Systemic Therapy in Metastatic Melanoma

Article Type
Changed
Fri, 11/10/2017 - 11:43
New targeted treatments and therapies for metastatic melanoma are improving patient prognosis and survival.

Melanoma is the most aggressive form of skin cancer, contributing to about 76,000 new cases and more than 9,000 deaths in 2014.1 Depending on the stage of the disease, 5-year melanoma survival can range from 15% to 97%. Patients with local and distant metastases have a 5-year survival of about 60% and 15%, respectively.2

The incidence of melanoma is rising, partly because of the increasing number of skin biopsies being performed.3 If melanoma is diagnosed early, surgical excision is the treatment of choice. In patients with oligometastatic disease (cancer that has spread, but only to 1 or a small number of sites), complete surgical excision of the metastases may provide prolonged overall survival (OS) and delay the need to use systemic therapy.4

Recently, many new drug therapies have shown promising results in clinical trials, which may improve the prognosis of metastatic disease. This article reviews currently available systemic treatment options for the management of metastatic melanoma, the role of cytotoxic chemotherapy and interleukin-2 (IL-2), and the latest therapies available, including immune checkpoint inhibitors.

Cytotoxic Chemotherapy and Interleukin-2

Cytotoxic chemotherapy does not have an established role in the initial treatment of metastatic melanoma. Currently, cytotoxic chemotherapy is used in patients who have not responded to immunotherapy or molecular targeted therapy. The most commonly used drugs include dacarbazine and its prodrug, temozolomide. Several studies have failed to demonstrate a survival benefit using a single-agent chemotherapy with either dacarbazine or temozolomide.5,6

Other agents used in metastatic melanoma include nitrosoureas (fotemustine), platinum compounds (cisplatin, carboplatin), vinca alkaloids (vincristine),
and taxanes (paclitaxel). None of these agents provide a survival benefit, but an objective response may be seen in a minority of cases. Combination chemotherapy regimens have not shown an advantage over singleagent dacarbazine or temozolomide.7,8

High-dose IL-2 has been used in cases of metastatic melanoma with good performance status (PS) and organ function. Studies have shown a complete response rate of 3% to 7% and a prolonged disease-free survival in a minority of patients.9-11 The use of highdose IL-2, however, is limited by the high incidence of adverse effects (AEs), which include bacterial sepsis, pulmonary edema, arrhythmias, fever, and on some occasions, death due to complications.10 The use of  IL-2 requires admission of the patient to a specialized unit for AE monitoring and management. Because of its ability to “cure” a minority of patients, a role still exists for IL-2 therapy in the treatment of younger, healthy patients with no evidence of organ dysfunction at baseline.

Immune Checkpoint Inhibitors

Checkpoint inhibitors are a class of drugs that unmask the immune system to fight against cancer cells. This class of drugs has shown significant activity and survival advantage in recent phase 2 and 3 trials. The class includes the anticytotoxic T-lymphocyte antigen 4 (CTLA-4) antibody ipilimumab and monoclonal antibodies targeting the programmed death 1 protein (PD-1) or its ligand (PD-L1).

Anti-CTLA-4 Antibodies: Ipilimumab

Cytotoxic T-lymphocyte antigen 4 is the antigen responsible for inhibition of cytotoxic T-cell-mediated immunity against foreign antigens presented by the antigen presenting cells (APCs). The APCs cause activation of the T cells when peptide fragments of intracellular proteins are presented in combination with mixed histocompatibility complex molecules. This step requires interaction of a costimulatory molecule (B7) on the APCs with a cluster of differentiation 28 protein (CD28) receptor located on T cells. CTLA-4 competes with CD28 to bind with the B7 molecule, thereby inhibiting the activation of the cytotoxic T cells (Figure 1). This pathway is thought to help with development of tolerance to host tissue antigens. Ipilimumab is a human monoclonal antibody that inhibits this CTLA-4 molecule and facilitates T-cell mediated antitumor activity.12 By blocking the CTLA-4 molecule, ipilimumab also mediates its autoimmune AEs on the host tissues.

Hodi and colleagues conducted a phase 3 trial of ipilimumab, including 676 patients who progressed after prior treatment for stage III or IV melanoma, and found that median OS was significantly better in the ipilimumab groups: 10 months in the ipilimumab plus gp100 peptide vaccine group vs 6.4 months in the gp100 vaccine alone group; 10.1 months in the ipilimumab alone group vs 6.4 months in the gp100 vaccine alone group.13 In another phase 3 trial comparing ipilimumab plus dacarbazine to dacarbazine alone, the ipilimumab group had a significantly improved OS (11.2 months vs 9.1 months).1 Survival rates with ipilimumab were prolonged for up to 3 years compared with the dacarbazine plus placebo group. However, the combination was associated with increased incidence of hepatotoxicity, thereby limiting its use.

A long-term survival analysis of 10 prospective and 2 retrospective studies of ipilimumab showed a median OS of 11.4 months and a long-term survival that began at 3 years with a plateau at 10 years of 21%, which was independent of prior therapy or ipilimumab dose.14 The immune-related AEs of ipilimumab are secondary to its activity against the host antigens and include dermatitis, enterocolitis, hepatitis, and endocrinopathies.15

A recent phase 2 trial studied the combination of ipilimumab with granulocyte-macrophage colonystimulating factor in 245 patients with stage III and IV melanoma. Median OS after 13 months was significantly higher with the combination compared with ipilimumab alone. The 1-year survival rate was 69% with
the combination and 53% with ipilimumab alone. There was no difference in the overall response rate (ORR) or progression-free survival (PFS) between the 2 groups. However, the AEs were significantly reduced with the combination (45% vs 58%).16 The dose of ipilimumab used in the trial was higher than the approved dose, making it difficult to apply the results in practice without further studies on the combination.

Anti-PD-1 Antibodies

Programmed death 1 ligands (PD-L1 and PD-L2) are expressed by tumor or stromal cells to inhibit the T-cell mediated antitumor activity. These ligands bind to the PD-1 protein on the surface of activated T cells to mediate their immunosuppressive effects. Interruption of this interaction by either anti-PD-1 antibodies or anti-PD-L1 antibodies facilitates tumor cell killing by activated T cells.17

Pembrozilumab and nivolumab are the 2 anti-PD-1 monoclonal antibodies that have been approved for treatment of metastatic melanoma. In a phase 1 trial
of pembrolizumab, 411 patients with advanced melanoma (consisting of both ipilimumab-naïve [IPI-N] and ipilimumab-treated [IPI-T] patients), ORR was 40% in IPI-N and 28% in IPI-T patients with a 1-year OS of 71% in all patients. Median PFS was 24 weeks in IPI-N and 23 weeks in IPI-T pts.18 There was no difference in outcomes and safety profiles across the various dosing regimens.18,19 Of note, pembrolizumab had antitumor activity irrespective of the PS, lactate dehydrogenase levels, BRAF (B-Raf proto-oncogene, serine/threonine kinase) gene mutation, metastatic stage, and number and type of prior therapy. In a subgroup analysis, 173 patients who had progression after treatment with ipilimumab were randomly assigned to pembrolizumab 2 mg/kg every 3 weeks (q3w) or 10 mg/kg q3w dosing regimens. Both groups had no significant difference in the ORR (26% in both) and safety profiles.20

In the 2012 KEYNOTE-002 clinical trial, a randomized phase 2 trial involving 540 patients with ipilimumab-refractory advanced melanoma, patients were randomized 1:1:1 to pembrolizumab 2 mg/kg or 10 mg/kg q3w or investigator-choice chemotherapy (control arm consisting of carboplatin plus paclitaxel, carboplatin, paclitaxel, dacarbazine, or temozolomide). The 6-month PFS was significantly improved with pembrolizumab (34% and 38% for pembrolizumab 2 mg/kg and 10 mg/kg, respectively) compared with 16% with chemotherapy. The ORR was significantly better with pembrolizumab (21% at 2 mg/kg, 25% at 10 mg/kg) compared with the control arm (4%).21 These findings led to the approval of pembrolizumab by the FDA for treatment of patients with advanced melanoma who have progressed on ipilimumab. Pembrolizumab is generally well tolerated. The most common AEs include fatigue, pruritus, and rash.

