pressure gradient, using peak velocity of tricuspid flow regurgitation with continuous wave Doppler in the apical 4‐chamber view and the modified Bernoulli equation.10 Right atrial pressure was considered equal to 5 mm Hg or 10 mm Hg, according to whether the inferior vena cava collapsed during inspiration, respectively.11

Computed Tomographic Pulmonary Angiography

All CTPAs were performed within 6 hours of patient arrival to the emergency room. Nonionic contrast (100 mL) was administered intravenously at an injection rate of 2.5 mL/second for 40 seconds. Images were obtained on the same single‐detector scanner (Somatron PQ 2000S; Picker International, Cleveland, OH). If possible, scanning was performed during a 32‐second breath hold. If patients were dyspneic, spiral CT angiography was performed during shallow breathing. The scans were obtained using 125 mA and 130 kV. A 5 mm/second table feed was used to scan a 16‐cm volume in the craniocaudal direction (pitch of 1.5). An imaging delay of 20 seconds was used, and overlapping images were reconstructed every 1.5 mm and viewed on films at lung and mediastinal settings.

PE was diagnosed by the on‐duty radiologist if an intraluminal filling defect was seen. For the purpose of the study, the images were reviewed by a senior radiologist. The pulmonary obstruction index was determined according to Qanadli et al.12 The RV to LV diameter ratio was obtained by computing the ratio between the widths of the right and the left ventricular cavities assessed on axial images obtained at the plane of maximal distance between the ventricular endocardial free wall and the interventricular septum, perpendicular to the long axis. A RV/LV ratio > 1 was accepted as suggestive of RV dysfunction. The diameter of the main pulmonary artery (PA) was measured just before the division into its 2 main branches.

Definition of Clinical Endpoints

Primary outcome measures were the prevalence of echocardiography‐assessed RVD or PH at diagnosis, the prevalence of CTPA signs of RVD, the CTPA‐measured severity of the pulmonary vascular obstruction, and the degree of hypoxemia.

Statistical Analysis

Normal distribution of data was assessed using the D'agostino‐Pearson test. Data are shown as mean standard deviation (for normally distributed data) or median (interquartile range) (for non‐normally distributed data). Comparisons between different groups were made using unpaired t test or Wilcoxon rank‐sum test, as appropriate. Correlation between the ECG score and the different variables measured was assessed using Spearman's rank correlation coefficient. Receiver operating characteristic (ROC) curves were constructed to assess the value of ECG score to predict echocardiography or CTPA‐detected RVD. Kappa measurement was performed to examine the interobserver agreement for the ECG score. A 2‐tailed value of P < 0.05 was considered significant.

Results

A total of 103 patients were included in the study (mean age = 69 15 years). One patient died from fatal PE soon after the diagnosis, before echocardiography could be performed. During the 6‐month follow‐up, one patient died from septic shock and another suffered a thromboembolic recurrence (deep vein thrombosis). Echocardiography‐assessed RVD was found in 25 cases (24.5%) and isolated pulmonary hypertension in 20 (19.6%). Median value of the vascular obstruction index was 43.8% (25‐65). Thirty‐three patients (32%) had signs of RVD in CTPA.

Median value of ECG score was 2.5 (1‐6). The level of interobserver agreement regarding ECG score was substantial ( = 0.80). Table 2 shows correlation between ECG score and echocardiography, arterial blood gases, and CTPA parameters. ECG correlated significantly with all the variables measured. Correlation was higher for the vascular obstruction index and the RV/LV ratio, measured with echocardiography. ECG scores were significantly different for groups with varying degrees of severity of PE. Table 3 shows the ECG scores for patients grouped according to different criteria of severity of the disease.

Correlation Between ECG Score and Echocardiography/CTPA Parameters
VariableR (95% CI)P
  • PASP: pulmonary artery systolic pressure; pO2 (A‐a): alveolar‐arterial gradient of oxygen. PA: pulmonary artery. RV/LV: right ventricle to left ventricle. CTPA: computed tomography pulmonary angiography.

Vascular obstruction index0.41 (0.220.57)<0.001
PASP0.31 (0.090.49)0.006
pO2(A‐a)0.29 (0.080.47)0.007
PA diameter0.28 (0.070.47)0.011
RV/LV ratio (echocardiography)0.42 (0.220.57)<0.001
RV/LV ratio (CTPA)0.36 (0.130.56)0.004
ECG Scores for Groups of Patients with Varying Severity of PE According to Different Criteria
CriteriaECG scoreP
  • RVD: right ventricle dysfunction; PH: pulmonary hypertension; CTPA: computed tomography pulmonary angiography; PaO2: arterial oxygen tension; PE: pulmonary embolism. Results are expressed as median [IQR].

RVD (echocardiography) present10 [615]<0.0001
RVD (echocardiography) absent2 [04] 
PH (echocardiography) present6 [1.7512.00]0.029
PH (echocardiography) absent2 [04] 
RVD (CTPA) present6 [311]0.0015
RVD (CTPA) absent2 [13] 
Vascular obstruction score >50%7 [211]0.0005
Vascular obstruction score 50%2 [03] 
PaO2 < 60 mmHg4 [1.252.75]0.015
PaO2 60 mmHg2 [14] 
Bilateral PE5 [111]0.028
Unilateral PE2 [0.754.00] 
Central PE4 [210]0.0011
Peripheral PE2 [03] 

Figure 1 shows the ROC curve for the ECG score and the presence of echocardiography‐detected RVD. Area under the ROC curve was 0.82 (95% confidence interval [CI]: 0.72‐0.89). A cut‐off point of 6 for the ECG score showed the highest accuracy for detecting RVD; sensitivity = 76.5% (95% CI: 50.1‐93.0), specificity = 88.6% (95% CI: 78.7‐94.9), positive likelihood ratio (+LR) = 6.69, negative likelihood ratio (LR) = 0.37. If the ECG score was to be used as a screening method to exclude RVD, a cut‐off point of 0 showed sensitivity = 94.1% (95% CI: 71.2‐99.0), specificity = 27.1% (95% CI: 17.2‐39.1), +LR = 1.29, LR = 0.22. If the ECG score was considered for confirming RVD, the more useful value was 9; sensitivity = 58.8% (95% CI: 33.0‐81.5), specificity = 92.0% (95% CI: 84.1‐97.6), +LR = 8.23, LR = 0.44. Area under the ROC curve to predict CTPA‐detected RVD was slightly lower than for echocardiography: 0.74 (95% CI: 0.61‐0.84).

Figure 1
ROC curve for the ECG score and the presence of echocardiography‐detected RVD.

Discussion

This study shows that a simple, easy‐to‐use ECG scoring system correlates well with the severity of PE, assessed by different methods, in hemodynamically stable patients. Interobserver agreement regarding the scoring system was substantial. Several previous studies have found that ECG signs of RV strain correlate with the presence of RVD.6, 1316 However, some of these studies are limited by a retrospective design that could lead to selection bias or by the inclusion of relatively few (eg, <100) cases.6, 13, 14 Moreover, only a few of these articles have specifically examined patients with normal blood pressure.6, 15 Although our study is still small, one of its strengths is that it prospectively recruited a population of consecutive, normotensive patients and both echocardiography and CTPA were systematically performed. Classifying cases with normal blood pressure by severity is particularly relevant, because of the uncertainty about best treatment when hemodynamic instability is not present. These patients who are not at high risk for PE can be further stratified according to the presence of RVD and/or myocardial injury into intermediate‐risk and low‐risk PE.17 Early hospital discharge or initial outpatient treatment might be considered in low‐risk patients.17, 18 However, echocardiography can be difficult to perform in the emergency setting. Also, the presence of RVD is not easy to predict on clinical grounds. Our study suggests that a standard 12‐lead ECG can be useful to prioritize patients for more exhaustive monitoring or additional risk stratifying tests. An ECG score of 0 would exclude RVD with enough sensitivity to avoid getting an echocardiogram in these cases. On the other hand, a score 9 would suggest the possibility that RVD was present, and an echocardiography ought to be considered in these patients.

Previous studies regarding correlation between ECG and anatomic extension of the pulmonary vascular obstruction in PE have revealed conflicting data. Iles et al. found that ECG score identified those patients with the greatest percentage of perfusion defect on ventilation/perfusion scan.19 However, Kanbay et al. did not find significant differences in ECG scores for patients with pulmonary vascular obstruction scores <50% or 50%, also assessed with ventilation/perfusion scan.20 Subramaniam et al. used CTPA to assess clot burden score, and they did not see a significant association between ECG score and the anatomic severity of PE.21 This study found a significant association between higher ECG score and a more severe vascular obstruction index. This finding might have potential clinical implications, because some reports suggest that a higher CTPA‐measured clot load score can be associated with poorer outcome in PE.22 However, the utility of CTPA scores of vascular obstruction severity as a marker of clinical evolution is controversial, and several other authors have found that these scores are poor predictors of mortality or adverse outcome events.23, 24 Therefore, the positive correlation between ECG and the vascular obstruction index in our study is interesting mainly in the sense that it independently supports the validity of the ECG score as a marker of a potentially more severe PE.

In this study, we have not systematically excluded patients with comorbidities (eg, chronic obstructive pulmonary disease) that could account for RVD or PH. Our intention was to reflect a real‐life clinical scenario. When a patient attends the emergency room with PE, clinical decisions must usually be taken without complete information on comorbidities that may contribute to the existence of RV overload. Also, these subjects have a reduced cardiorespiratory reserve, and the PE might impose an unbearable strain to the RV, so risk stratification is especially important. Therefore, we felt that the study should include all patients presenting with PE, so that the results can be extrapolated to usual clinical practice.

Our results suggest that, by selecting the appropriate cut‐off value, the ECG score can be used with adequate sensitivity and specificity to exclude and confirm, respectively, RVD in hemodynamically stable PE patients. This finding implies that ECG may be useful to design management strategies in these cases, selecting patients that may benefit from further tests.

There are some limitations for the present study. Echocardiography was not immediately performed after the ECG. Thus, we might have missed some patients who presented with transient RVD, which resolved between tests. It is likely that the correlation between ECG and echocardiography would have improved if this delay had been avoided. Also, we did not use a multidetector scanner for performing CTPAs. Consequently, small emboli might have been missed and the clot score might have been underestimated. It is plausible that this may have reduced to some extent the correlation between ECG and CTPA. Also, this a single‐center study, with relatively few patients studied. Finally, due to the low incidence of adverse events in our patients, our study is underpowered to make conclusions about the independent prognostic value of ECG for predicting adverse events.

In conclusion, an easy‐to‐use ECG score correlates significantly with the severity of PE, as assessed with echocardiography, CTPA, and arterial blood gas analysis, in normotensive patients. It can be used to predict with acceptable sensitivity and specificity the presence of RVD in these cases and, accordingly, it has potential value in risk‐stratification strategies. Larger, multicenter studies, should confirm these results before they can be applied to clinical practice.

References
  1. Goldhaber SZ,Visani L,De Rosa M.Acute pulmonary embolism: clinical outcomes in the International Cooperative Pulmonary Embolism Registry (ICOPER).Lancet.1999;353:13861389.
  2. Wan S,Quinlan DJ,Agnelli G,Eikelboom JW.Thrombolysis compared with heparin for the initial treatment of pulmonary embolism. A meta‐analysis of the randomized controlled trials.Circulation.2004;110:744749.
  3. Kasper W,Konstantinides S,Geibel A, et al.Management strategies and determinants of outcome in acute major pulmonary embolism: results of a multicenter registry.J Am Coll Cardiol.1997;30:11651171
  4. Ten Wolde M,Söhne M,Quak E,Mac Gillavry MR,Büller HR.Prognostic value of echocardiographycally assessed right ventricular dysfunction in patients with pulmonary embolism.Arch Intern Med.2004;164:16851689.
  5. Schoepf UJ,Kucher N,Kipfmueller F, et al.Right ventricular enlargement on chest computed tomography. A predictor of early death in acute pulmonary embolism.Circulation.2004;110:32763280.
  6. Punukollu G,Gowda RM,Vasavada BC,Khan IA.Role of electrocardiography in identifying right ventricular dysfunction in acute pulmonary embolism.Am J Cardiol.2005;96:450452.
  7. Golpe R,Pérez‐de‐Llano LA,Castro‐Añón O, et al.Right ventricle dysfunction and pulmonary hypertension in hemodynamically stable pulmonary embolism.Respir Med.2010;104:13701376.
  8. Daniel KR,Courtney M,Kline JA.Assessment of cardiac stress from massive pulmonary embolism with 12‐lead ECG.Chest.2001;120:474481.
  9. McQuillan BM,Picard MH,Leavitt M, et al.Clinical correlates and reference intervals for pulmonary artery systolic pressure among echocardiographically normal subjects.Circulation.2001;104:27972802.
  10. Berger M,Haimowitz A,Van Tosh A, et al.Quantitative assessment of pulmonary hypertension in patients with tricuspid regurgitation using continuous wave Doppler ultrasound.J Am Coll Cardiol.1985;6:359365.
  11. Kircher BJ,Himelman RB,Schiller NB.Noninvasive estimation of right atrial pressure from the inspiratory collapse of the inferior vena cava.Am J Cardiol.1990;66:493496.
  12. Qanadli SD,El Hajjam M,Vieillard‐Baron A, et al.New CT index to quantify arterial obstruction in pulmonary embolism: comparison with angiographic index and echocardiography.AJR Am J Roentgenol.2001;176:14151420.
  13. Toosi MS,Merlino JD,Leeper KV.Electrocardiographic score and short‐term outcomes of acute pulmonary embolism.Am J Cardiol.2007;100:11721176.
  14. Kostrubiec M,Hrynkiewicz A,Pedowska‐Wtoszek J, et al.Is it possible to use standard electrocardiography for risk assessment of patients with pulmonary embolism?Kardiol Pol.2009;67:744750.
  15. Vanni S,Polidori G,Vergara R, et al.Prognostic value of ECG among patients with acute pulmonary embolism and normal blood pressure.Am J Med.2009;122:257264.
  16. Kucher N,Walpoth N,Wustmann K,Noveanu M,Gertsch M.QR in V1‐an ECG sign associated with right ventricular strain and adverse clinical outcome in pulmonary embolism.Eur Heart J.2003;24:11131119.
  17. Torbicki A,Perrier A,Konstantinides S, et al.Guidelines on the diagnosis and management of acute pulmonary embolism. The Task Force for the Diagnosis and Management of Acute Pulmonary Embolism of the European Society of Cardiology (ESC).Eur Heart J.2008;29:22762315.
  18. Konstantinides S.Acute pulmonary embolism.N Eng J Med.2008;359:28042813.
  19. Iles S,Le Heron CJ,Davies G,Turner JG,Beckert LEL.ECG score predicts those with the greatest percentage of perfusion defects due to acute pulmonary thromboembolic disease.Chest.2004;125:16511656.
  20. Kanbay A,Kokturk N,Kaya MG, et al.Electrocardiography and Wells scoring in predicting the anatomic severity of pulmonary embolism.Respir Med.2007;101:11711176.
  21. Subramaniam RM,Mandrekar J,Chang C, et al.Pulmonary embolism outcome: a prospective evaluation of CT pulmonary angiographic clot burden score and ECG score.AJR Am J Roentgenol.2008;190:15991604.
  22. Van der Meer RW,Pattynama PMT,Van Strijen MJL, et al.Right ventricular dysfunction and pulmonary obstruction index at helical CT: prediction of clinical outcome during 3‐month follow‐up in patients with acute pulmonary embolism.Radiology.2005;235:798803.
  23. Ghuysen AE,Ghaye B,Villems V,Dondelinger RF,D'Orio V.Acute PE mortality prediction by analysis of helical computed tomography angiography and hemodynamic evaluation.Thorax.2005;60:956961.
  24. Araoz PA,Gotway MB,Harrington JR,Harmsen WS,Mandrekar JN.Pulmonary embolism: prognostic CT findings.Radiology.2007;242:889893.
Article PDF
Issue
Journal of Hospital Medicine - 6(5)
Page Number
285-289
Legacy Keywords
electrocardiography, pulmonary embolism, right ventricular function, computer tomography, echocardiography
Sections
Article PDF
Article PDF

The clinical outcome of patients with pulmonary embolism (PE) has a wide spectrum, ranging from a benign course, with early resolution of symptoms, to death. Prognostic stratification of patients with PE is therefore essential to guide therapy. Currently, this stratification is primarily based on blood pressure on admission. Hemodynamic instability or shock carries a bad prognosis, and is associated with a 3‐fold to 7‐fold increase in mortality.1 Thrombolytic therapy is warranted in these cases.2 There is a subset of patients that, despite normal blood pressure on presentation, can subsequently suffer hemodynamic deterioration and death.3

Echocardiography has been proposed as a useful tool to identify a subset of patients at a higher risk. It has been suggested that the finding of right ventricle dysfunction (RVD) in echocardiography predicts a worse prognosis, even in patients with normal blood pressure on presentation.4 Also, computed tomography pulmonary angiography (CTPA) is increasingly used as the main test to diagnose PE. This technique can also assess the degree of pulmonary arterial obstruction and the presence of signs suggestive of RVD. It has been suggested that CTPA findings can predict outcome in PE.5

Electrocardiography (ECG) has a widespread use and is routinely carried out and interpreted in the emergency room. Some ECG findings in patients with PE are suggestive of right ventricular strain and might therefore be used to prioritize cases that require closer monitoring.6

This study was designed with the objectives of assessing the correlation of standard ECG findings with echocardiography and CTPA signs of RVD, with the anatomic severity of the pulmonary vascular obstruction, and with the degree of hypoxemia, in hemodynamically stable patients with PE. Secondary objectives were determining if ECG could accurately predict RVD, and determining the interobserver agreement for the ECG score.

Patients and Methods

Patients and Study Design

We recently carried out a prospective, descriptive, single‐center follow‐up study to assess the prevalence of RVD and pulmonary hypertension (PH) in PE patients who presented with hemodynamic stability.7 We herein analyze ECG findings in these cases. The present, noninterventional study was approved by our institution's review board. The population studied included consecutive patients presenting with PE to our emergency room. Inclusion criteria were age >18 years and PE confirmed by CTPA. Exclusion criteria were high‐risk PE (presence of 2 consecutive systolic blood pressure measurements <90 mm Hg, measured >15 minutes apart or requiring inotropic support); comorbidity predictive of a 6‐month mortality >50% (eg, metastatic cancer or end‐stage respiratory or heart disease); creatinine clearance <35 mL/minute; or allergy to ionidated contrast media. All the patients gave informed consent to participate in the study.

Protocol

Initial assessment included clinical history, physical examination, ECG, arterial blood gas analysis, echocardiography, CTPA, and lower‐limb venous ultrasonography. All included individuals were treated with anticoagulation, and fibrinolytic therapy was not used.

ECG

A standard 12‐lead ECG was promptly obtained for all patients in the emergency room. We used the scoring system described by Daniel et al.8 (Table 1). This score is based on 4 signs that were previously found to be associated with high‐risk PE: tachycardia, incomplete or complete right bundle branch block, T wave inversion, and S1Q3T3 pattern. Two investigators trained in the interpretation of ECG results scored the ECG independently, and were blinded to patient clinical status and to echocardiography and CTPA findings.

ECG Scoring Method Used in the Study
CharacteristicsScore
  • If present check maximum only.

Tachycardia (>100 beats/min)2
Incomplete right bundle‐branch block2
Complete right bundle‐branch block3
T‐wave inversion in lead V1 through V44
T wave inversion in lead V1*
<1 mm0
12 mm1
>2 mm2
T wave inversion in lead V2*
<1 mm1
12 mm2
>2 mm3
T wave inversion in lead V3*
<1 mm1
12 mm2
>2 mm3
S wave in lead 10
Q wave in lead 31
Inverted T wave in lead 31
If all S1Q3T3, add2
Total score (maximum: 21)

Echocardiography

The study was done as soon as possible after admission using a SONOS 5500 (Hewlett Packard, Palo Alto, CA) ultrasound imaging system with a S4 transducer. In all cases M‐mode, 2‐dimensional, Doppler (continuous and pulsed wave), and color Doppler was performed with the patient in the left lateral position. The structure of the mitral, aortic, tricuspid, and pulmonary valves (different grades of regurgitation and stenosis were assessed), and the systolic and diastolic function of the left ventricle (LV) were systematically assessed.

We assessed if patients had either RVD or isolated PH. Individuals with at least 1 of the following findings were diagnosed as having RVD: right ventricle (RV) hypokinesis (asymmetrical or delayed contraction, usually in the RV base), paradoxical septal systolic motion or right ventricular dilatation (end‐diastolic diameter > 30 mm or right to left ventricular end‐diastolic diameter ratio 1 in the apical 4‐chamber view); PH was defined as pulmonary arterial systolic pressure (PASP) > 40 mm Hg.9 PASP was derived from the right atrioventricular pressure gradient, using peak velocity of tricuspid flow regurgitation with continuous wave Doppler in the apical 4‐chamber view and the modified Bernoulli equation.10 Right atrial pressure was considered equal to 5 mm Hg or 10 mm Hg, according to whether the inferior vena cava collapsed during inspiration, respectively.11

Computed Tomographic Pulmonary Angiography

All CTPAs were performed within 6 hours of patient arrival to the emergency room. Nonionic contrast (100 mL) was administered intravenously at an injection rate of 2.5 mL/second for 40 seconds. Images were obtained on the same single‐detector scanner (Somatron PQ 2000S; Picker International, Cleveland, OH). If possible, scanning was performed during a 32‐second breath hold. If patients were dyspneic, spiral CT angiography was performed during shallow breathing. The scans were obtained using 125 mA and 130 kV. A 5 mm/second table feed was used to scan a 16‐cm volume in the craniocaudal direction (pitch of 1.5). An imaging delay of 20 seconds was used, and overlapping images were reconstructed every 1.5 mm and viewed on films at lung and mediastinal settings.

PE was diagnosed by the on‐duty radiologist if an intraluminal filling defect was seen. For the purpose of the study, the images were reviewed by a senior radiologist. The pulmonary obstruction index was determined according to Qanadli et al.12 The RV to LV diameter ratio was obtained by computing the ratio between the widths of the right and the left ventricular cavities assessed on axial images obtained at the plane of maximal distance between the ventricular endocardial free wall and the interventricular septum, perpendicular to the long axis. A RV/LV ratio > 1 was accepted as suggestive of RV dysfunction. The diameter of the main pulmonary artery (PA) was measured just before the division into its 2 main branches.

Definition of Clinical Endpoints

Primary outcome measures were the prevalence of echocardiography‐assessed RVD or PH at diagnosis, the prevalence of CTPA signs of RVD, the CTPA‐measured severity of the pulmonary vascular obstruction, and the degree of hypoxemia.

Statistical Analysis

Normal distribution of data was assessed using the D'agostino‐Pearson test. Data are shown as mean standard deviation (for normally distributed data) or median (interquartile range) (for non‐normally distributed data). Comparisons between different groups were made using unpaired t test or Wilcoxon rank‐sum test, as appropriate. Correlation between the ECG score and the different variables measured was assessed using Spearman's rank correlation coefficient. Receiver operating characteristic (ROC) curves were constructed to assess the value of ECG score to predict echocardiography or CTPA‐detected RVD. Kappa measurement was performed to examine the interobserver agreement for the ECG score. A 2‐tailed value of P < 0.05 was considered significant.

Results

A total of 103 patients were included in the study (mean age = 69 15 years). One patient died from fatal PE soon after the diagnosis, before echocardiography could be performed. During the 6‐month follow‐up, one patient died from septic shock and another suffered a thromboembolic recurrence (deep vein thrombosis). Echocardiography‐assessed RVD was found in 25 cases (24.5%) and isolated pulmonary hypertension in 20 (19.6%). Median value of the vascular obstruction index was 43.8% (25‐65). Thirty‐three patients (32%) had signs of RVD in CTPA.

Median value of ECG score was 2.5 (1‐6). The level of interobserver agreement regarding ECG score was substantial ( = 0.80). Table 2 shows correlation between ECG score and echocardiography, arterial blood gases, and CTPA parameters. ECG correlated significantly with all the variables measured. Correlation was higher for the vascular obstruction index and the RV/LV ratio, measured with echocardiography. ECG scores were significantly different for groups with varying degrees of severity of PE. Table 3 shows the ECG scores for patients grouped according to different criteria of severity of the disease.

Correlation Between ECG Score and Echocardiography/CTPA Parameters
VariableR (95% CI)P
  • PASP: pulmonary artery systolic pressure; pO2 (A‐a): alveolar‐arterial gradient of oxygen. PA: pulmonary artery. RV/LV: right ventricle to left ventricle. CTPA: computed tomography pulmonary angiography.

Vascular obstruction index0.41 (0.220.57)<0.001
PASP0.31 (0.090.49)0.006
pO2(A‐a)0.29 (0.080.47)0.007
PA diameter0.28 (0.070.47)0.011
RV/LV ratio (echocardiography)0.42 (0.220.57)<0.001
RV/LV ratio (CTPA)0.36 (0.130.56)0.004
ECG Scores for Groups of Patients with Varying Severity of PE According to Different Criteria
CriteriaECG scoreP
  • RVD: right ventricle dysfunction; PH: pulmonary hypertension; CTPA: computed tomography pulmonary angiography; PaO2: arterial oxygen tension; PE: pulmonary embolism. Results are expressed as median [IQR].

RVD (echocardiography) present10 [615]<0.0001
RVD (echocardiography) absent2 [04] 
PH (echocardiography) present6 [1.7512.00]0.029
PH (echocardiography) absent2 [04] 
RVD (CTPA) present6 [311]0.0015
RVD (CTPA) absent2 [13] 
Vascular obstruction score >50%7 [211]0.0005
Vascular obstruction score 50%2 [03] 
PaO2 < 60 mmHg4 [1.252.75]0.015
PaO2 60 mmHg2 [14] 
Bilateral PE5 [111]0.028
Unilateral PE2 [0.754.00] 
Central PE4 [210]0.0011
Peripheral PE2 [03] 

Figure 1 shows the ROC curve for the ECG score and the presence of echocardiography‐detected RVD. Area under the ROC curve was 0.82 (95% confidence interval [CI]: 0.72‐0.89). A cut‐off point of 6 for the ECG score showed the highest accuracy for detecting RVD; sensitivity = 76.5% (95% CI: 50.1‐93.0), specificity = 88.6% (95% CI: 78.7‐94.9), positive likelihood ratio (+LR) = 6.69, negative likelihood ratio (LR) = 0.37. If the ECG score was to be used as a screening method to exclude RVD, a cut‐off point of 0 showed sensitivity = 94.1% (95% CI: 71.2‐99.0), specificity = 27.1% (95% CI: 17.2‐39.1), +LR = 1.29, LR = 0.22. If the ECG score was considered for confirming RVD, the more useful value was 9; sensitivity = 58.8% (95% CI: 33.0‐81.5), specificity = 92.0% (95% CI: 84.1‐97.6), +LR = 8.23, LR = 0.44. Area under the ROC curve to predict CTPA‐detected RVD was slightly lower than for echocardiography: 0.74 (95% CI: 0.61‐0.84).

Figure 1
ROC curve for the ECG score and the presence of echocardiography‐detected RVD.

Discussion

This study shows that a simple, easy‐to‐use ECG scoring system correlates well with the severity of PE, assessed by different methods, in hemodynamically stable patients. Interobserver agreement regarding the scoring system was substantial. Several previous studies have found that ECG signs of RV strain correlate with the presence of RVD.6, 1316 However, some of these studies are limited by a retrospective design that could lead to selection bias or by the inclusion of relatively few (eg, <100) cases.6, 13, 14 Moreover, only a few of these articles have specifically examined patients with normal blood pressure.6, 15 Although our study is still small, one of its strengths is that it prospectively recruited a population of consecutive, normotensive patients and both echocardiography and CTPA were systematically performed. Classifying cases with normal blood pressure by severity is particularly relevant, because of the uncertainty about best treatment when hemodynamic instability is not present. These patients who are not at high risk for PE can be further stratified according to the presence of RVD and/or myocardial injury into intermediate‐risk and low‐risk PE.17 Early hospital discharge or initial outpatient treatment might be considered in low‐risk patients.17, 18 However, echocardiography can be difficult to perform in the emergency setting. Also, the presence of RVD is not easy to predict on clinical grounds. Our study suggests that a standard 12‐lead ECG can be useful to prioritize patients for more exhaustive monitoring or additional risk stratifying tests. An ECG score of 0 would exclude RVD with enough sensitivity to avoid getting an echocardiogram in these cases. On the other hand, a score 9 would suggest the possibility that RVD was present, and an echocardiography ought to be considered in these patients.

Previous studies regarding correlation between ECG and anatomic extension of the pulmonary vascular obstruction in PE have revealed conflicting data. Iles et al. found that ECG score identified those patients with the greatest percentage of perfusion defect on ventilation/perfusion scan.19 However, Kanbay et al. did not find significant differences in ECG scores for patients with pulmonary vascular obstruction scores <50% or 50%, also assessed with ventilation/perfusion scan.20 Subramaniam et al. used CTPA to assess clot burden score, and they did not see a significant association between ECG score and the anatomic severity of PE.21 This study found a significant association between higher ECG score and a more severe vascular obstruction index. This finding might have potential clinical implications, because some reports suggest that a higher CTPA‐measured clot load score can be associated with poorer outcome in PE.22 However, the utility of CTPA scores of vascular obstruction severity as a marker of clinical evolution is controversial, and several other authors have found that these scores are poor predictors of mortality or adverse outcome events.23, 24 Therefore, the positive correlation between ECG and the vascular obstruction index in our study is interesting mainly in the sense that it independently supports the validity of the ECG score as a marker of a potentially more severe PE.

In this study, we have not systematically excluded patients with comorbidities (eg, chronic obstructive pulmonary disease) that could account for RVD or PH. Our intention was to reflect a real‐life clinical scenario. When a patient attends the emergency room with PE, clinical decisions must usually be taken without complete information on comorbidities that may contribute to the existence of RV overload. Also, these subjects have a reduced cardiorespiratory reserve, and the PE might impose an unbearable strain to the RV, so risk stratification is especially important. Therefore, we felt that the study should include all patients presenting with PE, so that the results can be extrapolated to usual clinical practice.

Our results suggest that, by selecting the appropriate cut‐off value, the ECG score can be used with adequate sensitivity and specificity to exclude and confirm, respectively, RVD in hemodynamically stable PE patients. This finding implies that ECG may be useful to design management strategies in these cases, selecting patients that may benefit from further tests.

There are some limitations for the present study. Echocardiography was not immediately performed after the ECG. Thus, we might have missed some patients who presented with transient RVD, which resolved between tests. It is likely that the correlation between ECG and echocardiography would have improved if this delay had been avoided. Also, we did not use a multidetector scanner for performing CTPAs. Consequently, small emboli might have been missed and the clot score might have been underestimated. It is plausible that this may have reduced to some extent the correlation between ECG and CTPA. Also, this a single‐center study, with relatively few patients studied. Finally, due to the low incidence of adverse events in our patients, our study is underpowered to make conclusions about the independent prognostic value of ECG for predicting adverse events.

In conclusion, an easy‐to‐use ECG score correlates significantly with the severity of PE, as assessed with echocardiography, CTPA, and arterial blood gas analysis, in normotensive patients. It can be used to predict with acceptable sensitivity and specificity the presence of RVD in these cases and, accordingly, it has potential value in risk‐stratification strategies. Larger, multicenter studies, should confirm these results before they can be applied to clinical practice.

The clinical outcome of patients with pulmonary embolism (PE) has a wide spectrum, ranging from a benign course, with early resolution of symptoms, to death. Prognostic stratification of patients with PE is therefore essential to guide therapy. Currently, this stratification is primarily based on blood pressure on admission. Hemodynamic instability or shock carries a bad prognosis, and is associated with a 3‐fold to 7‐fold increase in mortality.1 Thrombolytic therapy is warranted in these cases.2 There is a subset of patients that, despite normal blood pressure on presentation, can subsequently suffer hemodynamic deterioration and death.3

Echocardiography has been proposed as a useful tool to identify a subset of patients at a higher risk. It has been suggested that the finding of right ventricle dysfunction (RVD) in echocardiography predicts a worse prognosis, even in patients with normal blood pressure on presentation.4 Also, computed tomography pulmonary angiography (CTPA) is increasingly used as the main test to diagnose PE. This technique can also assess the degree of pulmonary arterial obstruction and the presence of signs suggestive of RVD. It has been suggested that CTPA findings can predict outcome in PE.5

Electrocardiography (ECG) has a widespread use and is routinely carried out and interpreted in the emergency room. Some ECG findings in patients with PE are suggestive of right ventricular strain and might therefore be used to prioritize cases that require closer monitoring.6

This study was designed with the objectives of assessing the correlation of standard ECG findings with echocardiography and CTPA signs of RVD, with the anatomic severity of the pulmonary vascular obstruction, and with the degree of hypoxemia, in hemodynamically stable patients with PE. Secondary objectives were determining if ECG could accurately predict RVD, and determining the interobserver agreement for the ECG score.

Patients and Methods

Patients and Study Design

We recently carried out a prospective, descriptive, single‐center follow‐up study to assess the prevalence of RVD and pulmonary hypertension (PH) in PE patients who presented with hemodynamic stability.7 We herein analyze ECG findings in these cases. The present, noninterventional study was approved by our institution's review board. The population studied included consecutive patients presenting with PE to our emergency room. Inclusion criteria were age >18 years and PE confirmed by CTPA. Exclusion criteria were high‐risk PE (presence of 2 consecutive systolic blood pressure measurements <90 mm Hg, measured >15 minutes apart or requiring inotropic support); comorbidity predictive of a 6‐month mortality >50% (eg, metastatic cancer or end‐stage respiratory or heart disease); creatinine clearance <35 mL/minute; or allergy to ionidated contrast media. All the patients gave informed consent to participate in the study.

Protocol

Initial assessment included clinical history, physical examination, ECG, arterial blood gas analysis, echocardiography, CTPA, and lower‐limb venous ultrasonography. All included individuals were treated with anticoagulation, and fibrinolytic therapy was not used.

ECG

A standard 12‐lead ECG was promptly obtained for all patients in the emergency room. We used the scoring system described by Daniel et al.8 (Table 1). This score is based on 4 signs that were previously found to be associated with high‐risk PE: tachycardia, incomplete or complete right bundle branch block, T wave inversion, and S1Q3T3 pattern. Two investigators trained in the interpretation of ECG results scored the ECG independently, and were blinded to patient clinical status and to echocardiography and CTPA findings.

ECG Scoring Method Used in the Study
CharacteristicsScore
  • If present check maximum only.

Tachycardia (>100 beats/min)2
Incomplete right bundle‐branch block2
Complete right bundle‐branch block3
T‐wave inversion in lead V1 through V44
T wave inversion in lead V1*
<1 mm0
12 mm1
>2 mm2
T wave inversion in lead V2*
<1 mm1
12 mm2
>2 mm3
T wave inversion in lead V3*
<1 mm1
12 mm2
>2 mm3
S wave in lead 10
Q wave in lead 31
Inverted T wave in lead 31
If all S1Q3T3, add2
Total score (maximum: 21)

Echocardiography

The study was done as soon as possible after admission using a SONOS 5500 (Hewlett Packard, Palo Alto, CA) ultrasound imaging system with a S4 transducer. In all cases M‐mode, 2‐dimensional, Doppler (continuous and pulsed wave), and color Doppler was performed with the patient in the left lateral position. The structure of the mitral, aortic, tricuspid, and pulmonary valves (different grades of regurgitation and stenosis were assessed), and the systolic and diastolic function of the left ventricle (LV) were systematically assessed.

We assessed if patients had either RVD or isolated PH. Individuals with at least 1 of the following findings were diagnosed as having RVD: right ventricle (RV) hypokinesis (asymmetrical or delayed contraction, usually in the RV base), paradoxical septal systolic motion or right ventricular dilatation (end‐diastolic diameter > 30 mm or right to left ventricular end‐diastolic diameter ratio 1 in the apical 4‐chamber view); PH was defined as pulmonary arterial systolic pressure (PASP) > 40 mm Hg.9 PASP was derived from the right atrioventricular pressure gradient, using peak velocity of tricuspid flow regurgitation with continuous wave Doppler in the apical 4‐chamber view and the modified Bernoulli equation.10 Right atrial pressure was considered equal to 5 mm Hg or 10 mm Hg, according to whether the inferior vena cava collapsed during inspiration, respectively.11

Computed Tomographic Pulmonary Angiography

All CTPAs were performed within 6 hours of patient arrival to the emergency room. Nonionic contrast (100 mL) was administered intravenously at an injection rate of 2.5 mL/second for 40 seconds. Images were obtained on the same single‐detector scanner (Somatron PQ 2000S; Picker International, Cleveland, OH). If possible, scanning was performed during a 32‐second breath hold. If patients were dyspneic, spiral CT angiography was performed during shallow breathing. The scans were obtained using 125 mA and 130 kV. A 5 mm/second table feed was used to scan a 16‐cm volume in the craniocaudal direction (pitch of 1.5). An imaging delay of 20 seconds was used, and overlapping images were reconstructed every 1.5 mm and viewed on films at lung and mediastinal settings.

PE was diagnosed by the on‐duty radiologist if an intraluminal filling defect was seen. For the purpose of the study, the images were reviewed by a senior radiologist. The pulmonary obstruction index was determined according to Qanadli et al.12 The RV to LV diameter ratio was obtained by computing the ratio between the widths of the right and the left ventricular cavities assessed on axial images obtained at the plane of maximal distance between the ventricular endocardial free wall and the interventricular septum, perpendicular to the long axis. A RV/LV ratio > 1 was accepted as suggestive of RV dysfunction. The diameter of the main pulmonary artery (PA) was measured just before the division into its 2 main branches.

Definition of Clinical Endpoints

Primary outcome measures were the prevalence of echocardiography‐assessed RVD or PH at diagnosis, the prevalence of CTPA signs of RVD, the CTPA‐measured severity of the pulmonary vascular obstruction, and the degree of hypoxemia.

Statistical Analysis

Normal distribution of data was assessed using the D'agostino‐Pearson test. Data are shown as mean standard deviation (for normally distributed data) or median (interquartile range) (for non‐normally distributed data). Comparisons between different groups were made using unpaired t test or Wilcoxon rank‐sum test, as appropriate. Correlation between the ECG score and the different variables measured was assessed using Spearman's rank correlation coefficient. Receiver operating characteristic (ROC) curves were constructed to assess the value of ECG score to predict echocardiography or CTPA‐detected RVD. Kappa measurement was performed to examine the interobserver agreement for the ECG score. A 2‐tailed value of P < 0.05 was considered significant.

Results

A total of 103 patients were included in the study (mean age = 69 15 years). One patient died from fatal PE soon after the diagnosis, before echocardiography could be performed. During the 6‐month follow‐up, one patient died from septic shock and another suffered a thromboembolic recurrence (deep vein thrombosis). Echocardiography‐assessed RVD was found in 25 cases (24.5%) and isolated pulmonary hypertension in 20 (19.6%). Median value of the vascular obstruction index was 43.8% (25‐65). Thirty‐three patients (32%) had signs of RVD in CTPA.

Median value of ECG score was 2.5 (1‐6). The level of interobserver agreement regarding ECG score was substantial ( = 0.80). Table 2 shows correlation between ECG score and echocardiography, arterial blood gases, and CTPA parameters. ECG correlated significantly with all the variables measured. Correlation was higher for the vascular obstruction index and the RV/LV ratio, measured with echocardiography. ECG scores were significantly different for groups with varying degrees of severity of PE. Table 3 shows the ECG scores for patients grouped according to different criteria of severity of the disease.

Correlation Between ECG Score and Echocardiography/CTPA Parameters
VariableR (95% CI)P
  • PASP: pulmonary artery systolic pressure; pO2 (A‐a): alveolar‐arterial gradient of oxygen. PA: pulmonary artery. RV/LV: right ventricle to left ventricle. CTPA: computed tomography pulmonary angiography.

Vascular obstruction index0.41 (0.220.57)<0.001
PASP0.31 (0.090.49)0.006
pO2(A‐a)0.29 (0.080.47)0.007
PA diameter0.28 (0.070.47)0.011
RV/LV ratio (echocardiography)0.42 (0.220.57)<0.001
RV/LV ratio (CTPA)0.36 (0.130.56)0.004
ECG Scores for Groups of Patients with Varying Severity of PE According to Different Criteria
CriteriaECG scoreP
  • RVD: right ventricle dysfunction; PH: pulmonary hypertension; CTPA: computed tomography pulmonary angiography; PaO2: arterial oxygen tension; PE: pulmonary embolism. Results are expressed as median [IQR].

RVD (echocardiography) present10 [615]<0.0001
RVD (echocardiography) absent2 [04] 
PH (echocardiography) present6 [1.7512.00]0.029
PH (echocardiography) absent2 [04] 
RVD (CTPA) present6 [311]0.0015
RVD (CTPA) absent2 [13] 
Vascular obstruction score >50%7 [211]0.0005
Vascular obstruction score 50%2 [03] 
PaO2 < 60 mmHg4 [1.252.75]0.015
PaO2 60 mmHg2 [14] 
Bilateral PE5 [111]0.028
Unilateral PE2 [0.754.00] 
Central PE4 [210]0.0011
Peripheral PE2 [03] 

Figure 1 shows the ROC curve for the ECG score and the presence of echocardiography‐detected RVD. Area under the ROC curve was 0.82 (95% confidence interval [CI]: 0.72‐0.89). A cut‐off point of 6 for the ECG score showed the highest accuracy for detecting RVD; sensitivity = 76.5% (95% CI: 50.1‐93.0), specificity = 88.6% (95% CI: 78.7‐94.9), positive likelihood ratio (+LR) = 6.69, negative likelihood ratio (LR) = 0.37. If the ECG score was to be used as a screening method to exclude RVD, a cut‐off point of 0 showed sensitivity = 94.1% (95% CI: 71.2‐99.0), specificity = 27.1% (95% CI: 17.2‐39.1), +LR = 1.29, LR = 0.22. If the ECG score was considered for confirming RVD, the more useful value was 9; sensitivity = 58.8% (95% CI: 33.0‐81.5), specificity = 92.0% (95% CI: 84.1‐97.6), +LR = 8.23, LR = 0.44. Area under the ROC curve to predict CTPA‐detected RVD was slightly lower than for echocardiography: 0.74 (95% CI: 0.61‐0.84).

Figure 1
ROC curve for the ECG score and the presence of echocardiography‐detected RVD.

Discussion

This study shows that a simple, easy‐to‐use ECG scoring system correlates well with the severity of PE, assessed by different methods, in hemodynamically stable patients. Interobserver agreement regarding the scoring system was substantial. Several previous studies have found that ECG signs of RV strain correlate with the presence of RVD.6, 1316 However, some of these studies are limited by a retrospective design that could lead to selection bias or by the inclusion of relatively few (eg, <100) cases.6, 13, 14 Moreover, only a few of these articles have specifically examined patients with normal blood pressure.6, 15 Although our study is still small, one of its strengths is that it prospectively recruited a population of consecutive, normotensive patients and both echocardiography and CTPA were systematically performed. Classifying cases with normal blood pressure by severity is particularly relevant, because of the uncertainty about best treatment when hemodynamic instability is not present. These patients who are not at high risk for PE can be further stratified according to the presence of RVD and/or myocardial injury into intermediate‐risk and low‐risk PE.17 Early hospital discharge or initial outpatient treatment might be considered in low‐risk patients.17, 18 However, echocardiography can be difficult to perform in the emergency setting. Also, the presence of RVD is not easy to predict on clinical grounds. Our study suggests that a standard 12‐lead ECG can be useful to prioritize patients for more exhaustive monitoring or additional risk stratifying tests. An ECG score of 0 would exclude RVD with enough sensitivity to avoid getting an echocardiogram in these cases. On the other hand, a score 9 would suggest the possibility that RVD was present, and an echocardiography ought to be considered in these patients.

Previous studies regarding correlation between ECG and anatomic extension of the pulmonary vascular obstruction in PE have revealed conflicting data. Iles et al. found that ECG score identified those patients with the greatest percentage of perfusion defect on ventilation/perfusion scan.19 However, Kanbay et al. did not find significant differences in ECG scores for patients with pulmonary vascular obstruction scores <50% or 50%, also assessed with ventilation/perfusion scan.20 Subramaniam et al. used CTPA to assess clot burden score, and they did not see a significant association between ECG score and the anatomic severity of PE.21 This study found a significant association between higher ECG score and a more severe vascular obstruction index. This finding might have potential clinical implications, because some reports suggest that a higher CTPA‐measured clot load score can be associated with poorer outcome in PE.22 However, the utility of CTPA scores of vascular obstruction severity as a marker of clinical evolution is controversial, and several other authors have found that these scores are poor predictors of mortality or adverse outcome events.23, 24 Therefore, the positive correlation between ECG and the vascular obstruction index in our study is interesting mainly in the sense that it independently supports the validity of the ECG score as a marker of a potentially more severe PE.

In this study, we have not systematically excluded patients with comorbidities (eg, chronic obstructive pulmonary disease) that could account for RVD or PH. Our intention was to reflect a real‐life clinical scenario. When a patient attends the emergency room with PE, clinical decisions must usually be taken without complete information on comorbidities that may contribute to the existence of RV overload. Also, these subjects have a reduced cardiorespiratory reserve, and the PE might impose an unbearable strain to the RV, so risk stratification is especially important. Therefore, we felt that the study should include all patients presenting with PE, so that the results can be extrapolated to usual clinical practice.

Our results suggest that, by selecting the appropriate cut‐off value, the ECG score can be used with adequate sensitivity and specificity to exclude and confirm, respectively, RVD in hemodynamically stable PE patients. This finding implies that ECG may be useful to design management strategies in these cases, selecting patients that may benefit from further tests.

There are some limitations for the present study. Echocardiography was not immediately performed after the ECG. Thus, we might have missed some patients who presented with transient RVD, which resolved between tests. It is likely that the correlation between ECG and echocardiography would have improved if this delay had been avoided. Also, we did not use a multidetector scanner for performing CTPAs. Consequently, small emboli might have been missed and the clot score might have been underestimated. It is plausible that this may have reduced to some extent the correlation between ECG and CTPA. Also, this a single‐center study, with relatively few patients studied. Finally, due to the low incidence of adverse events in our patients, our study is underpowered to make conclusions about the independent prognostic value of ECG for predicting adverse events.

In conclusion, an easy‐to‐use ECG score correlates significantly with the severity of PE, as assessed with echocardiography, CTPA, and arterial blood gas analysis, in normotensive patients. It can be used to predict with acceptable sensitivity and specificity the presence of RVD in these cases and, accordingly, it has potential value in risk‐stratification strategies. Larger, multicenter studies, should confirm these results before they can be applied to clinical practice.