Nivolumab was studied in a recent phase 1 trial in which 107 patients with previously treated advanced melanoma were treated with escalated doses every
2 weeks.22 The 2-year and 3-year OS rates were 48% and 41%, respectively. Objective responses were seen in 32% of the patients. The median response duration was 23 months.23

The first phase 3 trial was conducted in 418 patients with previously untreated metastatic melanoma BRAF mutation. Patients were randomized to receive either nivolumab or dacarbazine. The PFS and OS were significantly better with nivolumab compared with dacarbazine (PFS 5.1 months vs 2.2 months; OS 73% vs 42% at 1 year).24 The AE profile of nivolumab is similar to pembrolizumab and includes lung, skin, endocrine, renal, and gastrointestinal tract toxicities.

Preliminary results of another phase 3 trial were presented at the European Society of Medical Oncology 2014 meeting. Patients with previously treated metastatic melanoma (ipilimumab or BRAF inhibitor) were randomized in a 2:1 ratio to receive either nivolumab or investigators’ choice chemotherapy (dacarbazine or carboplatin plus paclitaxel). The ORR was significantly better with nivolumab (32% vs 11%), and 95% of patients were still responding after 6 months. The nivolumab group showed a complete remission in 3% of the patients with 34% of the responses lasting ≥ 6 months.25 This led to the recent approval of nivolumab for patients with metastatic melanoma with a BRAF mutation who have advanced on ipilimumab. In the phase 3 NCT01844505 trial patients are being randomized to receive ipilimumab, nivolumab, or both.

A newer PD-1 inhibitor, pidilizumab, was studied in a phase 2 trial that included 103 patients with metastatic melanoma, 51% of whom had received therapy with ipilimumab. The ORR in the study group was relatively lower (6%), but the OS at 1 year was 64.5%.26 Further studies are underway to evaluate the role of this drug in metastatic melanoma.

The response with both nivolumab and pembrolizumab is durable as well as sustained, even after discontinuation of therapy. None of the deaths in the aforementioned studies were atributed to drug-related toxicities. As evidenced by current data, these 2 drugs hold a great promise for the management of patients who progress after therapy with anti-CTLA-4 antibodies.

Anti-PD-L1 Antibodies

The anti-PD-L1 monoclonal antibodies work in a similar way to the PD-1 inhibitors and block the interaction between the PD-1 and its ligand, PD-L1. This causes sustained activation of cytotoxic T cells and facilitates their antitumor activity. Two of PD-L1 inhibitors have shown clinical activity against metastatic melanoma.

BMS-936559, the first PD-L1 antibody, is being studied in a phase 1 trial that includes 55 patients with advanced melanoma along with 152 patients with other solid malignancies. Three patients achieved a complete response, and 5 patients had an objective response lasting 1 year. The ORR for melanoma was 17%, with disease stabilization of ≥ 24 weeks in 27% of the patients.27 Common AEs included infusion reactions, diarrhea, fatigue, rash, hypothyroidism, and hepatitis.

The second PD-L1 antibody, MPDL3280A, was studied in a phase 1 trial of 45 patients with metastatic melanoma. An ORR of 29% was observed, along with a 24-week PFS of 43%.28 Commonly noted AEs included hyperglycemia and elevated liver aminotransferases.

A newer PD-L1 inhibitor, MEDI4736, is being studied for advanced malignancies in 8 patients with melanoma. In preliminary analysis, MEDI4736 demonstrated a partial response in 1 out of 8 melanoma patients with a disease control rate of 46%.29 Although the PD-L1 inhibitors seem promising, more information will help discern their role in the management of metastatic melanoma.

Combined Anti-CTLA-4 Plus Anti-PD-1 Antibody

The combination of ipilimumab and the PD-1 inhibitor nivolumab was tested in a phase 1 trial in which both drugs were used concurrently as well as sequentially in metastatic melanoma.30 The 1- and 2-year OS in patients who were treated concurrently was 82% and 75%, respectively. Complete remission was seen in 17% of the patients, and the responses were seen irrespective of the BRAF mutation status. The responses were durable, and about 64% of the objective responses remain in remission at last follow-up.31 Grade 3 to grade 4 AEs were noted in 53% of the patients, with 11 patients requiring discontinuation of the medications. More studies are required to ascertain the optimum dosage of the combination prior to its approval for use in metastatic melanoma.

Molecular Targeted Therapy

The RAS-RAF–mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway is activated in almost 90% of patients
with melanoma.32 This pathway is normally required for the growth and survival of nonmalignant cells. In malignant transformation, mutations and/or overexpression is seen at various levels including KIT, NRAS, BRAF, and the MEK protein. This leads to activation of serine and threonine protein kinases, which lead to uncontrolled cell proliferation and survival.33

Novel therapeutic approaches have tried inhibiting one or more of these pathways for melanoma treatment. The most important mediator of tumorigenesis is BRAF, which is a downstream receptor of NRAS, and is mutated in almost 50% of melanoma cases.34 NRAS mutations are seen in 15% to 20% of cutaneous melanomas.35,36 After its activation, the RAF enzyme—coded by the BRAF gene—causes phosphorylation of the MEK protein, which activates ERK. This ERK activation leads to growth signaling and is the final pathway in several malignancies (Figure 2).37,38

BRAF Inhibitors

BRAF is the first mediator whose inhibition led to clinically significant outcomes in patients with melanoma. The most common BRAF mutation consists of the
substitution of glutamic acid for valine at amino acid 600 (V600E mutation) with majority of the remainder consisting of an alternate substitution (V600V or V600K).34 Vemurafenib and dabrafenib are the 2 BRAF inhibitors that have been shown to improve tumor regression, PFS, and OS considerably, especially in combination with a MEK protein inhibitor. In the phase 3 BRIM-3 trial, the vemurafenib group had a significantly prolonged PFS and OS compared with dacarbazine (13.6 months vs 9.7 months; 6.9 months vs 1.6 months, respectively). It was the first study to show improved survival with vemurafenib in both the V600E and V600K BRAF mutant melanomas.39