References
  1. Goldhaber SZ,Visani L,De Rosa M.Acute pulmonary embolism: clinical outcomes in the International Cooperative Pulmonary Embolism Registry (ICOPER).Lancet.1999;353:13861389.
  2. Wan S,Quinlan DJ,Agnelli G,Eikelboom JW.Thrombolysis compared with heparin for the initial treatment of pulmonary embolism. A meta‐analysis of the randomized controlled trials.Circulation.2004;110:744749.
  3. Kasper W,Konstantinides S,Geibel A, et al.Management strategies and determinants of outcome in acute major pulmonary embolism: results of a multicenter registry.J Am Coll Cardiol.1997;30:11651171
  4. Ten Wolde M,Söhne M,Quak E,Mac Gillavry MR,Büller HR.Prognostic value of echocardiographycally assessed right ventricular dysfunction in patients with pulmonary embolism.Arch Intern Med.2004;164:16851689.
  5. Schoepf UJ,Kucher N,Kipfmueller F, et al.Right ventricular enlargement on chest computed tomography. A predictor of early death in acute pulmonary embolism.Circulation.2004;110:32763280.
  6. Punukollu G,Gowda RM,Vasavada BC,Khan IA.Role of electrocardiography in identifying right ventricular dysfunction in acute pulmonary embolism.Am J Cardiol.2005;96:450452.
  7. Golpe R,Pérez‐de‐Llano LA,Castro‐Añón O, et al.Right ventricle dysfunction and pulmonary hypertension in hemodynamically stable pulmonary embolism.Respir Med.2010;104:13701376.
  8. Daniel KR,Courtney M,Kline JA.Assessment of cardiac stress from massive pulmonary embolism with 12‐lead ECG.Chest.2001;120:474481.
  9. McQuillan BM,Picard MH,Leavitt M, et al.Clinical correlates and reference intervals for pulmonary artery systolic pressure among echocardiographically normal subjects.Circulation.2001;104:27972802.
  10. Berger M,Haimowitz A,Van Tosh A, et al.Quantitative assessment of pulmonary hypertension in patients with tricuspid regurgitation using continuous wave Doppler ultrasound.J Am Coll Cardiol.1985;6:359365.
  11. Kircher BJ,Himelman RB,Schiller NB.Noninvasive estimation of right atrial pressure from the inspiratory collapse of the inferior vena cava.Am J Cardiol.1990;66:493496.
  12. Qanadli SD,El Hajjam M,Vieillard‐Baron A, et al.New CT index to quantify arterial obstruction in pulmonary embolism: comparison with angiographic index and echocardiography.AJR Am J Roentgenol.2001;176:14151420.
  13. Toosi MS,Merlino JD,Leeper KV.Electrocardiographic score and short‐term outcomes of acute pulmonary embolism.Am J Cardiol.2007;100:11721176.
  14. Kostrubiec M,Hrynkiewicz A,Pedowska‐Wtoszek J, et al.Is it possible to use standard electrocardiography for risk assessment of patients with pulmonary embolism?Kardiol Pol.2009;67:744750.
  15. Vanni S,Polidori G,Vergara R, et al.Prognostic value of ECG among patients with acute pulmonary embolism and normal blood pressure.Am J Med.2009;122:257264.
  16. Kucher N,Walpoth N,Wustmann K,Noveanu M,Gertsch M.QR in V1‐an ECG sign associated with right ventricular strain and adverse clinical outcome in pulmonary embolism.Eur Heart J.2003;24:11131119.
  17. Torbicki A,Perrier A,Konstantinides S, et al.Guidelines on the diagnosis and management of acute pulmonary embolism. The Task Force for the Diagnosis and Management of Acute Pulmonary Embolism of the European Society of Cardiology (ESC).Eur Heart J.2008;29:22762315.
  18. Konstantinides S.Acute pulmonary embolism.N Eng J Med.2008;359:28042813.
  19. Iles S,Le Heron CJ,Davies G,Turner JG,Beckert LEL.ECG score predicts those with the greatest percentage of perfusion defects due to acute pulmonary thromboembolic disease.Chest.2004;125:16511656.
  20. Kanbay A,Kokturk N,Kaya MG, et al.Electrocardiography and Wells scoring in predicting the anatomic severity of pulmonary embolism.Respir Med.2007;101:11711176.
  21. Subramaniam RM,Mandrekar J,Chang C, et al.Pulmonary embolism outcome: a prospective evaluation of CT pulmonary angiographic clot burden score and ECG score.AJR Am J Roentgenol.2008;190:15991604.
  22. Van der Meer RW,Pattynama PMT,Van Strijen MJL, et al.Right ventricular dysfunction and pulmonary obstruction index at helical CT: prediction of clinical outcome during 3‐month follow‐up in patients with acute pulmonary embolism.Radiology.2005;235:798803.
  23. Ghuysen AE,Ghaye B,Villems V,Dondelinger RF,D'Orio V.Acute PE mortality prediction by analysis of helical computed tomography angiography and hemodynamic evaluation.Thorax.2005;60:956961.
  24. Araoz PA,Gotway MB,Harrington JR,Harmsen WS,Mandrekar JN.Pulmonary embolism: prognostic CT findings.Radiology.2007;242:889893.
References
  1. Goldhaber SZ,Visani L,De Rosa M.Acute pulmonary embolism: clinical outcomes in the International Cooperative Pulmonary Embolism Registry (ICOPER).Lancet.1999;353:13861389.
  2. Wan S,Quinlan DJ,Agnelli G,Eikelboom JW.Thrombolysis compared with heparin for the initial treatment of pulmonary embolism. A meta‐analysis of the randomized controlled trials.Circulation.2004;110:744749.
  3. Kasper W,Konstantinides S,Geibel A, et al.Management strategies and determinants of outcome in acute major pulmonary embolism: results of a multicenter registry.J Am Coll Cardiol.1997;30:11651171
  4. Ten Wolde M,Söhne M,Quak E,Mac Gillavry MR,Büller HR.Prognostic value of echocardiographycally assessed right ventricular dysfunction in patients with pulmonary embolism.Arch Intern Med.2004;164:16851689.
  5. Schoepf UJ,Kucher N,Kipfmueller F, et al.Right ventricular enlargement on chest computed tomography. A predictor of early death in acute pulmonary embolism.Circulation.2004;110:32763280.
  6. Punukollu G,Gowda RM,Vasavada BC,Khan IA.Role of electrocardiography in identifying right ventricular dysfunction in acute pulmonary embolism.Am J Cardiol.2005;96:450452.
  7. Golpe R,Pérez‐de‐Llano LA,Castro‐Añón O, et al.Right ventricle dysfunction and pulmonary hypertension in hemodynamically stable pulmonary embolism.Respir Med.2010;104:13701376.
  8. Daniel KR,Courtney M,Kline JA.Assessment of cardiac stress from massive pulmonary embolism with 12‐lead ECG.Chest.2001;120:474481.
  9. McQuillan BM,Picard MH,Leavitt M, et al.Clinical correlates and reference intervals for pulmonary artery systolic pressure among echocardiographically normal subjects.Circulation.2001;104:27972802.
  10. Berger M,Haimowitz A,Van Tosh A, et al.Quantitative assessment of pulmonary hypertension in patients with tricuspid regurgitation using continuous wave Doppler ultrasound.J Am Coll Cardiol.1985;6:359365.
  11. Kircher BJ,Himelman RB,Schiller NB.Noninvasive estimation of right atrial pressure from the inspiratory collapse of the inferior vena cava.Am J Cardiol.1990;66:493496.
  12. Qanadli SD,El Hajjam M,Vieillard‐Baron A, et al.New CT index to quantify arterial obstruction in pulmonary embolism: comparison with angiographic index and echocardiography.AJR Am J Roentgenol.2001;176:14151420.
  13. Toosi MS,Merlino JD,Leeper KV.Electrocardiographic score and short‐term outcomes of acute pulmonary embolism.Am J Cardiol.2007;100:11721176.
  14. Kostrubiec M,Hrynkiewicz A,Pedowska‐Wtoszek J, et al.Is it possible to use standard electrocardiography for risk assessment of patients with pulmonary embolism?Kardiol Pol.2009;67:744750.
  15. Vanni S,Polidori G,Vergara R, et al.Prognostic value of ECG among patients with acute pulmonary embolism and normal blood pressure.Am J Med.2009;122:257264.
  16. Kucher N,Walpoth N,Wustmann K,Noveanu M,Gertsch M.QR in V1‐an ECG sign associated with right ventricular strain and adverse clinical outcome in pulmonary embolism.Eur Heart J.2003;24:11131119.
  17. Torbicki A,Perrier A,Konstantinides S, et al.Guidelines on the diagnosis and management of acute pulmonary embolism. The Task Force for the Diagnosis and Management of Acute Pulmonary Embolism of the European Society of Cardiology (ESC).Eur Heart J.2008;29:22762315.
  18. Konstantinides S.Acute pulmonary embolism.N Eng J Med.2008;359:28042813.
  19. Iles S,Le Heron CJ,Davies G,Turner JG,Beckert LEL.ECG score predicts those with the greatest percentage of perfusion defects due to acute pulmonary thromboembolic disease.Chest.2004;125:16511656.
  20. Kanbay A,Kokturk N,Kaya MG, et al.Electrocardiography and Wells scoring in predicting the anatomic severity of pulmonary embolism.Respir Med.2007;101:11711176.
  21. Subramaniam RM,Mandrekar J,Chang C, et al.Pulmonary embolism outcome: a prospective evaluation of CT pulmonary angiographic clot burden score and ECG score.AJR Am J Roentgenol.2008;190:15991604.
  22. Van der Meer RW,Pattynama PMT,Van Strijen MJL, et al.Right ventricular dysfunction and pulmonary obstruction index at helical CT: prediction of clinical outcome during 3‐month follow‐up in patients with acute pulmonary embolism.Radiology.2005;235:798803.
  23. Ghuysen AE,Ghaye B,Villems V,Dondelinger RF,D'Orio V.Acute PE mortality prediction by analysis of helical computed tomography angiography and hemodynamic evaluation.Thorax.2005;60:956961.
  24. Araoz PA,Gotway MB,Harrington JR,Harmsen WS,Mandrekar JN.Pulmonary embolism: prognostic CT findings.Radiology.2007;242:889893.
Issue
Journal of Hospital Medicine - 6(5)
Issue
Journal of Hospital Medicine - 6(5)
Page Number
285-289
Page Number
285-289
Article Type
Display Headline
Electrocardiogram score predicts severity of pulmonary embolism in hemodynamically stable patients
Display Headline
Electrocardiogram score predicts severity of pulmonary embolism in hemodynamically stable patients
Legacy Keywords
electrocardiography, pulmonary embolism, right ventricular function, computer tomography, echocardiography
Legacy Keywords
electrocardiography, pulmonary embolism, right ventricular function, computer tomography, echocardiography
Sections
Article Source

Copyright © 2010 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Servicio de Neumología, Complexo Hospitalario Xeral‐Calde, c/Dr Ochoa, s/n. 27004 Lugo, Spain
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Continuing Medical Education Program in

Article Type
Changed
Mon, 05/22/2017 - 21:42
Display Headline
Continuing Medical Education Program in the Journal of Hospital Medicine

If you wish to receive credit for this activity, please refer to the website: www.wileyblackwellcme.com.

Accreditation and Designation Statement

Blackwell Futura Media Services designates this journal‐based CME activity for a maximum of 1 AMA PRA Category 1 Credit.. Physicians should only claim credit commensurate with the extent of their participation in the activity.

Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians.

Educational Objectives

Upon completion of this educational activity, participants will be better able to:

  • Illustrate the elements of a systematic approach to successful hospital smoking cessation programs.

  • Describe the efficacy of a coordinated real world&#148; hospital smoking cessation program in a U.S. hospital.

  • Evaluate the barriers to successful hospital smoking cessation programs.

 

This manuscript underwent peer review in line with the standards of editorial integrity and publication ethics maintained by Journal of Hospital Medicine. The peer reviewers have no relevant financial relationships. The peer review process for Journal of Hospital Medicine is blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review.

Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services's Policy on Activity Disclosure and Conflict of Interest. The primary resolution method used was peer review and review by a non‐conflicted expert.

Instructions on Receiving Credit

For information on applicability and acceptance of CME credit for this activity, please consult your professional licensing board.

This activity is designed to be completed within an hour; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial publication.

Follow these steps to earn credit:

  • Log on to www.wileyblackwellcme.com

  • Read the target audience, learning objectives, and author disclosures.

  • Read the article in print or online format.

  • Reflect on the article.

  • Access the CME Exam, and choose the best answer to each question.

  • Complete the required evaluation component of the activity.

 

This activity will be available for CME credit for twelve months following its publication date. At that time, it will be reviewed and potentially updated and extended for an additional twelve months.

Article PDF
Issue
Journal of Hospital Medicine - 6(5)
Page Number
290-290
Sections
Article PDF
Article PDF

If you wish to receive credit for this activity, please refer to the website: www.wileyblackwellcme.com.

Accreditation and Designation Statement

Blackwell Futura Media Services designates this journal‐based CME activity for a maximum of 1 AMA PRA Category 1 Credit.. Physicians should only claim credit commensurate with the extent of their participation in the activity.

Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians.

Educational Objectives

Upon completion of this educational activity, participants will be better able to:

  • Illustrate the elements of a systematic approach to successful hospital smoking cessation programs.

  • Describe the efficacy of a coordinated real world&#148; hospital smoking cessation program in a U.S. hospital.

  • Evaluate the barriers to successful hospital smoking cessation programs.

 

This manuscript underwent peer review in line with the standards of editorial integrity and publication ethics maintained by Journal of Hospital Medicine. The peer reviewers have no relevant financial relationships. The peer review process for Journal of Hospital Medicine is blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review.

Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services's Policy on Activity Disclosure and Conflict of Interest. The primary resolution method used was peer review and review by a non‐conflicted expert.

Instructions on Receiving Credit

For information on applicability and acceptance of CME credit for this activity, please consult your professional licensing board.

This activity is designed to be completed within an hour; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial publication.

Follow these steps to earn credit:

  • Log on to www.wileyblackwellcme.com

  • Read the target audience, learning objectives, and author disclosures.

  • Read the article in print or online format.

  • Reflect on the article.

  • Access the CME Exam, and choose the best answer to each question.

  • Complete the required evaluation component of the activity.

 

This activity will be available for CME credit for twelve months following its publication date. At that time, it will be reviewed and potentially updated and extended for an additional twelve months.

If you wish to receive credit for this activity, please refer to the website: www.wileyblackwellcme.com.

Accreditation and Designation Statement

Blackwell Futura Media Services designates this journal‐based CME activity for a maximum of 1 AMA PRA Category 1 Credit.. Physicians should only claim credit commensurate with the extent of their participation in the activity.

Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians.

Educational Objectives

Upon completion of this educational activity, participants will be better able to:

  • Illustrate the elements of a systematic approach to successful hospital smoking cessation programs.

  • Describe the efficacy of a coordinated real world&#148; hospital smoking cessation program in a U.S. hospital.

  • Evaluate the barriers to successful hospital smoking cessation programs.

 

This manuscript underwent peer review in line with the standards of editorial integrity and publication ethics maintained by Journal of Hospital Medicine. The peer reviewers have no relevant financial relationships. The peer review process for Journal of Hospital Medicine is blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review.

Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services's Policy on Activity Disclosure and Conflict of Interest. The primary resolution method used was peer review and review by a non‐conflicted expert.

Instructions on Receiving Credit

For information on applicability and acceptance of CME credit for this activity, please consult your professional licensing board.

This activity is designed to be completed within an hour; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial publication.

Follow these steps to earn credit:

  • Log on to www.wileyblackwellcme.com

  • Read the target audience, learning objectives, and author disclosures.

  • Read the article in print or online format.

  • Reflect on the article.

  • Access the CME Exam, and choose the best answer to each question.

  • Complete the required evaluation component of the activity.

 

This activity will be available for CME credit for twelve months following its publication date. At that time, it will be reviewed and potentially updated and extended for an additional twelve months.

Issue
Journal of Hospital Medicine - 6(5)
Issue
Journal of Hospital Medicine - 6(5)
Page Number
290-290
Page Number
290-290
Article Type
Display Headline
Continuing Medical Education Program in the Journal of Hospital Medicine
Display Headline
Continuing Medical Education Program in the Journal of Hospital Medicine
Sections
Article Source

Copyright © 2010 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Department of Preventive Medicine and Public Health, Mail‐Stop 1008, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Hospitalists and Sickle Cell Disease

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Frequently asked questions by hospitalists managing pain in adults with sickle cell disease

Severe, disabling pain, often requiring opioids, is the most common medical presentation for children and adults with sickle cell disease (SCD), an autosomal recessive red blood cell disorder affecting those of African, Mediterranean, and Asian descent.1, 2 A genetically controlled hemoglobin alteration impairs oxygen binding, and enables polymerization of deoxy‐hemoglobin, resulting in, classically, sickle‐shaped erythrocytes3 and a complex cascade of ischemia and vaso‐occlusion in the microcirculation.4, 5

Dramatic gains in the treatment of SCD in childhood have resulted in markedly improved survival through adulthood.68 Thus, the need for adult SCD care is relatively new and rapidly growing. In 2005, approximately 70% of the nearly 80,000 US SCD hospitalizations occurred in adults versus children (Table 1). These hospitalizations occurred in the context of a poorly coordinated American health care system,9 despite the hopes raised by the Patient‐Centered Medical Home10 and the Chronic Care Model.11

Adult and Pediatric Admissions for Sickle Cell Disease,* 2005 and 2008
2005 2008
Total No. of Discharges LOS Total No. of Discharges LOS
  • NOTE: Data extracted from the Healthcare Cost and Utilization Project database, http://www.ahrq.gov/data/hcup/.

  • Abbreviations: LOS, length of stay in days (mean).

  • International Classification of Diseases, Clinical Modification (ICD‐CM) principal diagnosis code(s) 282.60, 282.61, 282.62, 282.63, 282.64, 282.69.

All discharges 79,187 100.00% 5.3 70,121 100.00% 5.4
Age group <1 996 1.26% 2.5 513 0.73% 2.7
1‐17 23,134 29.21% 3.9 13,754 19.62% 3.8
18‐44 48,168 60.83% 6 48,021 68.48% 5.8
45‐64 6,527 8.24% 6 7,543 10.76% 5.6
65‐84 281 0.35% 6.4 221 0.32% 5.6
Missing 81 0.10% 4 70 0.10% 3.0

Adults with SCD are vulnerable both because they are usually members of racial and ethnic minority groups, and because they have a Food and Drug Administration (FDA)‐defined orphan disease.12 They often do not receive the only FDA‐approved medication for SCD, life‐saving hydroxyurea,13 recommended for adults with homozygous sickle cell anemia (Hb SS) and sickle‐othalassemia (Hb SoThal).14 Young adults often fail to experience a smooth transition of care from children's hospitals, falling into a medical abyss.15

Therefore, increasingly, hospitalists are managing adults with SCD, rather than adult hematologyoncology, pain, or palliative care specialists. Adults with SCD experience negative opinions, bilateral lack of trust, and conflict in the doctorpatient relationship, frequently cited in studies of SCD adults and providers in the literature.16, 17

Evidence Base

General guidelines for SCD management have been published by the National Institutes of Health (NIH)18 and the Agency for Healthcare Policy and Research.19 But one of us (K.L.H.) found evidence lacking with regard to SCD pain management.20 Published guidelines on general pain management, such as the World Health Organization's Analgesic Ladder,21 do not address SCD. A Cochrane Review of pain management in SCD found only 9 randomized controlled trials, all with small numbers of patients, addressing acute SCD pain only.22 As well, American and British consensus SCD pain guidelines23, 24 admit, and subsequent publications emphasize,25, 26 the lack of evidence for what to do or not do for SCD pain management. At least 1 well‐done summary of the SCD evidence base intended for hospitalists has been published, but it focuses on management of issues other than pain.27

Motivations and Fears

It is not surprising then that hospitalists may bring great fear and apprehension with them into their care of SCD patients. One of us (W.R.S.), a general internist, has been called by his own and 3 other academic medical centers, 2 with active Federally‐funded SCD research programs, to address the problems of high‐utilizing adults with SCD, including counseling hospitalists frustrated with the management of pain in these patients.

Hospitalists may be motivated to provide efficient inpatient management (Table 2), and be aware of pain as the primary symptom of SCD inpatients. But they may carry knowledge gaps and biases into their relationships with SCD inpatients. They may fear opioid administration (opiophobia), loss of licensure or governmental reprisals because of high‐dose prescription of opioids, or may believe that SCD patients are more often addicted than most.17, 28 Consequently, more troublesome hospital stays may occur when patients are not rapidly and adequately titrated to appropriate analgesic doses, or when unnecessary deleterious side effects result from opioid and other analgesics. We therefore offer answers to frequently asked questions (FAQs) about pain management by hospitalists caring for adults with SCD. We address FAQs arising during the prototypical situationa patient with SCD admitted for a painful exacerbation, and little or no acute comorbidity. We refer the reader to the aforementioned articles and guidelines to address other treatment issues in adults with SCD.

General Motivations and Principles of Efficient Inpatient Sickle Cell Pain Management, and the Obstacles to Inpatient Care of Sickle Cell Disease
Principle Obstacles
  • Abbreviations: DEA, Drug Enforcement Administration; ED, emergency department.

Make appropriate management handoffs for patients coming from the ED to promote continuity of care and shorten hospitalization Poor information systems and poor handoffs/continuity from ED management to hospital management
Get as much preexisting information about the patient as possible to inform acute care, avoid returns or further hospitalization Patient may have no primary care physician or may underutilize primary care
Patient may misuse ED and hospital (as primary care source)
Provide rapid and adequate analgesia Ignorance of the differences between tolerance, physical dependence, addiction, and pseudoaddiction
No specific data on pharmacodynamics of opioid analgesics in sickle cell disease
Don't lose licensure or arouse regulatory suspicion about prescribing patterns Ignorance of DEA monitoring and laws governing appropriate vs inappropriate prescribing of opioids
Get the patient discharged as soon as medically appropriate Difficulty assessing pain quality and intensity
Difficulty assessing/avoiding side effects of analgesics
Difficulty determining appropriateness/timing of changes in analgesic dosing, discharge planning
Make appropriate handoffs with the patient's usual source of continuity care (provide that source when necessary) to avoid returns or further hospitalization Patient may have no primary care physician or may underutilize primary care physician
Patient may be mis/underprescribed analgesics by primary care physician
Define and maintain appropriate roles for hospitalists vs physicians with sickle cell training, pain specialists, or other specialists Inadequate adult system of care for sickle cell disease (no/paucity of specialty care)
Multiple prescribers of opioids
Providers unwilling to care for or prescribe opioids for sickle cell patients

FAQS

  • Is there any objective way to tell when SCD patients really are in a crisis?

    Although the term crisis is used as if it were an objectively definable biological entity, no one has proposed a standard definition of a crisis based on pain intensity level, clinical features, or biomarkers. Measures of vaso‐occlusion are correlated with ischemic pain, including pain that is often called a crisis.2932 However, neither ischemic pain from SCD, nor the underlying vaso‐occlusive cascade that is associated with this pain, is a sudden, present‐or‐absent phenomenon. Instead, these are continua that can be measured using pain scales or various biomarkers.

    There is, however, correlative evidence of the intensity of SCD pain associated with various distinctive health states (admitted/not admitted, in crisis/not in crisis). The most visible measure of a crisis, health care utilization, was a strong predictor of mortality in the Cooperative Study of Sickle Cell Disease. Patients with 3 or more admissions per year had a lower 5‐year survival rate.33 In contrast, crisis in the landmark Pain in Sickle Cell Epidemiology Study (PiSCES) was self‐defined by patients.34 Despite being in pain on over half of their days on average, and despite a third of patients being in pain daily, most pain in PiSCES was not considered a crisis, and less than 5% of patients' days were spent in emergency departments (EDs) or hospitals. Ambulatory pain intensity reports were correlated with opioid use.35 A substantial minority (35%) of PiSCES patients made at least 3 ED visits per year. However, these high ED utilizers had worse laboratory values, more pain, more distress, and a lower quality of life.36

    Importantly, sometimes adults with SCD may have severe comorbidities which may not be addressed or may be mistakenly managed as an acute vaso‐occlusive episode without further investigation or timely specialist consultation. Although pain is primarily the individual's chief complaint, any potential relationship between the presence of medical comorbidities and pain should be examined when patients are admitted.

  • How can one know when opioid dosages should be changed, or when SCD pain is appropriately controlled to allow discharge?

    We recommend, as a standard of care, that SCD pain assessment and pain therapy be interwoven, despite a systematic review finding no evidence that directly linked the timing, frequency, or method of pain assessment with outcomes or safety in medical inpatients, and concluding that the safety and effectiveness of patient‐controlled analgesia (PCA) in medical patients had not been adequately studied.37 Hospitalists should focus the first 24 hours of inpatient SCD pain management on cycles of recurrent pain assessment and opioid dose titration as frequently as every 1 to 2 hours, to assure safe and rapidly efficacious analgesia. Pain intensity, duration, and character should be assessed directly. Intensity is often assessed using a visual analog scale (VAS) or numeric rating scale.38 Treating physicians should themselves directly assess pain during discussions of therapy with the patient, even though some assessment usually is done in hospitals during each nursing shift. Pain and pain relief can be assessed indirectly by monitoring opioid use.

    We recommend PCA for inpatients with SCD, administered as an intermittent demand dose (patient must push a button) of opioid, with or without a background opioid constant infusion.39 We usually set the interval between doses, or lockout, to 6 to 10 minutes. Both the lockout and the sedation from delivered doses prevent patients from pushing the demand button repetitively to the point of overdose. Use of a low‐level constant infusion (basal) may sustain pain control during times when the patient is asleep, avoiding recrudescent pain and lost ground due to inadequate analgesia during rest. Alternatively, long‐acting oral opioids may be continued if already used at home, or newly introduced to provide adequate baseline pain control which is augmented by the demand dosing. Most PCA pumps monitor hourly opioid dose demand (number of pushes), as well as hourly doses delivered. Both hourly opioid dose demand and hourly dose‐per‐demand ratio are measures of PCA efficacy or futility. Pumps record this data, and can be interrogated at the patient's bedside for up to several days of prior use. Physicians should combine pump interrogation with direct pain assessment to guide demand‐dose titration. Demand doses should be increased to 1.5 to 2 times the previous demand dose after several hours of failed reduction of pain intensity and duration, and/or persistently futile dose‐per‐demand ratios.

    PCA interrogation is also useful for conversion of parenteral opioids to oral opioids, as well as to guide the recommendation for discharge home. After the first 24‐48 hours of up‐titration, if opioid dose demand decreases concordantly with pain frequency and intensity, the demand dose may be safely decreased, and eventually daily PCA requirements may be summed and converted to oral medication using standard opioid dose conversion tables. At this point, physicians may use single measures or daily averages of directly assessed pain.

    Routine PCA use in SCD is backed by some evidence.40, 41 But we find it important that patients be taught and encouraged to use the demand feature of PCA. Still, for various reasons, some patients do not use PCA pumps well. Discordant or unreliable assessments (eg, high pain intensity but low‐opioid demand doses during the same interval) may result, and PCA potentially may fail as a dosing strategy. Management is more difficult for these patients. One alternative dosing strategy is prescription of scheduled doses of a short‐acting opioid, attaching to each dose the order, patient may refuse. This is different than dosing as needed, and allows counts of dose refusals over an interval, analogous to PCA pump interrogation.

  • How much is too much opioid? Should one rely on side effects, or on requests for medicine, or is there a ceiling dose?

    Addictionologists, pain specialists, oncologists, those involved in hospice care, and some hematologists caring for SCD patients agree that, in general, there should be no a priori dose limitations imposed on opioid prescribing for acute pain. Instead, titration of dose of opioid to pain relief is a central principle of acute pain management. Experts also agree that particular opioids carry particular side effects which warrant dose limitation, adjustments, or avoidance of that opioid altogether. A summary of opioids commonly used in SCD, along with warnings and implied dose limitations is found in Table 3.

    For safety, it is important to assess the history of prior opioid use to recognize a patient who is not tolerant to opioids (see below, FAQ 4), to avoid mistakenly overdosing a patient using doses often required by tolerant patients. In lieu of a pre‐written, individualized opioid dosing plan in place for the patient, the patient may be the best source of information regarding preferred medication and tolerated doses.

    The reader is referred to standard texts for a description of opioids, their pharmacokinetics and pharmacodynamics, and their addictive and abuse potential. The side‐effect profile of opioids is well‐known: nausea, vomiting, and itching frequently occur; hallucinations, respiratory suppression, and myoclonus occur infrequently.42 Meperidine may more readily cause central nervous system (CNS) dysfunction, including seizures, as compared to other commonly used opioids, because of its toxic metabolite nor‐meperidine. Use of meperidine is often avoided, especially use via PCA.43 Methadone may cause dysrhythmias, specificially corrected Q‐T interval (QTc) prolongation and torsades de pointes on an electrocardiogram, in doses above 200 mg per day.44 Some recommend baseline and yearly electrocardiogram monitoring when giving methadone chronically.

    Recognizing the potential dangers of opioids, it is also reasonable to look for opioid‐sparing analgesic strategies. Non‐opioid analgesics such as ketorolac45 and adjuvants such as ketamine46 that are opioid‐sparing should be considered whenever feasible. Complementary and alternative therapies such as transcutaneous electrical nerve stimulation (TENS)47 have less evidence of effectiveness, but have limited risks and may be of use for some individuals.

  • What are the major signs of substance abuse (opioids, street drugs) in SCD patients already on opioids, and can a hospitalist judge those signs acutely and intervene appropriately?

    Reports of underprescription of opioids in SCD have cited physician fear of abuse and addiction.48 A recent informal poll of adult sickle cell providers suggests policies vary on how potential abuse is monitored in ambulatory sickle cell patients. We note that physicians, especially upon meeting a patient for the first time, may be unable to reliably judge whether that patient is abusing opioids or street drugs. Both false‐positive and false‐negative diagnoses may be made.49 Repetitive reports of lost or stolen prescriptions or pill bottles, receipt of prescriptions from multiple providers, or repeated requests for early refills increase the suspicion of misuse or abuse, but are indirect evidence. Urine and serum monitoring may be useful, but may give incorrect information if misinterpreted or not conducted frequently enough to improve sensitivity.50

    It is important to distinguish between tolerance, the decreased analgesic response over time to repeated doses of the same drug; physical dependence, the production of withdrawal upon abrupt discontinuation of an opioid agonist or administration of an antagonist; and addiction, the psychological dependence upon opioids. Tolerance may be misperceived as true addiction. Its earliest symptom is shortening of the duration of effective analgesia. In contrast, addiction may be manifested by dose escalation in the absence of an increased pain stimulus, or by use of opioids for purposes other than pain relief.51 These are not easily distinguished during a single patient encounter.

    SCD patients' requests for specific opioid medications in specific doses, should not be taken as evidence of past or current abuse, but rather evidence of a well‐informed, self‐managing patient. Adults with SCD are clearly expected to be very knowledgeable about and tolerant to opioids if they have had a life of pain as a child, and will require higher doses of opioids than other patients treated by most hospitalists. The issue of medication abuse may be best handled in the ambulatory setting. Whenever possible, hospitalists should not rely only on data from the acute care setting to manage patients. Ambulatory providers may conduct random, unannounced urine and/or serum testing, as part of an opioid prescribing agreement that is written and filed in the patient's chart. Assays for prescribed opioids (especially long‐acting agents), as well as assays for common drugs of abuse, should be conducted. Comanagement with an addictionologist, psychiatrist, or psychologist should be considered in individuals suspected of opioid abuse.

    We do not suggest routine urine drug test monitoring of all SCD patients unless routine monitoring is done as a policy for all patients on opioids. Though the prevalence of addiction may be higher in subpopulations of patients with pain,52 and though prescription of opioids, prescription drug abuse, and accidental deaths from prescribed opioids have risen exponentially in the last several years,53 in our experience and in the published literature, drug misuse/abuse among SCD patients is no worse than among patients with other illnesses.5456 However, pseudoaddiction, the appropriate seeking of needed opioids from multiple physicians because of uncontrolled pain and opioid underprescription, may well be prevalent in SCD,57 and may be mistaken for true addiction.

  • How can patients' readiness for discharge be assessed? What can be done for the patient who has lengthy and/or multiple hospitalizations or frequent ED visits?

    The appropriate time for discharge in most patients is when they can manage their pain at home with oral opioids or less. Often, patients do not improve even after a few days of inpatient therapy.58 A typical pain episode may last much longer than the 6‐day average US hospital length of stay for a diagnosis of sickle cell crisis among 18‐44 year olds (Table 1).59 Patients may return and be readmitted.60, 61 But in the best cases, pain resolves or reverts to a usual chronic intensity level. As described in FAQ 2, daily or more frequent pain assessment is a bedrock for making discharge decisions. Patients well‐experienced in the use of pain intensity scales can report their usual pain intensity at home, and how close they are to their baseline pain intensity. Simply asking patients, Are you ready for discharge? is appropriate and may yield a surprising positive response. In a recent inpatient trial of PCA (manuscript in preparation), adult patients were admitted with a minimum pain intensity of 45 mm on a 100 mm horizontal VAS scale after treatment in the ED, and mean pain intensity of 76 mm 10 mm. All adults in this study were discharged with pain that was clinically significantly lower. Researchers have found a VAS change of 13.5 mm to be the minimum clinically significant change62 during treatment of vaso‐occlusive crisis.63

    Unremitting pain despite appropriate titration of opioids and prolonged hospital stays suggests the need for comprehensive evaluation for medical and psychosocial comorbidities, as is done for other patients with chronic pain syndromes. If not already done, discussion with the patient's primary care provider may reveal factors impacting on persistent pain. Consultation with a hematologist, pain or palliative care specialist, or other provider familiar with SCD may prove helpful. Implementation of adjuvant therapies as discussed in FAQ 3 and adding long‐acting oral opioids to continue postdischarge may also help. Hyperalgesia, or heightened sensitivity to pain, is normal after acute tissue injury, but is now suspected in SCD as a long‐term neuropathic pain syndrome, as a consequence either of repeated painful crises or of chronic opioid therapy.2 Only some centers have specialists qualified to test for and diagnose neuropathic pain.64

    Discharge planning should include identification of a source of outpatient follow‐up. Opioids prescribed at discharge should be sufficient to last at least until the first outpatient appointment, to avoid repeated ED or hospital visits. Communication with a primary care provider at discharge can enhance successful care transition. Otherwise, for patients without established providers, social workers and others may address barriers to follow‐up that frustrate both patient and provider.

Table of Opioids, Frequency of Use, and Special Considerations in SCD
Opioid Used Frequently (>20% of Patients) How Used Unique Side Effects and/or Dose Limitations
  • Abbreviations: SCD, sickle cell disease; QTc, corrected Q‐T interval on electrocardiogram.

Short‐acting
Codeine No Inpatient, parenteral; Ambulatory, oral
Oxycodone Yes Most commonly used ambulatory opioid
Morphine Yes Most commonly used inpatient opioid
Hydromorphone Yes Inpatient more than ambulatory
Fentanyl No Inpatient, parenteral Short‐acting
Hydrocodone No Ambulatory
Meperedine No Avoided Unpredictable seizure, coma, death
Propoxyphene No Ambulatory
Tramadol No Ambulatory
Long‐acting
Oxycodone No Ambulatory and as an oral basal in inpatients Abuse potential from capsule manipulation
Morphine Yes Ambulatory and as an oral basal in inpatients; most commonly used long‐acting opioid
Methadone No Ambulatory and as an oral basal in inpatients Dose‐dependent prolongation of QTc, torsades de pointes
Fentanyl No Ambulatory and as a transdermal basal in inpatients Abuse potential from transdermal patch manipulation

Support for Hospitalists Managing Adults With Sickle Cell Disease

Beside the general advice on pain management in SCD mentioned above or found in the bibliography of this article, at long last, a group of adult practitioners skilled in the care of SCD has formed nationally. The Sickle Cell Adult Provider Network [http://www.scapn.net] provides non‐binding advice and support to its members via an e‐mail listserve. Topics often include pain management. This advice fills a vacuum created by the lack of evidence‐based guidelines.

Ultimately, evidence and updated guidelines will be the best support for hospitalists and others managing pain in SCD. The hope is that SCD will receive the attention it deserves, so that practitioners and patients alike do not suffer continued pain from this disease or its management.

References
  1. Serjeant GR.Sickle‐cell disease.Lancet.1997;350(9079):725302.
  2. Smith WR,Scherer M.Sickle‐cell pain: advances in epidemiology and etiology.Hematology Am Soc Hematol Educ Program.2010;409415. PMID: 21239827.
  3. Steinberg MH.Management of sickle cell disease.N Engl J Med.1999;340:10211030.
  4. Hebbel RP,Vercellotti G,Nath KA.A systems biology consideration of the vasculopathy of sickle cell anemia: the need for multi‐modality chemo‐prophylaxsis.Cardiovasc Hematol Disord Drug Targets.2009;9(4):271292.
  5. Conran N,Franco‐Penteado CF,Costa FF.Newer aspects of the pathophysiology of sickle cell disease vaso‐occlusion [review].Hemoglobin.2009;33(1):116.
  6. Davis H,Schoendorf KC,Gergen PJ,Moore RM.National trends in the mortality of children with sickle cell disease, 1968 through 1992.Am J Public Health.1997;87(8):13171322.
  7. Adams RJ,McKie VC,Hsu L, et al.Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography.N Engl J Med.1998;339:511.
  8. Adams RJ,Brambilla D.Optimizing primary stroke prevention in sickle cell anemia (STOP 2) trial investigators. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease.N Engl J Med.2005;353(26):27692778.
  9. Sandy LG.Homeostasis without reserve—the risk of health system collapse.N Engl J Med.2002;347(24):19711975.
  10. The Advanced Medical Home: A Patient‐Centered, Physician‐Guided Model of Health Care [Policy Monograph].Philadelphia, PA:American College of Physicians;2006.
  11. Wagner EH,Glasgow RE,Davis C,Bonomi AE,Provost L,McCulloch D.Quality improvement in chronic illness care: a collaborative approach.Jt Comm J Qual Improv.2001;27:6380.
  12. Definition of Disease Prevalence for Therapies Qualifying Under the Orphan Drug Act. Subpart C, Designation of an Orphan Drug. Sec. 316.20. Content and format of a request for orphan‐drug designation. Available at: http://www.fda.gov/orphan/designat/prevalence.html. Accessed September 3,2008.
  13. Steinberg MH,Barton F,Castro O, et al.Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment.JAMA.2003;289:16451651.
  14. Lanzkron S,Haywood C,Hassell KL,Rand C.Provider barriers to hydroxyurea use in adults with sickle cell disease: a survey of the Sickle Cell Disease Adult Provider Network.J Natl Med Assoc.2008;100(8):968973.
  15. Treadwell M,Telfair J,Gibson RW,Johnson S,Osunkwo I.Transition from pediatric to adult care in sickle cell disease: establishing evidence‐based practice and directions for research.Am J Hematol.2011;86(1):116120. PMID: 21061308.
  16. Bobo L,Miller ST,Smith WR,Elam JT,Rosmarin PC,Lancaster DJ.Health perceptions and medical care opinions of inner‐city adults with sickle cell disease or asthma compared with those of their siblings.South Med J.1989;82(1):912.
  17. Shapiro BS,Benjamin LJ,Payne R,Heidrich G.Sickle cell‐related pain: perceptions of medical practitioners.J Pain Symptom Manage.1997;14(3):168174.
  18. The Management of Sickle Cell Disease.4th ed. NIH Publication 2002–2117.Washington, DC:National Institutes of Health, National Heart, Lung, and Blood Institute, Division of Blood Diseases and Resources; June2002.
  19. Pain Management Guideline.Washington, DC:Agency for Health Care Policy and Research;1992.
  20. Lottenberg R,Hassell KL.An evidence‐based approach to the treatment of adults with sickle cell disease.Hematology Am Soc Hematol Educ Program.2005;5865.
  21. World Health Organization: Cancer Pain Relief.Geneva, Switzerland:WHO,1986.
  22. Dunlop RJ,Bennett KC.Pain management for sickle cell disease.Cochrane Database Syst Rev. April 19,2006;(2):CD003350.
  23. Benjamin L,Dampier C,Jacox A;for the Guideline Committee.Guidelines for the Management of Acute and Chronic Pain in Sickle Cell Disease. APS Clinical Practice Guideline Series, No 1.Glenview, IL:American Pain Society;1999.
  24. Rees D.Guidelines for the management of the acute painful crisis of sickle cell disease.Br J Haematol.2003;120:744752.
  25. Field JJ,Knight‐Perry JE,DeBaun MR.Acute pain in children and adults with sickle cell disease: management in the absence of evidence‐based guidelines.Curr Opin Hematol.2009;16(3):173178.
  26. Rosenblum A,Marsch LA,Joseph H,Portenoy RK.Opioids and the treatment of chronic pain: controversies, current status, and future directions.Exp Clin Psychopharmacol.2008;16(5):405416.
  27. Axon RN.A sickle cell primer.The Hospitalist.2006;10(10):3941.
  28. Hill CS.The barriers to adequate pain management with opioid analgesics.Semin Oncol.1993;20(2 suppl 1):15.
  29. Dampier C,Setty BN,Eggleston B,Brodecki D,O'Neal P,Stuart M.Vaso‐occlusion in children with sickle cell disease: clinical characteristics and biologic correlates.J Pediatr Hematol Oncol.2004;26:785790. PMID: 15591896.
  30. Graido‐Gonzalez E,Doherty JC,Bergreen EW,Organ G,Telfer M,McMillen MA.Plasma endothelin‐1, cytokine, and prostaglandin E2 levels in sickle cell disease and acute vaso‐occlusive sickle crisis.Blood.1998;92:25512555. PMID: 9746797.
  31. Michaels LA,Ohene‐Frempong K,Zhao H,Douglas SD.Serum levels of substance P are elevated in patients with sickle cell disease and increase further during vaso‐occlusive crisis.Blood.1998;92:31483151. PMID: 9787150.
  32. Landburg PP,Teerlink T,Muskiet FA,Duits AJ,Schnog JJ;for the CURAMA study group.Plasma concentrations of asymmetric dimethylarginine, an endogenous nitric oxide synthase inhibitor, are elevated in sickle cell patients but do not increase further during painful crisis.Am J Hematol. February 27,2008; PMID: 18383318
  33. Platt OS,Thorington BD,Brambilla DJ, et al.Pain in sickle cell disease: rates and risk factors.N Engl J Med.1991;325:1116.
  34. Smith WR,Bovbjerg VE,Penberthy LT, et al.Understanding pain and improving management of sickle cell disease: the PiSCES Study.J Natl Med Assoc.2005;97(2):183193.
  35. Smith WR,McClish DK,Penberthy LT, et al.Daily assessment of pain in adults with sickle cell disease.Ann Intern Med.2008;148(2):94101.
  36. Aisiku IP,Smith WR,McClish DK, et al.Comparisons of high versus low emergency department utilizers in sickle cell disease.Ann Emerg Med.2009;53(5):587593.
  37. Helfand M,Freeman M.Assessment and management of acute pain in adult medical inpatients: a systematic review.Pain Med.2009;10(7):11831199. PMID: 19818030.
  38. Wallenstein SL.Scaling clinical pain and pain relief. In: Bromm B, ed.Pain Measurement in Man: Neorephysiological Correlates of Pain.New York:Elsevier Science Publishers,1984:389396.
  39. Dubois M.Patient‐controlled analgesia for acute pain.Clin J Pain.1989;5(suppl 1):S8S15. PMID: 2520435.
  40. Melzer‐Lange MD,Walsh‐Kelly CM,Lea G,Hillery CA,Scott JP.Patient‐controlled analgesia for sickle pain crisis in pediatric emergency department.Pediatr Emerg Care.2004;20:24.
  41. Trentadue N,Kachoyeanos MK,Lea G.A comparison of two regimens of patient‐controlled analgesia for children with sickle cell disease.J Pediatr Nurs.1998;13:1519.
  42. Narcotic analgesics, 2002 update.The DAWN Report;2004.
  43. Seifert CF,Kennedy S.Meperidine is alive and well in the new millennium: evaluation of meperidine usage patterns and frequency of adverse drug reactions.Pharmacotherapy.2004;24:776783.
  44. Porter BP,Coyne PJ,Smith WR.Methadone‐related torsades de pointes in a sickle cell patient treated for chronic pain.Am J Hematol.2005;78(4):316317.
  45. de Franceschi L,Finco G,Vassanelli A,Zaia B,Ischia S,Corrocher R.A pilot study on the efficacy of ketorolac plus tramadol infusion combined with erythrocytapheresis in the management of acute severe vaso‐occlusive crises and sickle cell pain.Haematologica.2004;89(11):13891391.
  46. Zempsky WT,Loiselle KA,Corsi JM,Hagstrom JN.Use of low‐dose ketamine infusion for pediatric patients with sickle cell disease‐related pain: a case series.Clin J Pain.2010;26(2):163167.
  47. Wang WC,George SL,Wilimas JA.Transcutaneous electrical nerve stimulation treatment of sickle cell pain crises.Acta Haematol.1988;80(2):99102.
  48. Midence K,Fuggle P,Davies SC.Psychosocial aspects of sickle cell disease (SCD) in childhood and adolescence: a review.Br J Clin Psychol.1993;32 (pt 3):271280.
  49. Weaver MF,Bond DS,Arnold BL,Waterhouse E,Towne A.Aberrant drug‐taking behaviors and headache: patient versus physician report.Am J Health Behav.2006;30(5):475482.
  50. Ballas SK.Current issues in sickle cell pain and its management [review].Hematology Am Soc Hematol Educ Program.2007;97105.
  51. Meehan WJ,Adelman SA,Rehman Z,Khoromi S.Opioid Abuse. Available at: http://www.emedicine.com/med/topic1673.htm. Updated April 18, 2006. Accessed August 23,2006.
  52. Savage SR.Assessment for addiction in pain‐treatment settings.Clin J Pain.2002;18(4 suppl):S28S38.
  53. Bohnert AS,Valenstein M,Bair MJ,Ganoczy D,McCarthy JF,Ilgen MA,Blow FC.Association between opioid prescribing patterns and opioid overdose‐related deaths.JAMA.2011;305(13):13151321.
  54. Payne R.American Pain Society workshop on the management of sickle cell pain.Saint Louis, MO;1990.
  55. Vichinsky EP,Johnson R,Lubin BH.Multidisciplinary approach to pain management in sickle cell disease.Am J Pediatr Hematol Oncol.1982;4:328333.
  56. Brozovic M,Davies S,Yardumian A,Bellingham A,Marsh G,Stephens AD.Pain relief in sickle cell crisis [letter].Lancet.1986;2:624625.
  57. Elander J,Lusher J,Bevan D,Telfer P,Burton B.Understanding the causes of problematic pain management in sickle cell disease: evidence that pseudoaddiction plays a more important role than genuine analgesic dependence.J Pain Symptom Manage.2004;27(2):156169.
  58. Ballas SK,Smith ED.Red blood cell changes during the evolution of the sickle cell painful crisis.Blood.1992;79:21542163.
  59. Brandow AM,Brousseau DC,Panepinto JA.Postdischarge pain, functional limitations and impact on caregivers of children with sickle cell disease treated for painful events.Br J Haematol.2009;144(5):782788.
  60. Ballas SK,Lusardi M.Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance.Am J Hematol.2005;79:1725.
  61. Brousseau DC,Owens PL,Mosso AL,Panepinto JA,Steiner CA.Acute care utilization and rehospitalizations for sickle cell disease.JAMA.2010;303(13):12881294.
  62. Todd KH,Funk KG,Funk JP, et al.Clinical significance of reported changes in pain severity.Ann Emerg Med.1996;27:485489.
  63. Lopez B,Flenders P,Davis‐Moon L,Corbin T,Ballas S.Clinically significant differences in visual analogue scale in acute vasoocclusive sickle cell crisis.Hemoglobin.2007;31:427432.
  64. Hansson P,Backonja M,Bouhassira D.Usefulness and limitations of quantitative sensory testing: clinical and research application in neuropathic pain states.Pain.2007;129:256259.
Article PDF
Issue
Journal of Hospital Medicine - 6(5)
Page Number
297-303
Legacy Keywords
hematology, pain, multidisciplinary care, quality improvement, sickle cell disease
Sections
Article PDF
Article PDF

Severe, disabling pain, often requiring opioids, is the most common medical presentation for children and adults with sickle cell disease (SCD), an autosomal recessive red blood cell disorder affecting those of African, Mediterranean, and Asian descent.1, 2 A genetically controlled hemoglobin alteration impairs oxygen binding, and enables polymerization of deoxy‐hemoglobin, resulting in, classically, sickle‐shaped erythrocytes3 and a complex cascade of ischemia and vaso‐occlusion in the microcirculation.4, 5

Dramatic gains in the treatment of SCD in childhood have resulted in markedly improved survival through adulthood.68 Thus, the need for adult SCD care is relatively new and rapidly growing. In 2005, approximately 70% of the nearly 80,000 US SCD hospitalizations occurred in adults versus children (Table 1). These hospitalizations occurred in the context of a poorly coordinated American health care system,9 despite the hopes raised by the Patient‐Centered Medical Home10 and the Chronic Care Model.11

Adult and Pediatric Admissions for Sickle Cell Disease,* 2005 and 2008
2005 2008
Total No. of Discharges LOS Total No. of Discharges LOS
  • NOTE: Data extracted from the Healthcare Cost and Utilization Project database, http://www.ahrq.gov/data/hcup/.