Another BRAF inhibitor, dabrafenib, was approved by the FDA for treatment of advanced melanoma with BRAF V600E mutation. It was tested in a phase 3 trial in which it was compared with dacarbazine in patients with advanced melanoma. Median OS in the dabrafenib arm was > 18 months and in dacarbazine arm > 15 months.40 Fifty-seven percent of the patients in dacarbazine arm were crossed over to the dabrafenib arm, thereby confounding the survival data for the former group. Another multicenter, phase 2 trial showed dabrafenib to have activity in melanoma patients with brain metastases, irrespective of previous therapy for the brain metastases.41 The long-term analysis of the BREAK-2 trial, which included 92 patients with metastatic melanoma treated with dabrafenib, showed a median OS of 12.9 months in BRAF V600K group and 13.1 months in BRAF V600E group.42

Adverse effects associated with BRAF inhibition include fatigue, rash, arthralgia, and photosensitivity reactions.43 Dermatologic complications may also include squamous cell carcinoma (SCC) (19%-26%), with keratoacanthoma being the most common subtype.44 These are believed to be likely secondary to the paradoxical activation of the MAPK signaling, since most of these lesions are found to have mutations in the RAS molecule.45 Other specific AEs of dabrafenib include hyperkeratosis (33%) and pyrexia (29%).42

Most patients treated with a BRAF inhibitor eventually have disease progression, likely secondary to reactivation of the MAPK pathway.46,47 This result has led to a heightened interest in combination therapies in an effort to improve outcomes. Combination therapy with ipilimumab and vemurafenib was studied and resulted in a higher incidence of hepatotoxicity (50%).48 However, no hepatotoxicity was seen in a phase 1 trial of combined dabrafenib and ipilimumab.49

Some studies have also suggested that extended BRAF inhibition after progression on a BRAF inhibitor may prolong survival.50,51 The phase 2 trial NCT01983124 is being conducted to evaluate the survival benefit with a combination of vemurafenib and a nitrosourea alkylating agent, fotemustine, in patients who have progressed on vemurafenib alone.

MEK Inhibitors

The inhibition of MEK can halt cell proliferation and induce apoptosis. The phase 3 METRIC trial, which compared the oral MEK inhibitor (trametinib) with chemotherapy, was conducted in 322 patients who had metastatic melanoma with a V600E or V600K BRAF mutation. The PFS and 6-month OS were significantly better with trametinib (4.8 months vs 1.5 months, 81% vs 66%) despite the crossover between the 2 groups.52 The AEs associated with trametinib included rash, diarrhea, and peripheral edema. Another phase 2 trial of trametinib including patients pretreated with a BRAF inhibitor showed no confirmed objective responses, 28% patients with stable disease, and minimal improvement in PFS (2 months). Among patients treated with prior chemotherapy and/or immunotherapy, trametinib showed significant improvement in complete responses, partial responses, stable disease, and the median PFS (2%, 23%, 51%, 4 months, respectively).53

The second MEK inhibitor, binimetinib, was studied in a phase 2 trial of advanced melanoma cases harboring a BRAF V600E or NRAS. Bimetinib demonstrated a PR in 20% cases of both the BRAF and NRAS mutant melanomas. Durable disease control was seen in 43% of the NRAS group and 32% of the BRAF group.54 The AE profile was similar to that seen with trametinib. Bimetinib is being studied in phase 1 and 2 trials with the CDK4/6 inhibitor as well as in the phase 3 trial NCT01763164 compared with dacarbazine in NRAS mutation positive melanomas.55

Selumetinib is a MEK inhibitor that has been compared with dacarbazine and temozolomide with no significant OS advantage. A novel highly specific inhibitor of MEK, cobimetinib, is currently being studied in combination with BRAF inhibitors.

Combined BRAF and MEK Inhibition

A randomized, double-blind, phase 3 study comparing the combination of dabrafenib and trametinib with dabrafenib and placebo in patients with advanced melanoma with a BRAF V600E mutation was presented at the 2014 American Society of Clinical Oncology meeting. Researchers found that after a median follow-up period of 9 months, there was a significant improvement with the combination in the PFS (9.3 months vs 8.8 months) and the ORR (67% vs 51%), with a similar incidence of AEs.56 The combination therapy group had fewer incidences of SCC of the skin but more incidence of pyrexia.

The combination of dabrafenib and trametinib was compared with vemurafenib monotherapy in a recent randomized phase 3 trial among 704 metastatic melanoma patients with a BRAF V600 mutation. Median PFS and ORR were significantly better with combination therapy compared with vemurafenib alone (11.4 months vs 7.3 months, 64% vs 51%, respectively). Overall survival rate at 1 year was significantly improved in the combination group as well (72% vs 65%).57 The incidence of SCC and keratoacanthoma was less in the combination (1%) compared with vemurafenib alone (18%). Another study investigating the coadministration and sequential administration of vemurafenib and trametinib is underway.58

The vemurafenib and cobimetinib combination was studied in a phase 3 trial of previously untreated unresectable locally advanced or metastatic BRAF V600
mutation-positive melanoma. The median PFS was 9.9 months in the combination group and 6.2 months in the control group. The interim analysis showed a 9-month survival rate of 81% in the combination group and 73% in the control group, with no significantly higher incidence of AEs in either arm.59 A longer follow-up will be needed to assess the OS benefit with the combination.

Encorafenib, a selective BRAF inhibitor, has been studied in a phase 1 trial in combination with binimetinib.60 This trial has paved the way to the initiation of a currently ongoing phase 3 trial (NCT01909453) comparing the combination with vemurafenib or encorafenib alone.

C-KIT Inhibitors

Mutations of c-KIT are seen more commonly in chronic sun damage-induced cutaneous melanomas, along with acral and mucosal melanomas.61,62 Earlier trials involving patients without selection for c-KIT mutation positivity failed to show benefit with imatinib. A single-arm, phase 2 trial of imatinib mesylate in patients with metastatic melanoma harboring the c-KIT mutation, an ORR of 23% was achieved, with a median PFS of 3.5 months.63 Imatinib showed an ORR of 29% in a phase 2 trial of mucosal, acral, and in chronic sun damage-induced melanoma patients with c-KIT amplifications and/or mutations. It was demonstrated that c-KIT amplification alone is not as responsive to imatinib compared with c-KIT mutation, suggesting that all patients with these specific melanomas should be tested for KIT mutation status.64

A second-generation c-KIT inhibitor, nilotinib, has shown some promising results with a favorable AE profile in small phase 2 trials.65,66 However, more clinical research will be needed before definite recommendations on its use in cutaneous melanomas can be made. Currently, its role seems to be limited to the management of acral, mucosal, and chronic sun damage-related melanomas with c-KIT mutations.

Future Directions

Angiogenesis promoters, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor, fibroblast growth factor, and interleukin-8, are overexpressed in melanoma. Bevacizumab, an anti-VEGF antibody, has been shown to have some benefit in combination with carboplatin and paclitaxel as a triple therapy.67 However, grade 3 AEs were seen in a portion of patients.