  • Abbreviations: LOS, length of stay in days (mean).

  • International Classification of Diseases, Clinical Modification (ICD‐CM) principal diagnosis code(s) 282.60, 282.61, 282.62, 282.63, 282.64, 282.69.

All discharges 79,187 100.00% 5.3 70,121 100.00% 5.4
Age group <1 996 1.26% 2.5 513 0.73% 2.7
1‐17 23,134 29.21% 3.9 13,754 19.62% 3.8
18‐44 48,168 60.83% 6 48,021 68.48% 5.8
45‐64 6,527 8.24% 6 7,543 10.76% 5.6
65‐84 281 0.35% 6.4 221 0.32% 5.6
Missing 81 0.10% 4 70 0.10% 3.0

Adults with SCD are vulnerable both because they are usually members of racial and ethnic minority groups, and because they have a Food and Drug Administration (FDA)‐defined orphan disease.12 They often do not receive the only FDA‐approved medication for SCD, life‐saving hydroxyurea,13 recommended for adults with homozygous sickle cell anemia (Hb SS) and sickle‐othalassemia (Hb SoThal).14 Young adults often fail to experience a smooth transition of care from children's hospitals, falling into a medical abyss.15

Therefore, increasingly, hospitalists are managing adults with SCD, rather than adult hematologyoncology, pain, or palliative care specialists. Adults with SCD experience negative opinions, bilateral lack of trust, and conflict in the doctorpatient relationship, frequently cited in studies of SCD adults and providers in the literature.16, 17

Evidence Base

General guidelines for SCD management have been published by the National Institutes of Health (NIH)18 and the Agency for Healthcare Policy and Research.19 But one of us (K.L.H.) found evidence lacking with regard to SCD pain management.20 Published guidelines on general pain management, such as the World Health Organization's Analgesic Ladder,21 do not address SCD. A Cochrane Review of pain management in SCD found only 9 randomized controlled trials, all with small numbers of patients, addressing acute SCD pain only.22 As well, American and British consensus SCD pain guidelines23, 24 admit, and subsequent publications emphasize,25, 26 the lack of evidence for what to do or not do for SCD pain management. At least 1 well‐done summary of the SCD evidence base intended for hospitalists has been published, but it focuses on management of issues other than pain.27

Motivations and Fears

It is not surprising then that hospitalists may bring great fear and apprehension with them into their care of SCD patients. One of us (W.R.S.), a general internist, has been called by his own and 3 other academic medical centers, 2 with active Federally‐funded SCD research programs, to address the problems of high‐utilizing adults with SCD, including counseling hospitalists frustrated with the management of pain in these patients.

Hospitalists may be motivated to provide efficient inpatient management (Table 2), and be aware of pain as the primary symptom of SCD inpatients. But they may carry knowledge gaps and biases into their relationships with SCD inpatients. They may fear opioid administration (opiophobia), loss of licensure or governmental reprisals because of high‐dose prescription of opioids, or may believe that SCD patients are more often addicted than most.17, 28 Consequently, more troublesome hospital stays may occur when patients are not rapidly and adequately titrated to appropriate analgesic doses, or when unnecessary deleterious side effects result from opioid and other analgesics. We therefore offer answers to frequently asked questions (FAQs) about pain management by hospitalists caring for adults with SCD. We address FAQs arising during the prototypical situationa patient with SCD admitted for a painful exacerbation, and little or no acute comorbidity. We refer the reader to the aforementioned articles and guidelines to address other treatment issues in adults with SCD.

General Motivations and Principles of Efficient Inpatient Sickle Cell Pain Management, and the Obstacles to Inpatient Care of Sickle Cell Disease
Principle Obstacles
  • Abbreviations: DEA, Drug Enforcement Administration; ED, emergency department.

Make appropriate management handoffs for patients coming from the ED to promote continuity of care and shorten hospitalization Poor information systems and poor handoffs/continuity from ED management to hospital management
Get as much preexisting information about the patient as possible to inform acute care, avoid returns or further hospitalization Patient may have no primary care physician or may underutilize primary care
Patient may misuse ED and hospital (as primary care source)
Provide rapid and adequate analgesia Ignorance of the differences between tolerance, physical dependence, addiction, and pseudoaddiction
No specific data on pharmacodynamics of opioid analgesics in sickle cell disease
Don't lose licensure or arouse regulatory suspicion about prescribing patterns Ignorance of DEA monitoring and laws governing appropriate vs inappropriate prescribing of opioids
Get the patient discharged as soon as medically appropriate Difficulty assessing pain quality and intensity
Difficulty assessing/avoiding side effects of analgesics
Difficulty determining appropriateness/timing of changes in analgesic dosing, discharge planning
Make appropriate handoffs with the patient's usual source of continuity care (provide that source when necessary) to avoid returns or further hospitalization Patient may have no primary care physician or may underutilize primary care physician
Patient may be mis/underprescribed analgesics by primary care physician
Define and maintain appropriate roles for hospitalists vs physicians with sickle cell training, pain specialists, or other specialists Inadequate adult system of care for sickle cell disease (no/paucity of specialty care)
Multiple prescribers of opioids
Providers unwilling to care for or prescribe opioids for sickle cell patients

FAQS

  • Is there any objective way to tell when SCD patients really are in a crisis?

    Although the term crisis is used as if it were an objectively definable biological entity, no one has proposed a standard definition of a crisis based on pain intensity level, clinical features, or biomarkers. Measures of vaso‐occlusion are correlated with ischemic pain, including pain that is often called a crisis.2932 However, neither ischemic pain from SCD, nor the underlying vaso‐occlusive cascade that is associated with this pain, is a sudden, present‐or‐absent phenomenon. Instead, these are continua that can be measured using pain scales or various biomarkers.

    There is, however, correlative evidence of the intensity of SCD pain associated with various distinctive health states (admitted/not admitted, in crisis/not in crisis). The most visible measure of a crisis, health care utilization, was a strong predictor of mortality in the Cooperative Study of Sickle Cell Disease. Patients with 3 or more admissions per year had a lower 5‐year survival rate.33 In contrast, crisis in the landmark Pain in Sickle Cell Epidemiology Study (PiSCES) was self‐defined by patients.34 Despite being in pain on over half of their days on average, and despite a third of patients being in pain daily, most pain in PiSCES was not considered a crisis, and less than 5% of patients' days were spent in emergency departments (EDs) or hospitals. Ambulatory pain intensity reports were correlated with opioid use.35 A substantial minority (35%) of PiSCES patients made at least 3 ED visits per year. However, these high ED utilizers had worse laboratory values, more pain, more distress, and a lower quality of life.36

    Importantly, sometimes adults with SCD may have severe comorbidities which may not be addressed or may be mistakenly managed as an acute vaso‐occlusive episode without further investigation or timely specialist consultation. Although pain is primarily the individual's chief complaint, any potential relationship between the presence of medical comorbidities and pain should be examined when patients are admitted.

  • How can one know when opioid dosages should be changed, or when SCD pain is appropriately controlled to allow discharge?

    We recommend, as a standard of care, that SCD pain assessment and pain therapy be interwoven, despite a systematic review finding no evidence that directly linked the timing, frequency, or method of pain assessment with outcomes or safety in medical inpatients, and concluding that the safety and effectiveness of patient‐controlled analgesia (PCA) in medical patients had not been adequately studied.37 Hospitalists should focus the first 24 hours of inpatient SCD pain management on cycles of recurrent pain assessment and opioid dose titration as frequently as every 1 to 2 hours, to assure safe and rapidly efficacious analgesia. Pain intensity, duration, and character should be assessed directly. Intensity is often assessed using a visual analog scale (VAS) or numeric rating scale.38 Treating physicians should themselves directly assess pain during discussions of therapy with the patient, even though some assessment usually is done in hospitals during each nursing shift. Pain and pain relief can be assessed indirectly by monitoring opioid use.

    We recommend PCA for inpatients with SCD, administered as an intermittent demand dose (patient must push a button) of opioid, with or without a background opioid constant infusion.39 We usually set the interval between doses, or lockout, to 6 to 10 minutes. Both the lockout and the sedation from delivered doses prevent patients from pushing the demand button repetitively to the point of overdose. Use of a low‐level constant infusion (basal) may sustain pain control during times when the patient is asleep, avoiding recrudescent pain and lost ground due to inadequate analgesia during rest. Alternatively, long‐acting oral opioids may be continued if already used at home, or newly introduced to provide adequate baseline pain control which is augmented by the demand dosing. Most PCA pumps monitor hourly opioid dose demand (number of pushes), as well as hourly doses delivered. Both hourly opioid dose demand and hourly dose‐per‐demand ratio are measures of PCA efficacy or futility. Pumps record this data, and can be interrogated at the patient's bedside for up to several days of prior use. Physicians should combine pump interrogation with direct pain assessment to guide demand‐dose titration. Demand doses should be increased to 1.5 to 2 times the previous demand dose after several hours of failed reduction of pain intensity and duration, and/or persistently futile dose‐per‐demand ratios.

    PCA interrogation is also useful for conversion of parenteral opioids to oral opioids, as well as to guide the recommendation for discharge home. After the first 24‐48 hours of up‐titration, if opioid dose demand decreases concordantly with pain frequency and intensity, the demand dose may be safely decreased, and eventually daily PCA requirements may be summed and converted to oral medication using standard opioid dose conversion tables. At this point, physicians may use single measures or daily averages of directly assessed pain.

    Routine PCA use in SCD is backed by some evidence.40, 41 But we find it important that patients be taught and encouraged to use the demand feature of PCA. Still, for various reasons, some patients do not use PCA pumps well. Discordant or unreliable assessments (eg, high pain intensity but low‐opioid demand doses during the same interval) may result, and PCA potentially may fail as a dosing strategy. Management is more difficult for these patients. One alternative dosing strategy is prescription of scheduled doses of a short‐acting opioid, attaching to each dose the order, patient may refuse. This is different than dosing as needed, and allows counts of dose refusals over an interval, analogous to PCA pump interrogation.

  • How much is too much opioid? Should one rely on side effects, or on requests for medicine, or is there a ceiling dose?

    Addictionologists, pain specialists, oncologists, those involved in hospice care, and some hematologists caring for SCD patients agree that, in general, there should be no a priori dose limitations imposed on opioid prescribing for acute pain. Instead, titration of dose of opioid to pain relief is a central principle of acute pain management. Experts also agree that particular opioids carry particular side effects which warrant dose limitation, adjustments, or avoidance of that opioid altogether. A summary of opioids commonly used in SCD, along with warnings and implied dose limitations is found in Table 3.

    For safety, it is important to assess the history of prior opioid use to recognize a patient who is not tolerant to opioids (see below, FAQ 4), to avoid mistakenly overdosing a patient using doses often required by tolerant patients. In lieu of a pre‐written, individualized opioid dosing plan in place for the patient, the patient may be the best source of information regarding preferred medication and tolerated doses.

    The reader is referred to standard texts for a description of opioids, their pharmacokinetics and pharmacodynamics, and their addictive and abuse potential. The side‐effect profile of opioids is well‐known: nausea, vomiting, and itching frequently occur; hallucinations, respiratory suppression, and myoclonus occur infrequently.42 Meperidine may more readily cause central nervous system (CNS) dysfunction, including seizures, as compared to other commonly used opioids, because of its toxic metabolite nor‐meperidine. Use of meperidine is often avoided, especially use via PCA.43 Methadone may cause dysrhythmias, specificially corrected Q‐T interval (QTc) prolongation and torsades de pointes on an electrocardiogram, in doses above 200 mg per day.44 Some recommend baseline and yearly electrocardiogram monitoring when giving methadone chronically.

    Recognizing the potential dangers of opioids, it is also reasonable to look for opioid‐sparing analgesic strategies. Non‐opioid analgesics such as ketorolac45 and adjuvants such as ketamine46 that are opioid‐sparing should be considered whenever feasible. Complementary and alternative therapies such as transcutaneous electrical nerve stimulation (TENS)47 have less evidence of effectiveness, but have limited risks and may be of use for some individuals.

  • What are the major signs of substance abuse (opioids, street drugs) in SCD patients already on opioids, and can a hospitalist judge those signs acutely and intervene appropriately?

    Reports of underprescription of opioids in SCD have cited physician fear of abuse and addiction.48 A recent informal poll of adult sickle cell providers suggests policies vary on how potential abuse is monitored in ambulatory sickle cell patients. We note that physicians, especially upon meeting a patient for the first time, may be unable to reliably judge whether that patient is abusing opioids or street drugs. Both false‐positive and false‐negative diagnoses may be made.49 Repetitive reports of lost or stolen prescriptions or pill bottles, receipt of prescriptions from multiple providers, or repeated requests for early refills increase the suspicion of misuse or abuse, but are indirect evidence. Urine and serum monitoring may be useful, but may give incorrect information if misinterpreted or not conducted frequently enough to improve sensitivity.50

    It is important to distinguish between tolerance, the decreased analgesic response over time to repeated doses of the same drug; physical dependence, the production of withdrawal upon abrupt discontinuation of an opioid agonist or administration of an antagonist; and addiction, the psychological dependence upon opioids. Tolerance may be misperceived as true addiction. Its earliest symptom is shortening of the duration of effective analgesia. In contrast, addiction may be manifested by dose escalation in the absence of an increased pain stimulus, or by use of opioids for purposes other than pain relief.51 These are not easily distinguished during a single patient encounter.

    SCD patients' requests for specific opioid medications in specific doses, should not be taken as evidence of past or current abuse, but rather evidence of a well‐informed, self‐managing patient. Adults with SCD are clearly expected to be very knowledgeable about and tolerant to opioids if they have had a life of pain as a child, and will require higher doses of opioids than other patients treated by most hospitalists. The issue of medication abuse may be best handled in the ambulatory setting. Whenever possible, hospitalists should not rely only on data from the acute care setting to manage patients. Ambulatory providers may conduct random, unannounced urine and/or serum testing, as part of an opioid prescribing agreement that is written and filed in the patient's chart. Assays for prescribed opioids (especially long‐acting agents), as well as assays for common drugs of abuse, should be conducted. Comanagement with an addictionologist, psychiatrist, or psychologist should be considered in individuals suspected of opioid abuse.

    We do not suggest routine urine drug test monitoring of all SCD patients unless routine monitoring is done as a policy for all patients on opioids. Though the prevalence of addiction may be higher in subpopulations of patients with pain,52 and though prescription of opioids, prescription drug abuse, and accidental deaths from prescribed opioids have risen exponentially in the last several years,53 in our experience and in the published literature, drug misuse/abuse among SCD patients is no worse than among patients with other illnesses.5456 However, pseudoaddiction, the appropriate seeking of needed opioids from multiple physicians because of uncontrolled pain and opioid underprescription, may well be prevalent in SCD,57 and may be mistaken for true addiction.

  • How can patients' readiness for discharge be assessed? What can be done for the patient who has lengthy and/or multiple hospitalizations or frequent ED visits?

    The appropriate time for discharge in most patients is when they can manage their pain at home with oral opioids or less. Often, patients do not improve even after a few days of inpatient therapy.58 A typical pain episode may last much longer than the 6‐day average US hospital length of stay for a diagnosis of sickle cell crisis among 18‐44 year olds (Table 1).59 Patients may return and be readmitted.60, 61 But in the best cases, pain resolves or reverts to a usual chronic intensity level. As described in FAQ 2, daily or more frequent pain assessment is a bedrock for making discharge decisions. Patients well‐experienced in the use of pain intensity scales can report their usual pain intensity at home, and how close they are to their baseline pain intensity. Simply asking patients, Are you ready for discharge? is appropriate and may yield a surprising positive response. In a recent inpatient trial of PCA (manuscript in preparation), adult patients were admitted with a minimum pain intensity of 45 mm on a 100 mm horizontal VAS scale after treatment in the ED, and mean pain intensity of 76 mm 10 mm. All adults in this study were discharged with pain that was clinically significantly lower. Researchers have found a VAS change of 13.5 mm to be the minimum clinically significant change62 during treatment of vaso‐occlusive crisis.63

    Unremitting pain despite appropriate titration of opioids and prolonged hospital stays suggests the need for comprehensive evaluation for medical and psychosocial comorbidities, as is done for other patients with chronic pain syndromes. If not already done, discussion with the patient's primary care provider may reveal factors impacting on persistent pain. Consultation with a hematologist, pain or palliative care specialist, or other provider familiar with SCD may prove helpful. Implementation of adjuvant therapies as discussed in FAQ 3 and adding long‐acting oral opioids to continue postdischarge may also help. Hyperalgesia, or heightened sensitivity to pain, is normal after acute tissue injury, but is now suspected in SCD as a long‐term neuropathic pain syndrome, as a consequence either of repeated painful crises or of chronic opioid therapy.2 Only some centers have specialists qualified to test for and diagnose neuropathic pain.64

    Discharge planning should include identification of a source of outpatient follow‐up. Opioids prescribed at discharge should be sufficient to last at least until the first outpatient appointment, to avoid repeated ED or hospital visits. Communication with a primary care provider at discharge can enhance successful care transition. Otherwise, for patients without established providers, social workers and others may address barriers to follow‐up that frustrate both patient and provider.

Table of Opioids, Frequency of Use, and Special Considerations in SCD
Opioid Used Frequently (>20% of Patients) How Used Unique Side Effects and/or Dose Limitations
  • Abbreviations: SCD, sickle cell disease; QTc, corrected Q‐T interval on electrocardiogram.

Short‐acting
Codeine No Inpatient, parenteral; Ambulatory, oral
Oxycodone Yes Most commonly used ambulatory opioid
Morphine Yes Most commonly used inpatient opioid
Hydromorphone Yes Inpatient more than ambulatory
Fentanyl No Inpatient, parenteral Short‐acting
Hydrocodone No Ambulatory
Meperedine No Avoided Unpredictable seizure, coma, death
Propoxyphene No Ambulatory
Tramadol No Ambulatory
Long‐acting
Oxycodone No Ambulatory and as an oral basal in inpatients Abuse potential from capsule manipulation
Morphine Yes Ambulatory and as an oral basal in inpatients; most commonly used long‐acting opioid
Methadone No Ambulatory and as an oral basal in inpatients Dose‐dependent prolongation of QTc, torsades de pointes
Fentanyl No Ambulatory and as a transdermal basal in inpatients Abuse potential from transdermal patch manipulation

Support for Hospitalists Managing Adults With Sickle Cell Disease

Beside the general advice on pain management in SCD mentioned above or found in the bibliography of this article, at long last, a group of adult practitioners skilled in the care of SCD has formed nationally. The Sickle Cell Adult Provider Network [http://www.scapn.net] provides non‐binding advice and support to its members via an e‐mail listserve. Topics often include pain management. This advice fills a vacuum created by the lack of evidence‐based guidelines.

Ultimately, evidence and updated guidelines will be the best support for hospitalists and others managing pain in SCD. The hope is that SCD will receive the attention it deserves, so that practitioners and patients alike do not suffer continued pain from this disease or its management.

Severe, disabling pain, often requiring opioids, is the most common medical presentation for children and adults with sickle cell disease (SCD), an autosomal recessive red blood cell disorder affecting those of African, Mediterranean, and Asian descent.1, 2 A genetically controlled hemoglobin alteration impairs oxygen binding, and enables polymerization of deoxy‐hemoglobin, resulting in, classically, sickle‐shaped erythrocytes3 and a complex cascade of ischemia and vaso‐occlusion in the microcirculation.4, 5

Dramatic gains in the treatment of SCD in childhood have resulted in markedly improved survival through adulthood.68 Thus, the need for adult SCD care is relatively new and rapidly growing. In 2005, approximately 70% of the nearly 80,000 US SCD hospitalizations occurred in adults versus children (Table 1). These hospitalizations occurred in the context of a poorly coordinated American health care system,9 despite the hopes raised by the Patient‐Centered Medical Home10 and the Chronic Care Model.11

Adult and Pediatric Admissions for Sickle Cell Disease,* 2005 and 2008
2005 2008
Total No. of Discharges LOS Total No. of Discharges LOS
  • NOTE: Data extracted from the Healthcare Cost and Utilization Project database, http://www.ahrq.gov/data/hcup/.

  • Abbreviations: LOS, length of stay in days (mean).

  • International Classification of Diseases, Clinical Modification (ICD‐CM) principal diagnosis code(s) 282.60, 282.61, 282.62, 282.63, 282.64, 282.69.

All discharges 79,187 100.00% 5.3 70,121 100.00% 5.4
Age group <1 996 1.26% 2.5 513 0.73% 2.7
1‐17 23,134 29.21% 3.9 13,754 19.62% 3.8
18‐44 48,168 60.83% 6 48,021 68.48% 5.8
45‐64 6,527 8.24% 6 7,543 10.76% 5.6
65‐84 281 0.35% 6.4 221 0.32% 5.6
Missing 81 0.10% 4 70 0.10% 3.0

Adults with SCD are vulnerable both because they are usually members of racial and ethnic minority groups, and because they have a Food and Drug Administration (FDA)‐defined orphan disease.12 They often do not receive the only FDA‐approved medication for SCD, life‐saving hydroxyurea,13 recommended for adults with homozygous sickle cell anemia (Hb SS) and sickle‐othalassemia (Hb SoThal).14 Young adults often fail to experience a smooth transition of care from children's hospitals, falling into a medical abyss.15

Therefore, increasingly, hospitalists are managing adults with SCD, rather than adult hematologyoncology, pain, or palliative care specialists. Adults with SCD experience negative opinions, bilateral lack of trust, and conflict in the doctorpatient relationship, frequently cited in studies of SCD adults and providers in the literature.16, 17

Evidence Base

General guidelines for SCD management have been published by the National Institutes of Health (NIH)18 and the Agency for Healthcare Policy and Research.19 But one of us (K.L.H.) found evidence lacking with regard to SCD pain management.20 Published guidelines on general pain management, such as the World Health Organization's Analgesic Ladder,21 do not address SCD. A Cochrane Review of pain management in SCD found only 9 randomized controlled trials, all with small numbers of patients, addressing acute SCD pain only.22 As well, American and British consensus SCD pain guidelines23, 24 admit, and subsequent publications emphasize,25, 26 the lack of evidence for what to do or not do for SCD pain management. At least 1 well‐done summary of the SCD evidence base intended for hospitalists has been published, but it focuses on management of issues other than pain.27

Motivations and Fears

It is not surprising then that hospitalists may bring great fear and apprehension with them into their care of SCD patients. One of us (W.R.S.), a general internist, has been called by his own and 3 other academic medical centers, 2 with active Federally‐funded SCD research programs, to address the problems of high‐utilizing adults with SCD, including counseling hospitalists frustrated with the management of pain in these patients.

Hospitalists may be motivated to provide efficient inpatient management (Table 2), and be aware of pain as the primary symptom of SCD inpatients. But they may carry knowledge gaps and biases into their relationships with SCD inpatients. They may fear opioid administration (opiophobia), loss of licensure or governmental reprisals because of high‐dose prescription of opioids, or may believe that SCD patients are more often addicted than most.17, 28 Consequently, more troublesome hospital stays may occur when patients are not rapidly and adequately titrated to appropriate analgesic doses, or when unnecessary deleterious side effects result from opioid and other analgesics. We therefore offer answers to frequently asked questions (FAQs) about pain management by hospitalists caring for adults with SCD. We address FAQs arising during the prototypical situationa patient with SCD admitted for a painful exacerbation, and little or no acute comorbidity. We refer the reader to the aforementioned articles and guidelines to address other treatment issues in adults with SCD.

General Motivations and Principles of Efficient Inpatient Sickle Cell Pain Management, and the Obstacles to Inpatient Care of Sickle Cell Disease
Principle Obstacles
  • Abbreviations: DEA, Drug Enforcement Administration; ED, emergency department.

Make appropriate management handoffs for patients coming from the ED to promote continuity of care and shorten hospitalization Poor information systems and poor handoffs/continuity from ED management to hospital management
Get as much preexisting information about the patient as possible to inform acute care, avoid returns or further hospitalization Patient may have no primary care physician or may underutilize primary care
Patient may misuse ED and hospital (as primary care source)
Provide rapid and adequate analgesia Ignorance of the differences between tolerance, physical dependence, addiction, and pseudoaddiction
No specific data on pharmacodynamics of opioid analgesics in sickle cell disease
Don't lose licensure or arouse regulatory suspicion about prescribing patterns Ignorance of DEA monitoring and laws governing appropriate vs inappropriate prescribing of opioids
Get the patient discharged as soon as medically appropriate Difficulty assessing pain quality and intensity
Difficulty assessing/avoiding side effects of analgesics
Difficulty determining appropriateness/timing of changes in analgesic dosing, discharge planning
Make appropriate handoffs with the patient's usual source of continuity care (provide that source when necessary) to avoid returns or further hospitalization Patient may have no primary care physician or may underutilize primary care physician
Patient may be mis/underprescribed analgesics by primary care physician
Define and maintain appropriate roles for hospitalists vs physicians with sickle cell training, pain specialists, or other specialists Inadequate adult system of care for sickle cell disease (no/paucity of specialty care)
Multiple prescribers of opioids
Providers unwilling to care for or prescribe opioids for sickle cell patients

FAQS

  • Is there any objective way to tell when SCD patients really are in a crisis?

    Although the term crisis is used as if it were an objectively definable biological entity, no one has proposed a standard definition of a crisis based on pain intensity level, clinical features, or biomarkers. Measures of vaso‐occlusion are correlated with ischemic pain, including pain that is often called a crisis.2932 However, neither ischemic pain from SCD, nor the underlying vaso‐occlusive cascade that is associated with this pain, is a sudden, present‐or‐absent phenomenon. Instead, these are continua that can be measured using pain scales or various biomarkers.

    There is, however, correlative evidence of the intensity of SCD pain associated with various distinctive health states (admitted/not admitted, in crisis/not in crisis). The most visible measure of a crisis, health care utilization, was a strong predictor of mortality in the Cooperative Study of Sickle Cell Disease. Patients with 3 or more admissions per year had a lower 5‐year survival rate.33 In contrast, crisis in the landmark Pain in Sickle Cell Epidemiology Study (PiSCES) was self‐defined by patients.34 Despite being in pain on over half of their days on average, and despite a third of patients being in pain daily, most pain in PiSCES was not considered a crisis, and less than 5% of patients' days were spent in emergency departments (EDs) or hospitals. Ambulatory pain intensity reports were correlated with opioid use.35 A substantial minority (35%) of PiSCES patients made at least 3 ED visits per year. However, these high ED utilizers had worse laboratory values, more pain, more distress, and a lower quality of life.36

    Importantly, sometimes adults with SCD may have severe comorbidities which may not be addressed or may be mistakenly managed as an acute vaso‐occlusive episode without further investigation or timely specialist consultation. Although pain is primarily the individual's chief complaint, any potential relationship between the presence of medical comorbidities and pain should be examined when patients are admitted.

  • How can one know when opioid dosages should be changed, or when SCD pain is appropriately controlled to allow discharge?

    We recommend, as a standard of care, that SCD pain assessment and pain therapy be interwoven, despite a systematic review finding no evidence that directly linked the timing, frequency, or method of pain assessment with outcomes or safety in medical inpatients, and concluding that the safety and effectiveness of patient‐controlled analgesia (PCA) in medical patients had not been adequately studied.37 Hospitalists should focus the first 24 hours of inpatient SCD pain management on cycles of recurrent pain assessment and opioid dose titration as frequently as every 1 to 2 hours, to assure safe and rapidly efficacious analgesia. Pain intensity, duration, and character should be assessed directly. Intensity is often assessed using a visual analog scale (VAS) or numeric rating scale.38 Treating physicians should themselves directly assess pain during discussions of therapy with the patient, even though some assessment usually is done in hospitals during each nursing shift. Pain and pain relief can be assessed indirectly by monitoring opioid use.

    We recommend PCA for inpatients with SCD, administered as an intermittent demand dose (patient must push a button) of opioid, with or without a background opioid constant infusion.39 We usually set the interval between doses, or lockout, to 6 to 10 minutes. Both the lockout and the sedation from delivered doses prevent patients from pushing the demand button repetitively to the point of overdose. Use of a low‐level constant infusion (basal) may sustain pain control during times when the patient is asleep, avoiding recrudescent pain and lost ground due to inadequate analgesia during rest. Alternatively, long‐acting oral opioids may be continued if already used at home, or newly introduced to provide adequate baseline pain control which is augmented by the demand dosing. Most PCA pumps monitor hourly opioid dose demand (number of pushes), as well as hourly doses delivered. Both hourly opioid dose demand and hourly dose‐per‐demand ratio are measures of PCA efficacy or futility. Pumps record this data, and can be interrogated at the patient's bedside for up to several days of prior use. Physicians should combine pump interrogation with direct pain assessment to guide demand‐dose titration. Demand doses should be increased to 1.5 to 2 times the previous demand dose after several hours of failed reduction of pain intensity and duration, and/or persistently futile dose‐per‐demand ratios.

    PCA interrogation is also useful for conversion of parenteral opioids to oral opioids, as well as to guide the recommendation for discharge home. After the first 24‐48 hours of up‐titration, if opioid dose demand decreases concordantly with pain frequency and intensity, the demand dose may be safely decreased, and eventually daily PCA requirements may be summed and converted to oral medication using standard opioid dose conversion tables. At this point, physicians may use single measures or daily averages of directly assessed pain.

    Routine PCA use in SCD is backed by some evidence.40, 41 But we find it important that patients be taught and encouraged to use the demand feature of PCA. Still, for various reasons, some patients do not use PCA pumps well. Discordant or unreliable assessments (eg, high pain intensity but low‐opioid demand doses during the same interval) may result, and PCA potentially may fail as a dosing strategy. Management is more difficult for these patients. One alternative dosing strategy is prescription of scheduled doses of a short‐acting opioid, attaching to each dose the order, patient may refuse. This is different than dosing as needed, and allows counts of dose refusals over an interval, analogous to PCA pump interrogation.

  • How much is too much opioid? Should one rely on side effects, or on requests for medicine, or is there a ceiling dose?

    Addictionologists, pain specialists, oncologists, those involved in hospice care, and some hematologists caring for SCD patients agree that, in general, there should be no a priori dose limitations imposed on opioid prescribing for acute pain. Instead, titration of dose of opioid to pain relief is a central principle of acute pain management. Experts also agree that particular opioids carry particular side effects which warrant dose limitation, adjustments, or avoidance of that opioid altogether. A summary of opioids commonly used in SCD, along with warnings and implied dose limitations is found in Table 3.

    For safety, it is important to assess the history of prior opioid use to recognize a patient who is not tolerant to opioids (see below, FAQ 4), to avoid mistakenly overdosing a patient using doses often required by tolerant patients. In lieu of a pre‐written, individualized opioid dosing plan in place for the patient, the patient may be the best source of information regarding preferred medication and tolerated doses.

    The reader is referred to standard texts for a description of opioids, their pharmacokinetics and pharmacodynamics, and their addictive and abuse potential. The side‐effect profile of opioids is well‐known: nausea, vomiting, and itching frequently occur; hallucinations, respiratory suppression, and myoclonus occur infrequently.42 Meperidine may more readily cause central nervous system (CNS) dysfunction, including seizures, as compared to other commonly used opioids, because of its toxic metabolite nor‐meperidine. Use of meperidine is often avoided, especially use via PCA.43 Methadone may cause dysrhythmias, specificially corrected Q‐T interval (QTc) prolongation and torsades de pointes on an electrocardiogram, in doses above 200 mg per day.44 Some recommend baseline and yearly electrocardiogram monitoring when giving methadone chronically.

    Recognizing the potential dangers of opioids, it is also reasonable to look for opioid‐sparing analgesic strategies. Non‐opioid analgesics such as ketorolac45 and adjuvants such as ketamine46 that are opioid‐sparing should be considered whenever feasible. Complementary and alternative therapies such as transcutaneous electrical nerve stimulation (TENS)47 have less evidence of effectiveness, but have limited risks and may be of use for some individuals.

  • What are the major signs of substance abuse (opioids, street drugs) in SCD patients already on opioids, and can a hospitalist judge those signs acutely and intervene appropriately?

    Reports of underprescription of opioids in SCD have cited physician fear of abuse and addiction.48 A recent informal poll of adult sickle cell providers suggests policies vary on how potential abuse is monitored in ambulatory sickle cell patients. We note that physicians, especially upon meeting a patient for the first time, may be unable to reliably judge whether that patient is abusing opioids or street drugs. Both false‐positive and false‐negative diagnoses may be made.49 Repetitive reports of lost or stolen prescriptions or pill bottles, receipt of prescriptions from multiple providers, or repeated requests for early refills increase the suspicion of misuse or abuse, but are indirect evidence. Urine and serum monitoring may be useful, but may give incorrect information if misinterpreted or not conducted frequently enough to improve sensitivity.50

    It is important to distinguish between tolerance, the decreased analgesic response over time to repeated doses of the same drug; physical dependence, the production of withdrawal upon abrupt discontinuation of an opioid agonist or administration of an antagonist; and addiction, the psychological dependence upon opioids. Tolerance may be misperceived as true addiction. Its earliest symptom is shortening of the duration of effective analgesia. In contrast, addiction may be manifested by dose escalation in the absence of an increased pain stimulus, or by use of opioids for purposes other than pain relief.51 These are not easily distinguished during a single patient encounter.

    SCD patients' requests for specific opioid medications in specific doses, should not be taken as evidence of past or current abuse, but rather evidence of a well‐informed, self‐managing patient. Adults with SCD are clearly expected to be very knowledgeable about and tolerant to opioids if they have had a life of pain as a child, and will require higher doses of opioids than other patients treated by most hospitalists. The issue of medication abuse may be best handled in the ambulatory setting. Whenever possible, hospitalists should not rely only on data from the acute care setting to manage patients. Ambulatory providers may conduct random, unannounced urine and/or serum testing, as part of an opioid prescribing agreement that is written and filed in the patient's chart. Assays for prescribed opioids (especially long‐acting agents), as well as assays for common drugs of abuse, should be conducted. Comanagement with an addictionologist, psychiatrist, or psychologist should be considered in individuals suspected of opioid abuse.

    We do not suggest routine urine drug test monitoring of all SCD patients unless routine monitoring is done as a policy for all patients on opioids. Though the prevalence of addiction may be higher in subpopulations of patients with pain,52 and though prescription of opioids, prescription drug abuse, and accidental deaths from prescribed opioids have risen exponentially in the last several years,53 in our experience and in the published literature, drug misuse/abuse among SCD patients is no worse than among patients with other illnesses.5456 However, pseudoaddiction, the appropriate seeking of needed opioids from multiple physicians because of uncontrolled pain and opioid underprescription, may well be prevalent in SCD,57 and may be mistaken for true addiction.

  • How can patients' readiness for discharge be assessed? What can be done for the patient who has lengthy and/or multiple hospitalizations or frequent ED visits?

    The appropriate time for discharge in most patients is when they can manage their pain at home with oral opioids or less. Often, patients do not improve even after a few days of inpatient therapy.58 A typical pain episode may last much longer than the 6‐day average US hospital length of stay for a diagnosis of sickle cell crisis among 18‐44 year olds (Table 1).59 Patients may return and be readmitted.60, 61 But in the best cases, pain resolves or reverts to a usual chronic intensity level. As described in FAQ 2, daily or more frequent pain assessment is a bedrock for making discharge decisions. Patients well‐experienced in the use of pain intensity scales can report their usual pain intensity at home, and how close they are to their baseline pain intensity. Simply asking patients, Are you ready for discharge? is appropriate and may yield a surprising positive response. In a recent inpatient trial of PCA (manuscript in preparation), adult patients were admitted with a minimum pain intensity of 45 mm on a 100 mm horizontal VAS scale after treatment in the ED, and mean pain intensity of 76 mm 10 mm. All adults in this study were discharged with pain that was clinically significantly lower. Researchers have found a VAS change of 13.5 mm to be the minimum clinically significant change62 during treatment of vaso‐occlusive crisis.63

    Unremitting pain despite appropriate titration of opioids and prolonged hospital stays suggests the need for comprehensive evaluation for medical and psychosocial comorbidities, as is done for other patients with chronic pain syndromes. If not already done, discussion with the patient's primary care provider may reveal factors impacting on persistent pain. Consultation with a hematologist, pain or palliative care specialist, or other provider familiar with SCD may prove helpful. Implementation of adjuvant therapies as discussed in FAQ 3 and adding long‐acting oral opioids to continue postdischarge may also help. Hyperalgesia, or heightened sensitivity to pain, is normal after acute tissue injury, but is now suspected in SCD as a long‐term neuropathic pain syndrome, as a consequence either of repeated painful crises or of chronic opioid therapy.2 Only some centers have specialists qualified to test for and diagnose neuropathic pain.64

    Discharge planning should include identification of a source of outpatient follow‐up. Opioids prescribed at discharge should be sufficient to last at least until the first outpatient appointment, to avoid repeated ED or hospital visits. Communication with a primary care provider at discharge can enhance successful care transition. Otherwise, for patients without established providers, social workers and others may address barriers to follow‐up that frustrate both patient and provider.

Table of Opioids, Frequency of Use, and Special Considerations in SCD
Opioid Used Frequently (>20% of Patients) How Used Unique Side Effects and/or Dose Limitations
  • Abbreviations: SCD, sickle cell disease; QTc, corrected Q‐T interval on electrocardiogram.

Short‐acting
Codeine No Inpatient, parenteral; Ambulatory, oral
Oxycodone Yes Most commonly used ambulatory opioid
Morphine Yes Most commonly used inpatient opioid
Hydromorphone Yes Inpatient more than ambulatory
Fentanyl No Inpatient, parenteral Short‐acting
Hydrocodone No Ambulatory
Meperedine No Avoided Unpredictable seizure, coma, death
Propoxyphene No Ambulatory
Tramadol No Ambulatory
Long‐acting
Oxycodone No Ambulatory and as an oral basal in inpatients Abuse potential from capsule manipulation
Morphine Yes Ambulatory and as an oral basal in inpatients; most commonly used long‐acting opioid
Methadone No Ambulatory and as an oral basal in inpatients Dose‐dependent prolongation of QTc, torsades de pointes
Fentanyl No Ambulatory and as a transdermal basal in inpatients Abuse potential from transdermal patch manipulation

Support for Hospitalists Managing Adults With Sickle Cell Disease

Beside the general advice on pain management in SCD mentioned above or found in the bibliography of this article, at long last, a group of adult practitioners skilled in the care of SCD has formed nationally. The Sickle Cell Adult Provider Network [http://www.scapn.net] provides non‐binding advice and support to its members via an e‐mail listserve. Topics often include pain management. This advice fills a vacuum created by the lack of evidence‐based guidelines.

Ultimately, evidence and updated guidelines will be the best support for hospitalists and others managing pain in SCD. The hope is that SCD will receive the attention it deserves, so that practitioners and patients alike do not suffer continued pain from this disease or its management.