The phosphatidylinositol-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway has also been studied as a target for melanoma therapy. Everolimus, an mTOR inhibitor, was studied in a phase 2 trial in combination with bevacizumab for treatment of metastatic melanoma. The combination showed improved median PFS and OS with the combination (4 months and 8.6 months, respectively), with 43% of patients alive after 12 months of follow-up.68 This study points to the direction of possible benefits with the combination of anti-VEGF and immunotherapy. A recent study failed to show survival advantage with combination of bevacizumab and temozolomide.69

Buparlisib (BKM120), a PI3K inhibitor, has been shown to have activity in vivo and in vitro against melanoma brain metastases.70 More studies need to be done to assess the possible combination with other established therapies.

Oblimersen is an antisense oligonucleotide that suppresses B-cell lymphoma-2, thereby suppressing its anti-apoptotic effect. The triple combination of oblimersen with temozolomide and albumin-bound paclitaxel has shown to be safe and efficacious in a phase 1 trial, thereby creating a need for further clinical trials.71

Treatment Approach

Systemic therapy for metastatic melanoma depends on several factors, including BRAF mutation status, functional status of the patient, disease burden, and severity of symptoms. Assessing the BRAF mutation status has become an important component in the management of patients with metastatic melanoma. It can help recognize patients who will benefit from molecular targeted therapy. In case of a BRAF-positive melanoma, treatment can be initiated with either immunotherapy or BRAF inhibitors. There are no randomized studies comparing immunotherapy to molecular targeted therapy.

Patients who have good PS and lymph node metastases can be treated initially with IL-2, which has the advantage of inducing cure in a minority of patients but should only be considered in patients with well-preserved organ function who can be monitored in an intensive care setting. On the other hand, patients who have bulky, symptomatic disease and poor PS should be treated initially with BRAF inhibitors. Combination of BRAF and MEK inhibitors can also be used and has an improved PFS and OS with potential to cause early tumor regression. There are studies to suggest suboptimal outcomes in patients who are treated with ipilimumab after progression on a BRAF inhibitor compared with initial treatment with ipilimumab followed by a BRAF inhibitor.72-74 However, all these studies are retrospective and there is no prospective data to suggest the above. BRAF mutation-positive patients who progress on a BRAF inhibitor
can be treated with PD-1 inhibitors.

Patients who do not have a BRAF mutation are unlikely to benefit from a BRAF inhibitor and primarily receive immunotherapy with ipilimumab or IL-2. Whenever possible, such patients should be enrolled in a clinical trial, as they have a poor prognosis. Patients who progress on ipilimumab can be treated with one of the PD-1 inhibitors (pembrolizumab, nivolumab). These PD-L1 inhibitors are still being investigated for use in such situations.

The role of chemotherapy in the management of metastatic melanoma has been limited by numerous studies showing significantly better survival with immunotherapy and molecular targeted therapy. Dacarbazine is the only FDA-approved drug for the treatment of melanoma. Its use is reserved mainly for patients who are not candidates for any of the other therapies available, including enrollment in a clinical trial.

Conclusion

Therapies for metastatic melanoma are in a state of flux. In the past decade, several new therapeutic agents have been introduced for the management of this potentially lethal disease. The treatment of metastatic melanoma has gradually shifted from cytotoxic chemotherapy toward a more individualized treatment that has a definite survival advantage over traditional counterparts. The advent of novel therapies has led to initiation of further studies to determine their role in the treatment of advanced melanoma, singly or in combination with other agents. In addition to evaluating new agents, more studies are needed to compare existing treatment modalities so that definitive treatment protocols can be formulated.

Acknowledgement
The authors would like to thank Felicia Ratnaraj, MD, for her assistance in creating the figures.

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

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

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Click here to read the digital edition.

References

1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin.2014;64(1):9-29.

2. Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27(36):6199-6206.

3. Welch HG, Woloshin S, Schwartz LM. Skin biopsy rates and incidence of melanoma:
population based ecological study. BMJ. 2005;331(7515):481.

4. Sosman JA, Moon J, Tuthill RJ, et al. A phase 2 trial of complete resection for stage IV melanoma: results of Southwest Oncology Group Clinical Trial S9430. Cancer. 2011;117(20):4740-4746.

5. Atkins MB. The role of cytotoxic chemotherapeutic agents either alone or in combination with biological response modifiers. In: Kirkwood JK, ed. Molecular Diagnosis, Prevention, & Therapy of Melanoma. New York, NY: Marcel Dekker;1997:219-225.

6. Patel PM, Suciu S, Mortier L, et al. Extended schedule, escalated dose temozolomide versus dacarbazine in stage IV melanoma: final results of a randomised phase III study (EORTC 18032). Eur J Cancer. 2011;47(10):1476-1483.

7. Chapman PB, Einhorn LH, Meyers ML, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 1999;17(9):2745-2751.

8. Flaherty KT, Lee SJ, Zhao F, et al. Phase III trial of carboplatin and paclitaxel with
or without sorafenib in metastatic melanoma. J Clin Oncol. 2013;31(3):373-379.

9. Rosenberg SA, Yang JC, Topalian SL, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. 1994;271(12):907-913.

10. Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol. 1999;17(7):2105-2116.

11. Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am. 2000;6(suppl 1):S11-S14.

12. Hoos A, Ibrahim R, Korman A, et al. Development of ipilimumab: contribution to a new paradigm for cancer immunotherapy. Semin Oncol. 2010;37(5):533-546.

13. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723.

14. Schadendorf D, Hodi FS, Robert C, et. al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma [published online ahead of print February 9, 2015]. J Clin Oncol. pii:JCO.2014.56.2736.

15. Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691-2697.

16. Hodi FS, Lee S, McDermott DF, et al. Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: a randomized clinical trial. JAMA. 2014;312(17):1744-1753.

17. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443-2454.

18. Ribas A, Hodi FS, Kefford R, et al. Efficacy and safety of the anti-PD-1 monoclonal antibody pembrolizumab (MK-3475) in 411 patients (pts) with melanoma (MEL) (Abstract LBA9000). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

19. Hamid O, Robert C, Ribas A, et al. Randomized comparison of two doses of the anti-PD-1 monoclonal antibody MK-3475 for ipilimumab-refractory (IPI-R) and IPI-naive (IPI-N) melanoma (MEL) (abstract 3000). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

20. Robert C, Ribas A, Wolchok JD, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014; 384(9948):1109-1117.

21. Dummer R, Daud A, Puzanov I, et. al. A randomized controlled comparison of pembrolizumab and chemotherapy in patients with ipilimumab-refractory melanoma. J Transl Med. 2015;13(suppl 1):O5.

22. Topalian SL, Sznol M, McDermott DF, et. al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020-1030.

23. Hodi FS, Sznol M, Kluger HM, et al. Long-term survival of ipilimumab-naive patients with advanced melanoma (MEL) treated with nivolumab (anti-PD-1, BMS-936558, ONO-4538) in a phase I trial (abstract 9002). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

24. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320-330.

25. Weber J, D’Angelo S, Gutzmer R, et al. A phase 3 randomized, open-label study of nivolumab versus investigator’s choice of chemotherapy in patients with advanced melanoma after prior anti-CTLA4 therapy (abstract LBA3). Paper presented at: European Society of Medical Oncology 2014 meeting; September 2014; Madrid, Spain.

26. Atkins MB, Kudchadkar RR, Sznol M, et al. Phase 2, multicenter, safety and efficacy study of pidilizumab in patients with metastatic melanoma (abstract 9001). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

27. Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455-2465.