References
  1. Serjeant GR.Sickle‐cell disease.Lancet.1997;350(9079):725302.
  2. Smith WR,Scherer M.Sickle‐cell pain: advances in epidemiology and etiology.Hematology Am Soc Hematol Educ Program.2010;409415. PMID: 21239827.
  3. Steinberg MH.Management of sickle cell disease.N Engl J Med.1999;340:10211030.
  4. Hebbel RP,Vercellotti G,Nath KA.A systems biology consideration of the vasculopathy of sickle cell anemia: the need for multi‐modality chemo‐prophylaxsis.Cardiovasc Hematol Disord Drug Targets.2009;9(4):271292.
  5. Conran N,Franco‐Penteado CF,Costa FF.Newer aspects of the pathophysiology of sickle cell disease vaso‐occlusion [review].Hemoglobin.2009;33(1):116.
  6. Davis H,Schoendorf KC,Gergen PJ,Moore RM.National trends in the mortality of children with sickle cell disease, 1968 through 1992.Am J Public Health.1997;87(8):13171322.
  7. Adams RJ,McKie VC,Hsu L, et al.Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography.N Engl J Med.1998;339:511.
  8. Adams RJ,Brambilla D.Optimizing primary stroke prevention in sickle cell anemia (STOP 2) trial investigators. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease.N Engl J Med.2005;353(26):27692778.
  9. Sandy LG.Homeostasis without reserve—the risk of health system collapse.N Engl J Med.2002;347(24):19711975.
  10. The Advanced Medical Home: A Patient‐Centered, Physician‐Guided Model of Health Care [Policy Monograph].Philadelphia, PA:American College of Physicians;2006.
  11. Wagner EH,Glasgow RE,Davis C,Bonomi AE,Provost L,McCulloch D.Quality improvement in chronic illness care: a collaborative approach.Jt Comm J Qual Improv.2001;27:6380.
  12. Definition of Disease Prevalence for Therapies Qualifying Under the Orphan Drug Act. Subpart C, Designation of an Orphan Drug. Sec. 316.20. Content and format of a request for orphan‐drug designation. Available at: http://www.fda.gov/orphan/designat/prevalence.html. Accessed September 3,2008.
  13. Steinberg MH,Barton F,Castro O, et al.Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment.JAMA.2003;289:16451651.
  14. Lanzkron S,Haywood C,Hassell KL,Rand C.Provider barriers to hydroxyurea use in adults with sickle cell disease: a survey of the Sickle Cell Disease Adult Provider Network.J Natl Med Assoc.2008;100(8):968973.
  15. Treadwell M,Telfair J,Gibson RW,Johnson S,Osunkwo I.Transition from pediatric to adult care in sickle cell disease: establishing evidence‐based practice and directions for research.Am J Hematol.2011;86(1):116120. PMID: 21061308.
  16. Bobo L,Miller ST,Smith WR,Elam JT,Rosmarin PC,Lancaster DJ.Health perceptions and medical care opinions of inner‐city adults with sickle cell disease or asthma compared with those of their siblings.South Med J.1989;82(1):912.
  17. Shapiro BS,Benjamin LJ,Payne R,Heidrich G.Sickle cell‐related pain: perceptions of medical practitioners.J Pain Symptom Manage.1997;14(3):168174.
  18. The Management of Sickle Cell Disease.4th ed. NIH Publication 2002–2117.Washington, DC:National Institutes of Health, National Heart, Lung, and Blood Institute, Division of Blood Diseases and Resources; June2002.
  19. Pain Management Guideline.Washington, DC:Agency for Health Care Policy and Research;1992.
  20. Lottenberg R,Hassell KL.An evidence‐based approach to the treatment of adults with sickle cell disease.Hematology Am Soc Hematol Educ Program.2005;5865.
  21. World Health Organization: Cancer Pain Relief.Geneva, Switzerland:WHO,1986.
  22. Dunlop RJ,Bennett KC.Pain management for sickle cell disease.Cochrane Database Syst Rev. April 19,2006;(2):CD003350.
  23. Benjamin L,Dampier C,Jacox A;for the Guideline Committee.Guidelines for the Management of Acute and Chronic Pain in Sickle Cell Disease. APS Clinical Practice Guideline Series, No 1.Glenview, IL:American Pain Society;1999.
  24. Rees D.Guidelines for the management of the acute painful crisis of sickle cell disease.Br J Haematol.2003;120:744752.
  25. Field JJ,Knight‐Perry JE,DeBaun MR.Acute pain in children and adults with sickle cell disease: management in the absence of evidence‐based guidelines.Curr Opin Hematol.2009;16(3):173178.
  26. Rosenblum A,Marsch LA,Joseph H,Portenoy RK.Opioids and the treatment of chronic pain: controversies, current status, and future directions.Exp Clin Psychopharmacol.2008;16(5):405416.
  27. Axon RN.A sickle cell primer.The Hospitalist.2006;10(10):3941.
  28. Hill CS.The barriers to adequate pain management with opioid analgesics.Semin Oncol.1993;20(2 suppl 1):15.
  29. Dampier C,Setty BN,Eggleston B,Brodecki D,O'Neal P,Stuart M.Vaso‐occlusion in children with sickle cell disease: clinical characteristics and biologic correlates.J Pediatr Hematol Oncol.2004;26:785790. PMID: 15591896.
  30. Graido‐Gonzalez E,Doherty JC,Bergreen EW,Organ G,Telfer M,McMillen MA.Plasma endothelin‐1, cytokine, and prostaglandin E2 levels in sickle cell disease and acute vaso‐occlusive sickle crisis.Blood.1998;92:25512555. PMID: 9746797.
  31. Michaels LA,Ohene‐Frempong K,Zhao H,Douglas SD.Serum levels of substance P are elevated in patients with sickle cell disease and increase further during vaso‐occlusive crisis.Blood.1998;92:31483151. PMID: 9787150.
  32. Landburg PP,Teerlink T,Muskiet FA,Duits AJ,Schnog JJ;for the CURAMA study group.Plasma concentrations of asymmetric dimethylarginine, an endogenous nitric oxide synthase inhibitor, are elevated in sickle cell patients but do not increase further during painful crisis.Am J Hematol. February 27,2008; PMID: 18383318
  33. Platt OS,Thorington BD,Brambilla DJ, et al.Pain in sickle cell disease: rates and risk factors.N Engl J Med.1991;325:1116.
  34. Smith WR,Bovbjerg VE,Penberthy LT, et al.Understanding pain and improving management of sickle cell disease: the PiSCES Study.J Natl Med Assoc.2005;97(2):183193.
  35. Smith WR,McClish DK,Penberthy LT, et al.Daily assessment of pain in adults with sickle cell disease.Ann Intern Med.2008;148(2):94101.
  36. Aisiku IP,Smith WR,McClish DK, et al.Comparisons of high versus low emergency department utilizers in sickle cell disease.Ann Emerg Med.2009;53(5):587593.
  37. Helfand M,Freeman M.Assessment and management of acute pain in adult medical inpatients: a systematic review.Pain Med.2009;10(7):11831199. PMID: 19818030.
  38. Wallenstein SL.Scaling clinical pain and pain relief. In: Bromm B, ed.Pain Measurement in Man: Neorephysiological Correlates of Pain.New York:Elsevier Science Publishers,1984:389396.
  39. Dubois M.Patient‐controlled analgesia for acute pain.Clin J Pain.1989;5(suppl 1):S8S15. PMID: 2520435.
  40. Melzer‐Lange MD,Walsh‐Kelly CM,Lea G,Hillery CA,Scott JP.Patient‐controlled analgesia for sickle pain crisis in pediatric emergency department.Pediatr Emerg Care.2004;20:24.
  41. Trentadue N,Kachoyeanos MK,Lea G.A comparison of two regimens of patient‐controlled analgesia for children with sickle cell disease.J Pediatr Nurs.1998;13:1519.
  42. Narcotic analgesics, 2002 update.The DAWN Report;2004.
  43. Seifert CF,Kennedy S.Meperidine is alive and well in the new millennium: evaluation of meperidine usage patterns and frequency of adverse drug reactions.Pharmacotherapy.2004;24:776783.
  44. Porter BP,Coyne PJ,Smith WR.Methadone‐related torsades de pointes in a sickle cell patient treated for chronic pain.Am J Hematol.2005;78(4):316317.
  45. de Franceschi L,Finco G,Vassanelli A,Zaia B,Ischia S,Corrocher R.A pilot study on the efficacy of ketorolac plus tramadol infusion combined with erythrocytapheresis in the management of acute severe vaso‐occlusive crises and sickle cell pain.Haematologica.2004;89(11):13891391.
  46. Zempsky WT,Loiselle KA,Corsi JM,Hagstrom JN.Use of low‐dose ketamine infusion for pediatric patients with sickle cell disease‐related pain: a case series.Clin J Pain.2010;26(2):163167.
  47. Wang WC,George SL,Wilimas JA.Transcutaneous electrical nerve stimulation treatment of sickle cell pain crises.Acta Haematol.1988;80(2):99102.
  48. Midence K,Fuggle P,Davies SC.Psychosocial aspects of sickle cell disease (SCD) in childhood and adolescence: a review.Br J Clin Psychol.1993;32 (pt 3):271280.
  49. Weaver MF,Bond DS,Arnold BL,Waterhouse E,Towne A.Aberrant drug‐taking behaviors and headache: patient versus physician report.Am J Health Behav.2006;30(5):475482.
  50. Ballas SK.Current issues in sickle cell pain and its management [review].Hematology Am Soc Hematol Educ Program.2007;97105.
  51. Meehan WJ,Adelman SA,Rehman Z,Khoromi S.Opioid Abuse. Available at: http://www.emedicine.com/med/topic1673.htm. Updated April 18, 2006. Accessed August 23,2006.
  52. Savage SR.Assessment for addiction in pain‐treatment settings.Clin J Pain.2002;18(4 suppl):S28S38.
  53. Bohnert AS,Valenstein M,Bair MJ,Ganoczy D,McCarthy JF,Ilgen MA,Blow FC.Association between opioid prescribing patterns and opioid overdose‐related deaths.JAMA.2011;305(13):13151321.
  54. Payne R.American Pain Society workshop on the management of sickle cell pain.Saint Louis, MO;1990.
  55. Vichinsky EP,Johnson R,Lubin BH.Multidisciplinary approach to pain management in sickle cell disease.Am J Pediatr Hematol Oncol.1982;4:328333.
  56. Brozovic M,Davies S,Yardumian A,Bellingham A,Marsh G,Stephens AD.Pain relief in sickle cell crisis [letter].Lancet.1986;2:624625.
  57. Elander J,Lusher J,Bevan D,Telfer P,Burton B.Understanding the causes of problematic pain management in sickle cell disease: evidence that pseudoaddiction plays a more important role than genuine analgesic dependence.J Pain Symptom Manage.2004;27(2):156169.
  58. Ballas SK,Smith ED.Red blood cell changes during the evolution of the sickle cell painful crisis.Blood.1992;79:21542163.
  59. Brandow AM,Brousseau DC,Panepinto JA.Postdischarge pain, functional limitations and impact on caregivers of children with sickle cell disease treated for painful events.Br J Haematol.2009;144(5):782788.
  60. Ballas SK,Lusardi M.Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance.Am J Hematol.2005;79:1725.
  61. Brousseau DC,Owens PL,Mosso AL,Panepinto JA,Steiner CA.Acute care utilization and rehospitalizations for sickle cell disease.JAMA.2010;303(13):12881294.
  62. Todd KH,Funk KG,Funk JP, et al.Clinical significance of reported changes in pain severity.Ann Emerg Med.1996;27:485489.
  63. Lopez B,Flenders P,Davis‐Moon L,Corbin T,Ballas S.Clinically significant differences in visual analogue scale in acute vasoocclusive sickle cell crisis.Hemoglobin.2007;31:427432.
  64. Hansson P,Backonja M,Bouhassira D.Usefulness and limitations of quantitative sensory testing: clinical and research application in neuropathic pain states.Pain.2007;129:256259.
References
  1. Serjeant GR.Sickle‐cell disease.Lancet.1997;350(9079):725302.
  2. Smith WR,Scherer M.Sickle‐cell pain: advances in epidemiology and etiology.Hematology Am Soc Hematol Educ Program.2010;409415. PMID: 21239827.
  3. Steinberg MH.Management of sickle cell disease.N Engl J Med.1999;340:10211030.
  4. Hebbel RP,Vercellotti G,Nath KA.A systems biology consideration of the vasculopathy of sickle cell anemia: the need for multi‐modality chemo‐prophylaxsis.Cardiovasc Hematol Disord Drug Targets.2009;9(4):271292.
  5. Conran N,Franco‐Penteado CF,Costa FF.Newer aspects of the pathophysiology of sickle cell disease vaso‐occlusion [review].Hemoglobin.2009;33(1):116.
  6. Davis H,Schoendorf KC,Gergen PJ,Moore RM.National trends in the mortality of children with sickle cell disease, 1968 through 1992.Am J Public Health.1997;87(8):13171322.
  7. Adams RJ,McKie VC,Hsu L, et al.Prevention of a first stroke by transfusions in children with sickle cell anemia and abnormal results on transcranial Doppler ultrasonography.N Engl J Med.1998;339:511.
  8. Adams RJ,Brambilla D.Optimizing primary stroke prevention in sickle cell anemia (STOP 2) trial investigators. Discontinuing prophylactic transfusions used to prevent stroke in sickle cell disease.N Engl J Med.2005;353(26):27692778.
  9. Sandy LG.Homeostasis without reserve—the risk of health system collapse.N Engl J Med.2002;347(24):19711975.
  10. The Advanced Medical Home: A Patient‐Centered, Physician‐Guided Model of Health Care [Policy Monograph].Philadelphia, PA:American College of Physicians;2006.
  11. Wagner EH,Glasgow RE,Davis C,Bonomi AE,Provost L,McCulloch D.Quality improvement in chronic illness care: a collaborative approach.Jt Comm J Qual Improv.2001;27:6380.
  12. Definition of Disease Prevalence for Therapies Qualifying Under the Orphan Drug Act. Subpart C, Designation of an Orphan Drug. Sec. 316.20. Content and format of a request for orphan‐drug designation. Available at: http://www.fda.gov/orphan/designat/prevalence.html. Accessed September 3,2008.
  13. Steinberg MH,Barton F,Castro O, et al.Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment.JAMA.2003;289:16451651.
  14. Lanzkron S,Haywood C,Hassell KL,Rand C.Provider barriers to hydroxyurea use in adults with sickle cell disease: a survey of the Sickle Cell Disease Adult Provider Network.J Natl Med Assoc.2008;100(8):968973.
  15. Treadwell M,Telfair J,Gibson RW,Johnson S,Osunkwo I.Transition from pediatric to adult care in sickle cell disease: establishing evidence‐based practice and directions for research.Am J Hematol.2011;86(1):116120. PMID: 21061308.
  16. Bobo L,Miller ST,Smith WR,Elam JT,Rosmarin PC,Lancaster DJ.Health perceptions and medical care opinions of inner‐city adults with sickle cell disease or asthma compared with those of their siblings.South Med J.1989;82(1):912.
  17. Shapiro BS,Benjamin LJ,Payne R,Heidrich G.Sickle cell‐related pain: perceptions of medical practitioners.J Pain Symptom Manage.1997;14(3):168174.
  18. The Management of Sickle Cell Disease.4th ed. NIH Publication 2002–2117.Washington, DC:National Institutes of Health, National Heart, Lung, and Blood Institute, Division of Blood Diseases and Resources; June2002.
  19. Pain Management Guideline.Washington, DC:Agency for Health Care Policy and Research;1992.
  20. Lottenberg R,Hassell KL.An evidence‐based approach to the treatment of adults with sickle cell disease.Hematology Am Soc Hematol Educ Program.2005;5865.
  21. World Health Organization: Cancer Pain Relief.Geneva, Switzerland:WHO,1986.
  22. Dunlop RJ,Bennett KC.Pain management for sickle cell disease.Cochrane Database Syst Rev. April 19,2006;(2):CD003350.
  23. Benjamin L,Dampier C,Jacox A;for the Guideline Committee.Guidelines for the Management of Acute and Chronic Pain in Sickle Cell Disease. APS Clinical Practice Guideline Series, No 1.Glenview, IL:American Pain Society;1999.
  24. Rees D.Guidelines for the management of the acute painful crisis of sickle cell disease.Br J Haematol.2003;120:744752.
  25. Field JJ,Knight‐Perry JE,DeBaun MR.Acute pain in children and adults with sickle cell disease: management in the absence of evidence‐based guidelines.Curr Opin Hematol.2009;16(3):173178.
  26. Rosenblum A,Marsch LA,Joseph H,Portenoy RK.Opioids and the treatment of chronic pain: controversies, current status, and future directions.Exp Clin Psychopharmacol.2008;16(5):405416.
  27. Axon RN.A sickle cell primer.The Hospitalist.2006;10(10):3941.
  28. Hill CS.The barriers to adequate pain management with opioid analgesics.Semin Oncol.1993;20(2 suppl 1):15.
  29. Dampier C,Setty BN,Eggleston B,Brodecki D,O'Neal P,Stuart M.Vaso‐occlusion in children with sickle cell disease: clinical characteristics and biologic correlates.J Pediatr Hematol Oncol.2004;26:785790. PMID: 15591896.
  30. Graido‐Gonzalez E,Doherty JC,Bergreen EW,Organ G,Telfer M,McMillen MA.Plasma endothelin‐1, cytokine, and prostaglandin E2 levels in sickle cell disease and acute vaso‐occlusive sickle crisis.Blood.1998;92:25512555. PMID: 9746797.
  31. Michaels LA,Ohene‐Frempong K,Zhao H,Douglas SD.Serum levels of substance P are elevated in patients with sickle cell disease and increase further during vaso‐occlusive crisis.Blood.1998;92:31483151. PMID: 9787150.
  32. Landburg PP,Teerlink T,Muskiet FA,Duits AJ,Schnog JJ;for the CURAMA study group.Plasma concentrations of asymmetric dimethylarginine, an endogenous nitric oxide synthase inhibitor, are elevated in sickle cell patients but do not increase further during painful crisis.Am J Hematol. February 27,2008; PMID: 18383318
  33. Platt OS,Thorington BD,Brambilla DJ, et al.Pain in sickle cell disease: rates and risk factors.N Engl J Med.1991;325:1116.
  34. Smith WR,Bovbjerg VE,Penberthy LT, et al.Understanding pain and improving management of sickle cell disease: the PiSCES Study.J Natl Med Assoc.2005;97(2):183193.
  35. Smith WR,McClish DK,Penberthy LT, et al.Daily assessment of pain in adults with sickle cell disease.Ann Intern Med.2008;148(2):94101.
  36. Aisiku IP,Smith WR,McClish DK, et al.Comparisons of high versus low emergency department utilizers in sickle cell disease.Ann Emerg Med.2009;53(5):587593.
  37. Helfand M,Freeman M.Assessment and management of acute pain in adult medical inpatients: a systematic review.Pain Med.2009;10(7):11831199. PMID: 19818030.
  38. Wallenstein SL.Scaling clinical pain and pain relief. In: Bromm B, ed.Pain Measurement in Man: Neorephysiological Correlates of Pain.New York:Elsevier Science Publishers,1984:389396.
  39. Dubois M.Patient‐controlled analgesia for acute pain.Clin J Pain.1989;5(suppl 1):S8S15. PMID: 2520435.
  40. Melzer‐Lange MD,Walsh‐Kelly CM,Lea G,Hillery CA,Scott JP.Patient‐controlled analgesia for sickle pain crisis in pediatric emergency department.Pediatr Emerg Care.2004;20:24.
  41. Trentadue N,Kachoyeanos MK,Lea G.A comparison of two regimens of patient‐controlled analgesia for children with sickle cell disease.J Pediatr Nurs.1998;13:1519.
  42. Narcotic analgesics, 2002 update.The DAWN Report;2004.
  43. Seifert CF,Kennedy S.Meperidine is alive and well in the new millennium: evaluation of meperidine usage patterns and frequency of adverse drug reactions.Pharmacotherapy.2004;24:776783.
  44. Porter BP,Coyne PJ,Smith WR.Methadone‐related torsades de pointes in a sickle cell patient treated for chronic pain.Am J Hematol.2005;78(4):316317.
  45. de Franceschi L,Finco G,Vassanelli A,Zaia B,Ischia S,Corrocher R.A pilot study on the efficacy of ketorolac plus tramadol infusion combined with erythrocytapheresis in the management of acute severe vaso‐occlusive crises and sickle cell pain.Haematologica.2004;89(11):13891391.
  46. Zempsky WT,Loiselle KA,Corsi JM,Hagstrom JN.Use of low‐dose ketamine infusion for pediatric patients with sickle cell disease‐related pain: a case series.Clin J Pain.2010;26(2):163167.
  47. Wang WC,George SL,Wilimas JA.Transcutaneous electrical nerve stimulation treatment of sickle cell pain crises.Acta Haematol.1988;80(2):99102.
  48. Midence K,Fuggle P,Davies SC.Psychosocial aspects of sickle cell disease (SCD) in childhood and adolescence: a review.Br J Clin Psychol.1993;32 (pt 3):271280.
  49. Weaver MF,Bond DS,Arnold BL,Waterhouse E,Towne A.Aberrant drug‐taking behaviors and headache: patient versus physician report.Am J Health Behav.2006;30(5):475482.
  50. Ballas SK.Current issues in sickle cell pain and its management [review].Hematology Am Soc Hematol Educ Program.2007;97105.
  51. Meehan WJ,Adelman SA,Rehman Z,Khoromi S.Opioid Abuse. Available at: http://www.emedicine.com/med/topic1673.htm. Updated April 18, 2006. Accessed August 23,2006.
  52. Savage SR.Assessment for addiction in pain‐treatment settings.Clin J Pain.2002;18(4 suppl):S28S38.
  53. Bohnert AS,Valenstein M,Bair MJ,Ganoczy D,McCarthy JF,Ilgen MA,Blow FC.Association between opioid prescribing patterns and opioid overdose‐related deaths.JAMA.2011;305(13):13151321.
  54. Payne R.American Pain Society workshop on the management of sickle cell pain.Saint Louis, MO;1990.
  55. Vichinsky EP,Johnson R,Lubin BH.Multidisciplinary approach to pain management in sickle cell disease.Am J Pediatr Hematol Oncol.1982;4:328333.
  56. Brozovic M,Davies S,Yardumian A,Bellingham A,Marsh G,Stephens AD.Pain relief in sickle cell crisis [letter].Lancet.1986;2:624625.
  57. Elander J,Lusher J,Bevan D,Telfer P,Burton B.Understanding the causes of problematic pain management in sickle cell disease: evidence that pseudoaddiction plays a more important role than genuine analgesic dependence.J Pain Symptom Manage.2004;27(2):156169.
  58. Ballas SK,Smith ED.Red blood cell changes during the evolution of the sickle cell painful crisis.Blood.1992;79:21542163.
  59. Brandow AM,Brousseau DC,Panepinto JA.Postdischarge pain, functional limitations and impact on caregivers of children with sickle cell disease treated for painful events.Br J Haematol.2009;144(5):782788.
  60. Ballas SK,Lusardi M.Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance.Am J Hematol.2005;79:1725.
  61. Brousseau DC,Owens PL,Mosso AL,Panepinto JA,Steiner CA.Acute care utilization and rehospitalizations for sickle cell disease.JAMA.2010;303(13):12881294.
  62. Todd KH,Funk KG,Funk JP, et al.Clinical significance of reported changes in pain severity.Ann Emerg Med.1996;27:485489.
  63. Lopez B,Flenders P,Davis‐Moon L,Corbin T,Ballas S.Clinically significant differences in visual analogue scale in acute vasoocclusive sickle cell crisis.Hemoglobin.2007;31:427432.
  64. Hansson P,Backonja M,Bouhassira D.Usefulness and limitations of quantitative sensory testing: clinical and research application in neuropathic pain states.Pain.2007;129:256259.
Issue
Journal of Hospital Medicine - 6(5)
Issue
Journal of Hospital Medicine - 6(5)
Page Number
297-303
Page Number
297-303
Article Type
Display Headline
Frequently asked questions by hospitalists managing pain in adults with sickle cell disease
Display Headline
Frequently asked questions by hospitalists managing pain in adults with sickle cell disease
Legacy Keywords
hematology, pain, multidisciplinary care, quality improvement, sickle cell disease
Legacy Keywords
hematology, pain, multidisciplinary care, quality improvement, sickle cell disease
Sections
Article Source
Copyright © 2011 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Division of General Internal Medicine, Adult Sickle Cell Program, Department of Internal Medicine, Virginia Commonwealth University, 1200 E. Broad St., Box 980306, Richmond, VA 23298‐0306
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media

SCD High Utilization

Article Type
Changed
Mon, 01/02/2017 - 19:34
Display Headline
Understanding and meeting the hospitalist's challenge: Caring for adults with sickle cell disease

In November 2010, the National Heart, Lung, and Blood Institute celebrated the 100th year anniversary of the discovery of sickle cell disease (SCD) in the United States by hosting the Herrick Symposium. Despite progress in the past 100 years, there is just one treatment available (hydroxyurea) and, while SCD is no longer considered only a disease of children, patients' life spans remain severely shortened (42 and 48 years, respectively, for males and females).1 With restrictions in residency hours and hospitals efforts to contain costs, hospitalists are increasingly being called upon to manage inpatient care for adults with SCD during their hospitalization. With the Centers for Medicare and Medicaid Services' recent plans to penalize hospitals with 30‐day readmission rates in excess of expected, hospitalists should address the challenges they face in providing care to adults with SCD and identify strategies to successfully meet them.

In this issue of the Journal of Hospital Medicine, Carroll and colleagues examined data from the California State Inpatient Database provided by the Healthcare Cost and Utilization Project (HCUP) for persons with International Classification of Diseases, Ninth Revision (ICD‐9) codes for SCD.2 They characterized patterns of hospital use during a 4‐year study period. Records for all patients, age 13 and older, with an admission for an SCD ICD‐9related cause were included. Patients with 4 or more hospitalizations in a 12‐month period were classified as having high hospital utilization, and 25% of the 1879 different patients evaluated fell into this category. A general perception exists that most persons with SCD have high hospital utilization, but data from Carroll et al. challenge this perception.2

While 1 in 4 patients in the cohort had high hospital utilization, why were not even more able to stay out of the hospital? Characteristics of these high utilizers shed light on contributing factors. Many patients died in the hospital (6.6%), consistent with research by others who also demonstrated the high risk of death associated with rehospitalization among patients with SCD.3, 4 In a prospective cohort study of 71 adults with either 70 hospital days or 6 admissions in a 12‐month period, 15% of the cohort died within a 24‐month study period.3 Patients with the highest number of hospital days, and those suffering from depression, were at highest risk of death. A separate prospective, longitudinal, 4‐year cohort study of adults with sickle cell anemia found an overall mortality of 14%.4 Mortality for those readmitted within 1 week of a painful crisis was 20%, compared to 11% for others in the cohort. High hospitalization use and hospital readmissions should be seen as worrisome markers of high risk for death, and patients should be carefully evaluated for life‐threatening complications, and not assumed to be purely drug seeking.

Why do some patients with SCD experience high readmission rates and mortality? Such patients with frequent hospitalizations have been found, in fact, to be sicker, and Carroll's research confirms that high utilizers were more likely to have comorbidities (acute chest syndrome, aseptic necrosis, renal disease) and complications (sepsis, pneumonia, pulmonary embolus, diabetes, mood disorders, and cocaine and alcohol use).2 Fortunately, high utilization appears to moderate over time for most patients, but those with persistent high utilization were more likely to have sepsis and mood disorders. Aisiku and colleagues studied a cohort of adults with SCD, in Virginia, in which emergency department (ED) utilization provided additional evidence for the association of high utilization with worse outcomes.5 Patients with 3 or more ED visits in 1 year were found to have lower hematocrits and higher white blood cell counts, to require more blood transfusions, and to report more pain, more pain days, more pain crises, and a worse quality of life. It is clear that patients with high hospital utilization are sicker and at an increased risk of death. While enlightening, this data does not tell the entire story.

Most admissions for patients with SCD begin with a vaso‐occlusive crisis, and frequently other complications may develop. Smith et al. provided the first prospectively collected data documenting that these patients reported pain on more than 54% of days, and many experience pain daily, yet infrequently access healthcare services.6 Hospitalists should appreciate that chronic pain is common for many adults with SCD. Pain management in this patient population is complex and often challenging, requiring high doses of opioids. In this current issue of the Journal, Smith and colleagues have contributed an excellent overview of how to manage pain in adults with SCD.7 The review specifically addresses some of the most challenging aspects of pain management in the hospital setting. Unfortunately, healthcare providers too frequently perceive patients as addicted to narcotics and abusing them, despite clear evidence that patients with SCD suffer from chronic pain. The consequent crisis of trust between the patient and provider commonly leads to inadequate treatment of pain and subsequent self‐discharge. Haywood and colleagues compared trust levels in adults with SCD and a history of sudden self‐discharge (ie, leaving against medical advice [AMA]) to those without such a history.8 Patients with a history of self‐discharge reported lower levels of trust of the medical staff and more negative interpersonal experiences. In a separate investigation, researchers compared scores on the Picker Patient Experience Questionnaire between a cohort of adults with SCD and national norms.9 Patients with SCD scored lower on 9 of 12 items. More specifically, 86% of respondents reported having insufficient involvement in decisions about care and treatment, and 50% reported staff did not do enough to control pain. Sadly, it appears the health system overall has undertreated and mismanaged patients with SCD suffering from pain crises.

Hospitalists face many challenges when managing care for adults with such a complex disease associated with high mortality, severe pain, and often a high readmission rate. The complexity of SCD calls for a comprehensive approach, and the need for each patient to have a clearly identified medical home.10 The hospital, emergency department, and hospitalist cannot and should not serve this role. Recently, Lindquist and Baker proposed a framework to understand and prevent hospital readmissions.11 They recommend optimizing the interfaces of transitional care among the patient, hospitalist, and primary care physician (PCP). By applying this framework of care, hospitalists must identify a PCP and provider with SCD expertise for follow‐up. Clear communication between the PCP, sickle cell expert, and hospitalist can be used to facilitate inpatient and emergency department care, and avoid inconsistent care that fosters mistrust. Individualized and consistent analgesic protocols established by outpatient providers, in collaboration with the patient, are more likely to deliver effective care, compared to variable attempts by whatever hospitalist happens to admit the patient.

Working with physicians expert in the care of patients with SCD, hospitalists might also identify patients who may benefit from hydroxyurea (HU) therapy. Despite clear evidence of HU's multiple salutary effects (decreased number of painful crises and need for hospital admissions, reduced number of blood transfusions and frequency of acute chest syndrome, and an overall benefit in mortality),12, 13 it remains underprescribed.14 In an analysis of a Medicaid managed care organization database in Maryland, 85% of patients never refilled a HU prescription. Moreover, patients with the highest rate of refills had the lowest number of hospital admissions and cost of care. Based on the evidence, hospitalists should screen all patients to determine whether or not HU has been prescribed, and if not, patients should be carefully assessed to determine if they are candidates for this effective therapy with communication to the patient's PCP and SCD expert.