28. Hamid O, Sosman JA, Lawrence DP, et. al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). J Clin Oncol. 2013;31(15)(suppl): Abstract 9010.

29. Lutzky J, Antonia SJ, Blake-Haskins A, et. al. A phase 1 study of MEDI4736, an anti–PD-L1 antibody, in patients with advanced solid tumors. J Clin Oncol. 2014;32(15)(suppl): Abstract 3001.

30. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced
melanoma. N Engl J Med. 2013;369(2):122-133.

31. Sznol M, Kluger HM, Callahan MK, et al. Survival, response duration, and activity by BRAF mutation (MT) status of nivolumab (NIVO, anti-PD-1, BMS-936558, ONO-4538) and ipilimumab (IPI) concurrent therapy in advanced melanoma (MEL) (abstract LBA9003). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

32. Omholt K, Platz A, Kanter L, Ringborg U, Hansson J. NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progression. Clin Cancer Res. 2003;9(17):6483-6488.

33. Wellbrock C, Hurlstone A. BRAF as therapeutic target in melanoma. Biochem Pharmacol. 2010;80(5):561-567.

34. Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol. 2011;29(10):1239-1246.

35. Ball NJ, Yohn JJ, Morelli JG, et al. Ras mutations in human melanoma: a marker of malignant progression. J Invest Dermatol. 1994;102(3):285-290.

36. Platz A, Ringborg U, Brahme EM, Lagerlöf B. Melanoma metastases from patients with hereditary cutaneous malignant melanoma contain a high frequency of N-ras activating mutations. Melanoma Res. 1994;4(3):169-177.

37. Beeram M, Patnaik A, Rowinsky EK. Raf: a strategic target for therapeutic development against cancer. J Clin Oncol. 2005;23(27):6771-6790.

38. Terai K, Matsuda M. The amino-terminal B-Raf-specific region mediates calcium-dependent homo- and hetero-dimerization of Raf. EMBO J. 2006;25(15):3556-3564.

39. McArthur GA, Chapman PB, Robert C, et al. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014;15(3):323-332.

40. Hauschild A, Grob JJ, Demidov LV, et al. An update on BREAK-3, a phase III, randomized trial: dabrafenib versus dacarbazine in patients with BRAF V600E-positive mutation metastatic melanoma (Abstract 9013). Paper presented at: American Society of Clinical Oncology 2013 meeting; May-June 2013; Chicago, IL.

41. Long GV, Trefzer U, Davies MA, et al. Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(11):1087-1095.

42. Ascierto PA, Minor DR, Ribas A, et. al., Long-term safety and overall survival update for BREAK-2, a phase 2, single-arm, open-label study of dabrafenib in previously treated metastatic melanoma (NCT01153763). J Clin Oncol. 2014;32(15)(suppl): Abstract 9034.

43. Larkin J, Del Vecchio M, Ascierto PA, et al. Vemurafenib in patients with
BRAF(V600) mutated metastatic melanoma: an open-label, multicentre, safety
study. Lancet Oncol. 2014;15(4):436-444.

44. Lacouture ME, Duvic M, Hauschild A, et al. Analysis of dermatologic events in vemurafenib-treated patients with melanoma. Oncologist. 2013;18(3):314-322.

45. Su F, Viros A, Milagre C, et al. RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors. N Engl J Med. 2012;366(3):207-215.

46. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364(26):2507-2516.

47. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380(9839):358-365.

48. Ribas A, Hodi FS, Callahan M, et. al. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med. 2014;368(14):1365-1366.

49. Linette GP, Puzanov I, Callahan MK, et al. Phase 1 study of the BRAF inhibitor dabrafenib (D) with or without the MEK inhibitor trametinib (T) in combination with ipilimumab (Ipi) for V600E/K mutation–positive unresectable or metastatic melanoma (MM). J Clin Oncol. 2014;32(15)(suppl): Abstract 2511.

50. Chan MMK, Haydu LE, Menzies AM, et al. The nature and management of metastatic melanoma after progression on BRAF inhibitors: effects of extended BRAF inhibition. Cancer. 2014;120(20):3142-3153.

51. Carlino MS, Gowrishankar K, Saunders CAB, et al. Antiproliferative effects of continued mitogen-activated protein kinase pathway inhibition following acquired resistance to BRAF and/or MEK inhibition in melanoma. Mol Cancer Ther. 2013;12(7):1332-1342.

52. Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367(2):107-114.

53. Kim KB, Kefford R, Pavlick AC, et. al. Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol. 2013;31(1):482-489.

54. Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol. 2013;14(3):249-256.

55. Sosman JA, Kittaneh M, Lolkema MP, et al. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: early encouraging clinical activity (abstract 9009). Paper presented at: 2014 American Society of Clinical Oncology meeting ; May-June 2014; Chicago, IL.

56. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. 2014;371(20):1877-1888.

57. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30-39.

58. Gogas H, Schadendorf D, Dummer R. Vemurafenib treatment in patients with BRAF-mutated melanoma failing MEK inhibition with trametinib. J Clin Oncol. 2014;32(15)(suppl): Abstract 9061.

59. Larkin J, Ascierto PA, Dréno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371(20):1867-1876.

60. Kefford R, Miller WH, Tan DS, et al. Preliminary results from a phase Ib/II, openlabel, dose-escalation study of the oral BRAF inhibitor LGX818 in combination with the oral MEK1/2 inhibitor MEK162 in BRAF V600-dependent advanced solid tumors (abstract 9019). Paper presented at: 2013 American Society of Clinical Oncology meeting; May-June 2014; Chicago, IL.

61. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct
subtypes of melanoma. J Clin Oncol. 2006;24(26):4340-4346.

62. Jin SA, Chun SM, Choi YD, et al. BRAF mutations and KIT aberrations and their clinicopathological correlation in 202 Korean melanomas. J Invest Dermatol. 2013;133(2):579-582.

63. Guo J, Si L, Kong Y et. al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol. 2011;29(21):2904-2909.

64. Hodi FS, Corless CL, Giobbie-Hurder A, et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol. 2013;31(26):3182-3190.

65. Cho JH, Kim KM, Kwon M, Kim JH, Lee J. Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Invest New Drugs. 2012;30(5): 2008-2014.

66. Lebbe C, Chevret S, Jouary T, et. al. Phase II multicentric uncontrolled national trial assessing the efficacy of nilotinib in the treatment of advanced melanomas with c-KIT mutation or amplification. J Clin Oncol. 2014;32(15)(suppl): Abstract 9032.

67. Perez DG, Suman VJ, Fitch TR, et al. Phase 2 trial of carboplatin, weekly paclitaxel, and biweekly bevacizumab in patients with unresectable stage IV melanoma: a North Central Cancer Treatment Group study, N047A. Cancer. 2009;115(1):119-127.

68. Hainsworth JD, Infante JR, Spigel DR, et al. Bevacizumab and everolimus in the treatment of patients with metastatic melanoma. Cancer. 2010;116(17): 4122-4129.

69. Dronca RS, Allred JB, Perez DG, et. al. Phase II study of temozolomide (TMZ) and everolimus (RAD001) therapy for metastatic melanoma: a North Central Cancer Treatment Group study, N0675. Am J Clin Oncol. 2014;37(4):369-376.