Carroll's analysis confirms that patients with sickle cell disease frequently admitted to hospital (high utilizers) suffer a heavier burden of their illness and are at remarkably high risk of further morbidity and mortality.2 Though admissions are usually for acute pain crises, these high utilizers also suffer greater risk of hematologic, cardiovascular, infectious, orthopedic, and psychiatric complications. The common psychiatric issues, including both mood disorders and substance abuse, emphasize the need for a multidisciplinary team of care providers to provide a comprehensive bio‐psycho‐social assessment of all patients with SCD who experience high hospital utilization. These patients will also benefit from system improvements that integrate and coordinate care across inpatient, outpatient, emergency department, and patient homes. Hospitalists are well positioned to engage in this model of care, as well as develop and improve processes to ensure seamless transitions across the various settings of care delivery. It is also crucial that hospitalists are engaged in research needed to better identify and understand risk factors that lead to high utilization. Only through collaborative efforts can we hope to solve the conundrum of frequently hospitalized patients with sickle cell pain crises.

References
  1. Platt OS,Brambilla DJ,Rosse WF, et al.Mortality in sickle cell disease. Life expectancy and risk factors for early death.N Engl J Med.1994;330:16391644.
  2. Carroll C,Haywood C,Lanzkron S.Prediction of onset and course of high hospital utilization in sickle cell disease.J Hosp Med.2011;6:248255.
  3. Houston‐Yu P,Rana SR,Beyer B,Castro O.Frequent and prolonged hospitalizations: a risk factor for early mortality in sickle cell disease patients.Am J Hematol.2003;72:201203.
  4. Ballas SK,Lusardi M.Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance.Am J Hematol.2005;79:1725.
  5. Aisiku IP,Smith WR,McClish DK,Levenson JL,Penberthy LT,Roseff SD.Comparison of high versus low emergency department utilizers in sickle cell disease.Ann Emerg Med.2009;53:587593.
  6. Smith WR,Penberthy LT,Bovbjerg VE, et al.Daily assessment of pain in adults with sickle cell disease.Ann Intern Med.2008;148:94101.
  7. Smith WR,Jordan LB,Hassell KL.Frequently asked questions by hospitalists managing pain in adults with sickle cell disease.J Hosp Med.2011;6:297303.
  8. Haywood C,Lanzkron S,Ratanawongsa N,Bediako SM,Lattimer‐Nelson L,Beach MC.Hospital self‐discharge among adults with sickle‐cell disease (SCD): associations with trust and interpersonal experiences with care.J Hosp Med.2010;5:289294.
  9. Lattimer L,Haywood C,Lanzkron S,Ratanawongsa N,Bediako SM,Beach MC.Problematic hospital experiences among adult patients with sickle cell disease.J Health Care Poor Underserved.2010;21:11141123.
  10. Hassell K,Pace B,Wang W, et al.Sickle cell disease summit: from clinical and research disparity to action.Am J Hematol.2008;84:3945.
  11. Lindquist L,Baker DW.Understanding preventable hospital readmissions: masqueraders, markers and true causal factors.J Hosp Med.2011;6:5153.
  12. Charache S,Terrin M,Moore R, et al.Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia.N Engl J Med.1995;332:13171322.
  13. Steinberg MH,Barton F,Castro O, et al.Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment.JAMA.2003;289:16451651.
  14. Lanzkron S,Haywood C,Fagan PJ,Rand CS.Examining the effectiveness of hydroxyurea in people with sickle cell disease.J Health Care Poor Underserved.2010;21:277286.
Article PDF
Issue
Journal of Hospital Medicine - 6(5)
Page Number
245-247
Sections
Article PDF
Article PDF

In November 2010, the National Heart, Lung, and Blood Institute celebrated the 100th year anniversary of the discovery of sickle cell disease (SCD) in the United States by hosting the Herrick Symposium. Despite progress in the past 100 years, there is just one treatment available (hydroxyurea) and, while SCD is no longer considered only a disease of children, patients' life spans remain severely shortened (42 and 48 years, respectively, for males and females).1 With restrictions in residency hours and hospitals efforts to contain costs, hospitalists are increasingly being called upon to manage inpatient care for adults with SCD during their hospitalization. With the Centers for Medicare and Medicaid Services' recent plans to penalize hospitals with 30‐day readmission rates in excess of expected, hospitalists should address the challenges they face in providing care to adults with SCD and identify strategies to successfully meet them.

In this issue of the Journal of Hospital Medicine, Carroll and colleagues examined data from the California State Inpatient Database provided by the Healthcare Cost and Utilization Project (HCUP) for persons with International Classification of Diseases, Ninth Revision (ICD‐9) codes for SCD.2 They characterized patterns of hospital use during a 4‐year study period. Records for all patients, age 13 and older, with an admission for an SCD ICD‐9related cause were included. Patients with 4 or more hospitalizations in a 12‐month period were classified as having high hospital utilization, and 25% of the 1879 different patients evaluated fell into this category. A general perception exists that most persons with SCD have high hospital utilization, but data from Carroll et al. challenge this perception.2

While 1 in 4 patients in the cohort had high hospital utilization, why were not even more able to stay out of the hospital? Characteristics of these high utilizers shed light on contributing factors. Many patients died in the hospital (6.6%), consistent with research by others who also demonstrated the high risk of death associated with rehospitalization among patients with SCD.3, 4 In a prospective cohort study of 71 adults with either 70 hospital days or 6 admissions in a 12‐month period, 15% of the cohort died within a 24‐month study period.3 Patients with the highest number of hospital days, and those suffering from depression, were at highest risk of death. A separate prospective, longitudinal, 4‐year cohort study of adults with sickle cell anemia found an overall mortality of 14%.4 Mortality for those readmitted within 1 week of a painful crisis was 20%, compared to 11% for others in the cohort. High hospitalization use and hospital readmissions should be seen as worrisome markers of high risk for death, and patients should be carefully evaluated for life‐threatening complications, and not assumed to be purely drug seeking.

Why do some patients with SCD experience high readmission rates and mortality? Such patients with frequent hospitalizations have been found, in fact, to be sicker, and Carroll's research confirms that high utilizers were more likely to have comorbidities (acute chest syndrome, aseptic necrosis, renal disease) and complications (sepsis, pneumonia, pulmonary embolus, diabetes, mood disorders, and cocaine and alcohol use).2 Fortunately, high utilization appears to moderate over time for most patients, but those with persistent high utilization were more likely to have sepsis and mood disorders. Aisiku and colleagues studied a cohort of adults with SCD, in Virginia, in which emergency department (ED) utilization provided additional evidence for the association of high utilization with worse outcomes.5 Patients with 3 or more ED visits in 1 year were found to have lower hematocrits and higher white blood cell counts, to require more blood transfusions, and to report more pain, more pain days, more pain crises, and a worse quality of life. It is clear that patients with high hospital utilization are sicker and at an increased risk of death. While enlightening, this data does not tell the entire story.

Most admissions for patients with SCD begin with a vaso‐occlusive crisis, and frequently other complications may develop. Smith et al. provided the first prospectively collected data documenting that these patients reported pain on more than 54% of days, and many experience pain daily, yet infrequently access healthcare services.6 Hospitalists should appreciate that chronic pain is common for many adults with SCD. Pain management in this patient population is complex and often challenging, requiring high doses of opioids. In this current issue of the Journal, Smith and colleagues have contributed an excellent overview of how to manage pain in adults with SCD.7 The review specifically addresses some of the most challenging aspects of pain management in the hospital setting. Unfortunately, healthcare providers too frequently perceive patients as addicted to narcotics and abusing them, despite clear evidence that patients with SCD suffer from chronic pain. The consequent crisis of trust between the patient and provider commonly leads to inadequate treatment of pain and subsequent self‐discharge. Haywood and colleagues compared trust levels in adults with SCD and a history of sudden self‐discharge (ie, leaving against medical advice [AMA]) to those without such a history.8 Patients with a history of self‐discharge reported lower levels of trust of the medical staff and more negative interpersonal experiences. In a separate investigation, researchers compared scores on the Picker Patient Experience Questionnaire between a cohort of adults with SCD and national norms.9 Patients with SCD scored lower on 9 of 12 items. More specifically, 86% of respondents reported having insufficient involvement in decisions about care and treatment, and 50% reported staff did not do enough to control pain. Sadly, it appears the health system overall has undertreated and mismanaged patients with SCD suffering from pain crises.

Hospitalists face many challenges when managing care for adults with such a complex disease associated with high mortality, severe pain, and often a high readmission rate. The complexity of SCD calls for a comprehensive approach, and the need for each patient to have a clearly identified medical home.10 The hospital, emergency department, and hospitalist cannot and should not serve this role. Recently, Lindquist and Baker proposed a framework to understand and prevent hospital readmissions.11 They recommend optimizing the interfaces of transitional care among the patient, hospitalist, and primary care physician (PCP). By applying this framework of care, hospitalists must identify a PCP and provider with SCD expertise for follow‐up. Clear communication between the PCP, sickle cell expert, and hospitalist can be used to facilitate inpatient and emergency department care, and avoid inconsistent care that fosters mistrust. Individualized and consistent analgesic protocols established by outpatient providers, in collaboration with the patient, are more likely to deliver effective care, compared to variable attempts by whatever hospitalist happens to admit the patient.

Working with physicians expert in the care of patients with SCD, hospitalists might also identify patients who may benefit from hydroxyurea (HU) therapy. Despite clear evidence of HU's multiple salutary effects (decreased number of painful crises and need for hospital admissions, reduced number of blood transfusions and frequency of acute chest syndrome, and an overall benefit in mortality),12, 13 it remains underprescribed.14 In an analysis of a Medicaid managed care organization database in Maryland, 85% of patients never refilled a HU prescription. Moreover, patients with the highest rate of refills had the lowest number of hospital admissions and cost of care. Based on the evidence, hospitalists should screen all patients to determine whether or not HU has been prescribed, and if not, patients should be carefully assessed to determine if they are candidates for this effective therapy with communication to the patient's PCP and SCD expert.

Carroll's analysis confirms that patients with sickle cell disease frequently admitted to hospital (high utilizers) suffer a heavier burden of their illness and are at remarkably high risk of further morbidity and mortality.2 Though admissions are usually for acute pain crises, these high utilizers also suffer greater risk of hematologic, cardiovascular, infectious, orthopedic, and psychiatric complications. The common psychiatric issues, including both mood disorders and substance abuse, emphasize the need for a multidisciplinary team of care providers to provide a comprehensive bio‐psycho‐social assessment of all patients with SCD who experience high hospital utilization. These patients will also benefit from system improvements that integrate and coordinate care across inpatient, outpatient, emergency department, and patient homes. Hospitalists are well positioned to engage in this model of care, as well as develop and improve processes to ensure seamless transitions across the various settings of care delivery. It is also crucial that hospitalists are engaged in research needed to better identify and understand risk factors that lead to high utilization. Only through collaborative efforts can we hope to solve the conundrum of frequently hospitalized patients with sickle cell pain crises.

In November 2010, the National Heart, Lung, and Blood Institute celebrated the 100th year anniversary of the discovery of sickle cell disease (SCD) in the United States by hosting the Herrick Symposium. Despite progress in the past 100 years, there is just one treatment available (hydroxyurea) and, while SCD is no longer considered only a disease of children, patients' life spans remain severely shortened (42 and 48 years, respectively, for males and females).1 With restrictions in residency hours and hospitals efforts to contain costs, hospitalists are increasingly being called upon to manage inpatient care for adults with SCD during their hospitalization. With the Centers for Medicare and Medicaid Services' recent plans to penalize hospitals with 30‐day readmission rates in excess of expected, hospitalists should address the challenges they face in providing care to adults with SCD and identify strategies to successfully meet them.

In this issue of the Journal of Hospital Medicine, Carroll and colleagues examined data from the California State Inpatient Database provided by the Healthcare Cost and Utilization Project (HCUP) for persons with International Classification of Diseases, Ninth Revision (ICD‐9) codes for SCD.2 They characterized patterns of hospital use during a 4‐year study period. Records for all patients, age 13 and older, with an admission for an SCD ICD‐9related cause were included. Patients with 4 or more hospitalizations in a 12‐month period were classified as having high hospital utilization, and 25% of the 1879 different patients evaluated fell into this category. A general perception exists that most persons with SCD have high hospital utilization, but data from Carroll et al. challenge this perception.2

While 1 in 4 patients in the cohort had high hospital utilization, why were not even more able to stay out of the hospital? Characteristics of these high utilizers shed light on contributing factors. Many patients died in the hospital (6.6%), consistent with research by others who also demonstrated the high risk of death associated with rehospitalization among patients with SCD.3, 4 In a prospective cohort study of 71 adults with either 70 hospital days or 6 admissions in a 12‐month period, 15% of the cohort died within a 24‐month study period.3 Patients with the highest number of hospital days, and those suffering from depression, were at highest risk of death. A separate prospective, longitudinal, 4‐year cohort study of adults with sickle cell anemia found an overall mortality of 14%.4 Mortality for those readmitted within 1 week of a painful crisis was 20%, compared to 11% for others in the cohort. High hospitalization use and hospital readmissions should be seen as worrisome markers of high risk for death, and patients should be carefully evaluated for life‐threatening complications, and not assumed to be purely drug seeking.

Why do some patients with SCD experience high readmission rates and mortality? Such patients with frequent hospitalizations have been found, in fact, to be sicker, and Carroll's research confirms that high utilizers were more likely to have comorbidities (acute chest syndrome, aseptic necrosis, renal disease) and complications (sepsis, pneumonia, pulmonary embolus, diabetes, mood disorders, and cocaine and alcohol use).2 Fortunately, high utilization appears to moderate over time for most patients, but those with persistent high utilization were more likely to have sepsis and mood disorders. Aisiku and colleagues studied a cohort of adults with SCD, in Virginia, in which emergency department (ED) utilization provided additional evidence for the association of high utilization with worse outcomes.5 Patients with 3 or more ED visits in 1 year were found to have lower hematocrits and higher white blood cell counts, to require more blood transfusions, and to report more pain, more pain days, more pain crises, and a worse quality of life. It is clear that patients with high hospital utilization are sicker and at an increased risk of death. While enlightening, this data does not tell the entire story.

Most admissions for patients with SCD begin with a vaso‐occlusive crisis, and frequently other complications may develop. Smith et al. provided the first prospectively collected data documenting that these patients reported pain on more than 54% of days, and many experience pain daily, yet infrequently access healthcare services.6 Hospitalists should appreciate that chronic pain is common for many adults with SCD. Pain management in this patient population is complex and often challenging, requiring high doses of opioids. In this current issue of the Journal, Smith and colleagues have contributed an excellent overview of how to manage pain in adults with SCD.7 The review specifically addresses some of the most challenging aspects of pain management in the hospital setting. Unfortunately, healthcare providers too frequently perceive patients as addicted to narcotics and abusing them, despite clear evidence that patients with SCD suffer from chronic pain. The consequent crisis of trust between the patient and provider commonly leads to inadequate treatment of pain and subsequent self‐discharge. Haywood and colleagues compared trust levels in adults with SCD and a history of sudden self‐discharge (ie, leaving against medical advice [AMA]) to those without such a history.8 Patients with a history of self‐discharge reported lower levels of trust of the medical staff and more negative interpersonal experiences. In a separate investigation, researchers compared scores on the Picker Patient Experience Questionnaire between a cohort of adults with SCD and national norms.9 Patients with SCD scored lower on 9 of 12 items. More specifically, 86% of respondents reported having insufficient involvement in decisions about care and treatment, and 50% reported staff did not do enough to control pain. Sadly, it appears the health system overall has undertreated and mismanaged patients with SCD suffering from pain crises.

Hospitalists face many challenges when managing care for adults with such a complex disease associated with high mortality, severe pain, and often a high readmission rate. The complexity of SCD calls for a comprehensive approach, and the need for each patient to have a clearly identified medical home.10 The hospital, emergency department, and hospitalist cannot and should not serve this role. Recently, Lindquist and Baker proposed a framework to understand and prevent hospital readmissions.11 They recommend optimizing the interfaces of transitional care among the patient, hospitalist, and primary care physician (PCP). By applying this framework of care, hospitalists must identify a PCP and provider with SCD expertise for follow‐up. Clear communication between the PCP, sickle cell expert, and hospitalist can be used to facilitate inpatient and emergency department care, and avoid inconsistent care that fosters mistrust. Individualized and consistent analgesic protocols established by outpatient providers, in collaboration with the patient, are more likely to deliver effective care, compared to variable attempts by whatever hospitalist happens to admit the patient.

Working with physicians expert in the care of patients with SCD, hospitalists might also identify patients who may benefit from hydroxyurea (HU) therapy. Despite clear evidence of HU's multiple salutary effects (decreased number of painful crises and need for hospital admissions, reduced number of blood transfusions and frequency of acute chest syndrome, and an overall benefit in mortality),12, 13 it remains underprescribed.14 In an analysis of a Medicaid managed care organization database in Maryland, 85% of patients never refilled a HU prescription. Moreover, patients with the highest rate of refills had the lowest number of hospital admissions and cost of care. Based on the evidence, hospitalists should screen all patients to determine whether or not HU has been prescribed, and if not, patients should be carefully assessed to determine if they are candidates for this effective therapy with communication to the patient's PCP and SCD expert.

Carroll's analysis confirms that patients with sickle cell disease frequently admitted to hospital (high utilizers) suffer a heavier burden of their illness and are at remarkably high risk of further morbidity and mortality.2 Though admissions are usually for acute pain crises, these high utilizers also suffer greater risk of hematologic, cardiovascular, infectious, orthopedic, and psychiatric complications. The common psychiatric issues, including both mood disorders and substance abuse, emphasize the need for a multidisciplinary team of care providers to provide a comprehensive bio‐psycho‐social assessment of all patients with SCD who experience high hospital utilization. These patients will also benefit from system improvements that integrate and coordinate care across inpatient, outpatient, emergency department, and patient homes. Hospitalists are well positioned to engage in this model of care, as well as develop and improve processes to ensure seamless transitions across the various settings of care delivery. It is also crucial that hospitalists are engaged in research needed to better identify and understand risk factors that lead to high utilization. Only through collaborative efforts can we hope to solve the conundrum of frequently hospitalized patients with sickle cell pain crises.

References
  1. Platt OS,Brambilla DJ,Rosse WF, et al.Mortality in sickle cell disease. Life expectancy and risk factors for early death.N Engl J Med.1994;330:16391644.
  2. Carroll C,Haywood C,Lanzkron S.Prediction of onset and course of high hospital utilization in sickle cell disease.J Hosp Med.2011;6:248255.
  3. Houston‐Yu P,Rana SR,Beyer B,Castro O.Frequent and prolonged hospitalizations: a risk factor for early mortality in sickle cell disease patients.Am J Hematol.2003;72:201203.
  4. Ballas SK,Lusardi M.Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance.Am J Hematol.2005;79:1725.
  5. Aisiku IP,Smith WR,McClish DK,Levenson JL,Penberthy LT,Roseff SD.Comparison of high versus low emergency department utilizers in sickle cell disease.Ann Emerg Med.2009;53:587593.
  6. Smith WR,Penberthy LT,Bovbjerg VE, et al.Daily assessment of pain in adults with sickle cell disease.Ann Intern Med.2008;148:94101.
  7. Smith WR,Jordan LB,Hassell KL.Frequently asked questions by hospitalists managing pain in adults with sickle cell disease.J Hosp Med.2011;6:297303.
  8. Haywood C,Lanzkron S,Ratanawongsa N,Bediako SM,Lattimer‐Nelson L,Beach MC.Hospital self‐discharge among adults with sickle‐cell disease (SCD): associations with trust and interpersonal experiences with care.J Hosp Med.2010;5:289294.
  9. Lattimer L,Haywood C,Lanzkron S,Ratanawongsa N,Bediako SM,Beach MC.Problematic hospital experiences among adult patients with sickle cell disease.J Health Care Poor Underserved.2010;21:11141123.
  10. Hassell K,Pace B,Wang W, et al.Sickle cell disease summit: from clinical and research disparity to action.Am J Hematol.2008;84:3945.
  11. Lindquist L,Baker DW.Understanding preventable hospital readmissions: masqueraders, markers and true causal factors.J Hosp Med.2011;6:5153.
  12. Charache S,Terrin M,Moore R, et al.Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia.N Engl J Med.1995;332:13171322.
  13. Steinberg MH,Barton F,Castro O, et al.Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment.JAMA.2003;289:16451651.
  14. Lanzkron S,Haywood C,Fagan PJ,Rand CS.Examining the effectiveness of hydroxyurea in people with sickle cell disease.J Health Care Poor Underserved.2010;21:277286.
References
  1. Platt OS,Brambilla DJ,Rosse WF, et al.Mortality in sickle cell disease. Life expectancy and risk factors for early death.N Engl J Med.1994;330:16391644.
  2. Carroll C,Haywood C,Lanzkron S.Prediction of onset and course of high hospital utilization in sickle cell disease.J Hosp Med.2011;6:248255.
  3. Houston‐Yu P,Rana SR,Beyer B,Castro O.Frequent and prolonged hospitalizations: a risk factor for early mortality in sickle cell disease patients.Am J Hematol.2003;72:201203.
  4. Ballas SK,Lusardi M.Hospital readmission for adult acute sickle cell painful episodes: frequency, etiology, and prognostic significance.Am J Hematol.2005;79:1725.
  5. Aisiku IP,Smith WR,McClish DK,Levenson JL,Penberthy LT,Roseff SD.Comparison of high versus low emergency department utilizers in sickle cell disease.Ann Emerg Med.2009;53:587593.
  6. Smith WR,Penberthy LT,Bovbjerg VE, et al.Daily assessment of pain in adults with sickle cell disease.Ann Intern Med.2008;148:94101.
  7. Smith WR,Jordan LB,Hassell KL.Frequently asked questions by hospitalists managing pain in adults with sickle cell disease.J Hosp Med.2011;6:297303.
  8. Haywood C,Lanzkron S,Ratanawongsa N,Bediako SM,Lattimer‐Nelson L,Beach MC.Hospital self‐discharge among adults with sickle‐cell disease (SCD): associations with trust and interpersonal experiences with care.J Hosp Med.2010;5:289294.
  9. Lattimer L,Haywood C,Lanzkron S,Ratanawongsa N,Bediako SM,Beach MC.Problematic hospital experiences among adult patients with sickle cell disease.J Health Care Poor Underserved.2010;21:11141123.
  10. Hassell K,Pace B,Wang W, et al.Sickle cell disease summit: from clinical and research disparity to action.Am J Hematol.2008;84:3945.
  11. Lindquist L,Baker DW.Understanding preventable hospital readmissions: masqueraders, markers and true causal factors.J Hosp Med.2011;6:5153.
  12. Charache S,Terrin M,Moore R, et al.Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia.N Engl J Med.1995;332:13171322.
  13. Steinberg MH,Barton F,Castro O, et al.Effect of hydroxyurea on mortality and morbidity in adult sickle cell anemia: risks and benefits up to 9 years of treatment.JAMA.2003;289:16451651.
  14. Lanzkron S,Haywood C,Fagan PJ,Rand CS.Examining the effectiveness of hydroxyurea in people with sickle cell disease.J Health Care Poor Underserved.2010;21:277286.
Issue
Journal of Hospital Medicine - 6(5)
Issue
Journal of Hospital Medicine - 6(5)
Page Number
245-247
Page Number
245-247
Article Type
Display Headline
Understanding and meeting the hospitalist's challenge: Caring for adults with sickle cell disease
Display Headline
Understanding and meeting the hospitalist's challenge: Caring for adults with sickle cell disease
Sections
Article Source
Copyright © 2011 Society of Hospital Medicine
Disallow All Ads
Correspondence Location
Northwestern University, 750 N. Lake Shore Drive, 10th Floor, #139, Chicago, IL 60611
Content Gating
Gated (full article locked unless allowed per User)
Gating Strategy
First Peek Free
Article PDF Media