70. Meier FE, Niessner H, Schmitz J, et al. The PI3K inhibitor BKM120 has potent antitumor activity in melanoma brain metastases in vitro and in vivo. J Clin Oncol. 2013;31(15)(suppl): Abstract e20050.

71. Ott PA, Chang J, Madden K, et al. Oblimersen in combination with temozolomide and albumin-bound paclitaxel in patients with advanced melanoma: a phase I trial. Cancer Chemother Pharmacol. 2013;71(1);183-191.

72. Ackerman A, Klein O, McDermott DF, et al. Outcomes of patients with metastatic
melanoma treated with immunotherapy prior to or after BRAF inhibitors. Cancer. 2014;120(11):1695-1701.

73. Ascierto PA, Margolin K. Ipilimumab before BRAF inhibitor treatment may be
more beneficial than vice versa for the majority of patients with advanced melanoma.
Cancer. 2014;120(11):1617-1619.

74. Ascierto PA, Simeone E, Sileni VC, et al. Sequential treatment with ipilimumab and BRAF inhibitors in patients with metastatic melanoma: data from the Italian cohort of the ipilimumab expanded access program. Cancer Invest. 2014;32(4):144-149.

Author and Disclosure Information

Dr. Goyal is a house officer in the Department of Internal Medicine and Dr. Silberstein is a professor and chief of Hematology/Oncology, both at CHI Health Creighton University Medical Center in Omaha, Nebraska. Dr. Silberstein is also the chief of oncology at VA Nebraska-Western Iowa Healthcare System in Omaha.

Issue
Federal Practitioner - 32(4)s
Publications
Topics
Sections
Author and Disclosure Information

Dr. Goyal is a house officer in the Department of Internal Medicine and Dr. Silberstein is a professor and chief of Hematology/Oncology, both at CHI Health Creighton University Medical Center in Omaha, Nebraska. Dr. Silberstein is also the chief of oncology at VA Nebraska-Western Iowa Healthcare System in Omaha.

Author and Disclosure Information

Dr. Goyal is a house officer in the Department of Internal Medicine and Dr. Silberstein is a professor and chief of Hematology/Oncology, both at CHI Health Creighton University Medical Center in Omaha, Nebraska. Dr. Silberstein is also the chief of oncology at VA Nebraska-Western Iowa Healthcare System in Omaha.

New targeted treatments and therapies for metastatic melanoma are improving patient prognosis and survival.
New targeted treatments and therapies for metastatic melanoma are improving patient prognosis and survival.

Melanoma is the most aggressive form of skin cancer, contributing to about 76,000 new cases and more than 9,000 deaths in 2014.1 Depending on the stage of the disease, 5-year melanoma survival can range from 15% to 97%. Patients with local and distant metastases have a 5-year survival of about 60% and 15%, respectively.2

The incidence of melanoma is rising, partly because of the increasing number of skin biopsies being performed.3 If melanoma is diagnosed early, surgical excision is the treatment of choice. In patients with oligometastatic disease (cancer that has spread, but only to 1 or a small number of sites), complete surgical excision of the metastases may provide prolonged overall survival (OS) and delay the need to use systemic therapy.4

Recently, many new drug therapies have shown promising results in clinical trials, which may improve the prognosis of metastatic disease. This article reviews currently available systemic treatment options for the management of metastatic melanoma, the role of cytotoxic chemotherapy and interleukin-2 (IL-2), and the latest therapies available, including immune checkpoint inhibitors.

Cytotoxic Chemotherapy and Interleukin-2

Cytotoxic chemotherapy does not have an established role in the initial treatment of metastatic melanoma. Currently, cytotoxic chemotherapy is used in patients who have not responded to immunotherapy or molecular targeted therapy. The most commonly used drugs include dacarbazine and its prodrug, temozolomide. Several studies have failed to demonstrate a survival benefit using a single-agent chemotherapy with either dacarbazine or temozolomide.5,6

Other agents used in metastatic melanoma include nitrosoureas (fotemustine), platinum compounds (cisplatin, carboplatin), vinca alkaloids (vincristine),
and taxanes (paclitaxel). None of these agents provide a survival benefit, but an objective response may be seen in a minority of cases. Combination chemotherapy regimens have not shown an advantage over singleagent dacarbazine or temozolomide.7,8

High-dose IL-2 has been used in cases of metastatic melanoma with good performance status (PS) and organ function. Studies have shown a complete response rate of 3% to 7% and a prolonged disease-free survival in a minority of patients.9-11 The use of highdose IL-2, however, is limited by the high incidence of adverse effects (AEs), which include bacterial sepsis, pulmonary edema, arrhythmias, fever, and on some occasions, death due to complications.10 The use of  IL-2 requires admission of the patient to a specialized unit for AE monitoring and management. Because of its ability to “cure” a minority of patients, a role still exists for IL-2 therapy in the treatment of younger, healthy patients with no evidence of organ dysfunction at baseline.

Immune Checkpoint Inhibitors

Checkpoint inhibitors are a class of drugs that unmask the immune system to fight against cancer cells. This class of drugs has shown significant activity and survival advantage in recent phase 2 and 3 trials. The class includes the anticytotoxic T-lymphocyte antigen 4 (CTLA-4) antibody ipilimumab and monoclonal antibodies targeting the programmed death 1 protein (PD-1) or its ligand (PD-L1).

Anti-CTLA-4 Antibodies: Ipilimumab

Cytotoxic T-lymphocyte antigen 4 is the antigen responsible for inhibition of cytotoxic T-cell-mediated immunity against foreign antigens presented by the antigen presenting cells (APCs). The APCs cause activation of the T cells when peptide fragments of intracellular proteins are presented in combination with mixed histocompatibility complex molecules. This step requires interaction of a costimulatory molecule (B7) on the APCs with a cluster of differentiation 28 protein (CD28) receptor located on T cells. CTLA-4 competes with CD28 to bind with the B7 molecule, thereby inhibiting the activation of the cytotoxic T cells (Figure 1). This pathway is thought to help with development of tolerance to host tissue antigens. Ipilimumab is a human monoclonal antibody that inhibits this CTLA-4 molecule and facilitates T-cell mediated antitumor activity.12 By blocking the CTLA-4 molecule, ipilimumab also mediates its autoimmune AEs on the host tissues.

Hodi and colleagues conducted a phase 3 trial of ipilimumab, including 676 patients who progressed after prior treatment for stage III or IV melanoma, and found that median OS was significantly better in the ipilimumab groups: 10 months in the ipilimumab plus gp100 peptide vaccine group vs 6.4 months in the gp100 vaccine alone group; 10.1 months in the ipilimumab alone group vs 6.4 months in the gp100 vaccine alone group.13 In another phase 3 trial comparing ipilimumab plus dacarbazine to dacarbazine alone, the ipilimumab group had a significantly improved OS (11.2 months vs 9.1 months).1 Survival rates with ipilimumab were prolonged for up to 3 years compared with the dacarbazine plus placebo group. However, the combination was associated with increased incidence of hepatotoxicity, thereby limiting its use.

A long-term survival analysis of 10 prospective and 2 retrospective studies of ipilimumab showed a median OS of 11.4 months and a long-term survival that began at 3 years with a plateau at 10 years of 21%, which was independent of prior therapy or ipilimumab dose.14 The immune-related AEs of ipilimumab are secondary to its activity against the host antigens and include dermatitis, enterocolitis, hepatitis, and endocrinopathies.15

A recent phase 2 trial studied the combination of ipilimumab with granulocyte-macrophage colonystimulating factor in 245 patients with stage III and IV melanoma. Median OS after 13 months was significantly higher with the combination compared with ipilimumab alone. The 1-year survival rate was 69% with
the combination and 53% with ipilimumab alone. There was no difference in the overall response rate (ORR) or progression-free survival (PFS) between the 2 groups. However, the AEs were significantly reduced with the combination (45% vs 58%).16 The dose of ipilimumab used in the trial was higher than the approved dose, making it difficult to apply the results in practice without further studies on the combination.

Anti-PD-1 Antibodies

Programmed death 1 ligands (PD-L1 and PD-L2) are expressed by tumor or stromal cells to inhibit the T-cell mediated antitumor activity. These ligands bind to the PD-1 protein on the surface of activated T cells to mediate their immunosuppressive effects. Interruption of this interaction by either anti-PD-1 antibodies or anti-PD-L1 antibodies facilitates tumor cell killing by activated T cells.17

Pembrozilumab and nivolumab are the 2 anti-PD-1 monoclonal antibodies that have been approved for treatment of metastatic melanoma. In a phase 1 trial
of pembrolizumab, 411 patients with advanced melanoma (consisting of both ipilimumab-naïve [IPI-N] and ipilimumab-treated [IPI-T] patients), ORR was 40% in IPI-N and 28% in IPI-T patients with a 1-year OS of 71% in all patients. Median PFS was 24 weeks in IPI-N and 23 weeks in IPI-T pts.18 There was no difference in outcomes and safety profiles across the various dosing regimens.18,19 Of note, pembrolizumab had antitumor activity irrespective of the PS, lactate dehydrogenase levels, BRAF (B-Raf proto-oncogene, serine/threonine kinase) gene mutation, metastatic stage, and number and type of prior therapy. In a subgroup analysis, 173 patients who had progression after treatment with ipilimumab were randomly assigned to pembrolizumab 2 mg/kg every 3 weeks (q3w) or 10 mg/kg q3w dosing regimens. Both groups had no significant difference in the ORR (26% in both) and safety profiles.20

In the 2012 KEYNOTE-002 clinical trial, a randomized phase 2 trial involving 540 patients with ipilimumab-refractory advanced melanoma, patients were randomized 1:1:1 to pembrolizumab 2 mg/kg or 10 mg/kg q3w or investigator-choice chemotherapy (control arm consisting of carboplatin plus paclitaxel, carboplatin, paclitaxel, dacarbazine, or temozolomide). The 6-month PFS was significantly improved with pembrolizumab (34% and 38% for pembrolizumab 2 mg/kg and 10 mg/kg, respectively) compared with 16% with chemotherapy. The ORR was significantly better with pembrolizumab (21% at 2 mg/kg, 25% at 10 mg/kg) compared with the control arm (4%).21 These findings led to the approval of pembrolizumab by the FDA for treatment of patients with advanced melanoma who have progressed on ipilimumab. Pembrolizumab is generally well tolerated. The most common AEs include fatigue, pruritus, and rash.

Nivolumab was studied in a recent phase 1 trial in which 107 patients with previously treated advanced melanoma were treated with escalated doses every
2 weeks.22 The 2-year and 3-year OS rates were 48% and 41%, respectively. Objective responses were seen in 32% of the patients. The median response duration was 23 months.23

The first phase 3 trial was conducted in 418 patients with previously untreated metastatic melanoma BRAF mutation. Patients were randomized to receive either nivolumab or dacarbazine. The PFS and OS were significantly better with nivolumab compared with dacarbazine (PFS 5.1 months vs 2.2 months; OS 73% vs 42% at 1 year).24 The AE profile of nivolumab is similar to pembrolizumab and includes lung, skin, endocrine, renal, and gastrointestinal tract toxicities.

Preliminary results of another phase 3 trial were presented at the European Society of Medical Oncology 2014 meeting. Patients with previously treated metastatic melanoma (ipilimumab or BRAF inhibitor) were randomized in a 2:1 ratio to receive either nivolumab or investigators’ choice chemotherapy (dacarbazine or carboplatin plus paclitaxel). The ORR was significantly better with nivolumab (32% vs 11%), and 95% of patients were still responding after 6 months. The nivolumab group showed a complete remission in 3% of the patients with 34% of the responses lasting ≥ 6 months.25 This led to the recent approval of nivolumab for patients with metastatic melanoma with a BRAF mutation who have advanced on ipilimumab. In the phase 3 NCT01844505 trial patients are being randomized to receive ipilimumab, nivolumab, or both.

A newer PD-1 inhibitor, pidilizumab, was studied in a phase 2 trial that included 103 patients with metastatic melanoma, 51% of whom had received therapy with ipilimumab. The ORR in the study group was relatively lower (6%), but the OS at 1 year was 64.5%.26 Further studies are underway to evaluate the role of this drug in metastatic melanoma.

The response with both nivolumab and pembrolizumab is durable as well as sustained, even after discontinuation of therapy. None of the deaths in the aforementioned studies were atributed to drug-related toxicities. As evidenced by current data, these 2 drugs hold a great promise for the management of patients who progress after therapy with anti-CTLA-4 antibodies.

Anti-PD-L1 Antibodies

The anti-PD-L1 monoclonal antibodies work in a similar way to the PD-1 inhibitors and block the interaction between the PD-1 and its ligand, PD-L1. This causes sustained activation of cytotoxic T cells and facilitates their antitumor activity. Two of PD-L1 inhibitors have shown clinical activity against metastatic melanoma.

BMS-936559, the first PD-L1 antibody, is being studied in a phase 1 trial that includes 55 patients with advanced melanoma along with 152 patients with other solid malignancies. Three patients achieved a complete response, and 5 patients had an objective response lasting 1 year. The ORR for melanoma was 17%, with disease stabilization of ≥ 24 weeks in 27% of the patients.27 Common AEs included infusion reactions, diarrhea, fatigue, rash, hypothyroidism, and hepatitis.

The second PD-L1 antibody, MPDL3280A, was studied in a phase 1 trial of 45 patients with metastatic melanoma. An ORR of 29% was observed, along with a 24-week PFS of 43%.28 Commonly noted AEs included hyperglycemia and elevated liver aminotransferases.

A newer PD-L1 inhibitor, MEDI4736, is being studied for advanced malignancies in 8 patients with melanoma. In preliminary analysis, MEDI4736 demonstrated a partial response in 1 out of 8 melanoma patients with a disease control rate of 46%.29 Although the PD-L1 inhibitors seem promising, more information will help discern their role in the management of metastatic melanoma.

Combined Anti-CTLA-4 Plus Anti-PD-1 Antibody

The combination of ipilimumab and the PD-1 inhibitor nivolumab was tested in a phase 1 trial in which both drugs were used concurrently as well as sequentially in metastatic melanoma.30 The 1- and 2-year OS in patients who were treated concurrently was 82% and 75%, respectively. Complete remission was seen in 17% of the patients, and the responses were seen irrespective of the BRAF mutation status. The responses were durable, and about 64% of the objective responses remain in remission at last follow-up.31 Grade 3 to grade 4 AEs were noted in 53% of the patients, with 11 patients requiring discontinuation of the medications. More studies are required to ascertain the optimum dosage of the combination prior to its approval for use in metastatic melanoma.

Molecular Targeted Therapy

The RAS-RAF–mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway is activated in almost 90% of patients
with melanoma.32 This pathway is normally required for the growth and survival of nonmalignant cells. In malignant transformation, mutations and/or overexpression is seen at various levels including KIT, NRAS, BRAF, and the MEK protein. This leads to activation of serine and threonine protein kinases, which lead to uncontrolled cell proliferation and survival.33

Novel therapeutic approaches have tried inhibiting one or more of these pathways for melanoma treatment. The most important mediator of tumorigenesis is BRAF, which is a downstream receptor of NRAS, and is mutated in almost 50% of melanoma cases.34 NRAS mutations are seen in 15% to 20% of cutaneous melanomas.35,36 After its activation, the RAF enzyme—coded by the BRAF gene—causes phosphorylation of the MEK protein, which activates ERK. This ERK activation leads to growth signaling and is the final pathway in several malignancies (Figure 2).37,38

BRAF Inhibitors

BRAF is the first mediator whose inhibition led to clinically significant outcomes in patients with melanoma. The most common BRAF mutation consists of the
substitution of glutamic acid for valine at amino acid 600 (V600E mutation) with majority of the remainder consisting of an alternate substitution (V600V or V600K).34 Vemurafenib and dabrafenib are the 2 BRAF inhibitors that have been shown to improve tumor regression, PFS, and OS considerably, especially in combination with a MEK protein inhibitor. In the phase 3 BRIM-3 trial, the vemurafenib group had a significantly prolonged PFS and OS compared with dacarbazine (13.6 months vs 9.7 months; 6.9 months vs 1.6 months, respectively). It was the first study to show improved survival with vemurafenib in both the V600E and V600K BRAF mutant melanomas.39

Another BRAF inhibitor, dabrafenib, was approved by the FDA for treatment of advanced melanoma with BRAF V600E mutation. It was tested in a phase 3 trial in which it was compared with dacarbazine in patients with advanced melanoma. Median OS in the dabrafenib arm was > 18 months and in dacarbazine arm > 15 months.40 Fifty-seven percent of the patients in dacarbazine arm were crossed over to the dabrafenib arm, thereby confounding the survival data for the former group. Another multicenter, phase 2 trial showed dabrafenib to have activity in melanoma patients with brain metastases, irrespective of previous therapy for the brain metastases.41 The long-term analysis of the BREAK-2 trial, which included 92 patients with metastatic melanoma treated with dabrafenib, showed a median OS of 12.9 months in BRAF V600K group and 13.1 months in BRAF V600E group.42

Adverse effects associated with BRAF inhibition include fatigue, rash, arthralgia, and photosensitivity reactions.43 Dermatologic complications may also include squamous cell carcinoma (SCC) (19%-26%), with keratoacanthoma being the most common subtype.44 These are believed to be likely secondary to the paradoxical activation of the MAPK signaling, since most of these lesions are found to have mutations in the RAS molecule.45 Other specific AEs of dabrafenib include hyperkeratosis (33%) and pyrexia (29%).42

Most patients treated with a BRAF inhibitor eventually have disease progression, likely secondary to reactivation of the MAPK pathway.46,47 This result has led to a heightened interest in combination therapies in an effort to improve outcomes. Combination therapy with ipilimumab and vemurafenib was studied and resulted in a higher incidence of hepatotoxicity (50%).48 However, no hepatotoxicity was seen in a phase 1 trial of combined dabrafenib and ipilimumab.49

Some studies have also suggested that extended BRAF inhibition after progression on a BRAF inhibitor may prolong survival.50,51 The phase 2 trial NCT01983124 is being conducted to evaluate the survival benefit with a combination of vemurafenib and a nitrosourea alkylating agent, fotemustine, in patients who have progressed on vemurafenib alone.

MEK Inhibitors

The inhibition of MEK can halt cell proliferation and induce apoptosis. The phase 3 METRIC trial, which compared the oral MEK inhibitor (trametinib) with chemotherapy, was conducted in 322 patients who had metastatic melanoma with a V600E or V600K BRAF mutation. The PFS and 6-month OS were significantly better with trametinib (4.8 months vs 1.5 months, 81% vs 66%) despite the crossover between the 2 groups.52 The AEs associated with trametinib included rash, diarrhea, and peripheral edema. Another phase 2 trial of trametinib including patients pretreated with a BRAF inhibitor showed no confirmed objective responses, 28% patients with stable disease, and minimal improvement in PFS (2 months). Among patients treated with prior chemotherapy and/or immunotherapy, trametinib showed significant improvement in complete responses, partial responses, stable disease, and the median PFS (2%, 23%, 51%, 4 months, respectively).53

The second MEK inhibitor, binimetinib, was studied in a phase 2 trial of advanced melanoma cases harboring a BRAF V600E or NRAS. Bimetinib demonstrated a PR in 20% cases of both the BRAF and NRAS mutant melanomas. Durable disease control was seen in 43% of the NRAS group and 32% of the BRAF group.54 The AE profile was similar to that seen with trametinib. Bimetinib is being studied in phase 1 and 2 trials with the CDK4/6 inhibitor as well as in the phase 3 trial NCT01763164 compared with dacarbazine in NRAS mutation positive melanomas.55

Selumetinib is a MEK inhibitor that has been compared with dacarbazine and temozolomide with no significant OS advantage. A novel highly specific inhibitor of MEK, cobimetinib, is currently being studied in combination with BRAF inhibitors.

Combined BRAF and MEK Inhibition

A randomized, double-blind, phase 3 study comparing the combination of dabrafenib and trametinib with dabrafenib and placebo in patients with advanced melanoma with a BRAF V600E mutation was presented at the 2014 American Society of Clinical Oncology meeting. Researchers found that after a median follow-up period of 9 months, there was a significant improvement with the combination in the PFS (9.3 months vs 8.8 months) and the ORR (67% vs 51%), with a similar incidence of AEs.56 The combination therapy group had fewer incidences of SCC of the skin but more incidence of pyrexia.

The combination of dabrafenib and trametinib was compared with vemurafenib monotherapy in a recent randomized phase 3 trial among 704 metastatic melanoma patients with a BRAF V600 mutation. Median PFS and ORR were significantly better with combination therapy compared with vemurafenib alone (11.4 months vs 7.3 months, 64% vs 51%, respectively). Overall survival rate at 1 year was significantly improved in the combination group as well (72% vs 65%).57 The incidence of SCC and keratoacanthoma was less in the combination (1%) compared with vemurafenib alone (18%). Another study investigating the coadministration and sequential administration of vemurafenib and trametinib is underway.58

The vemurafenib and cobimetinib combination was studied in a phase 3 trial of previously untreated unresectable locally advanced or metastatic BRAF V600
mutation-positive melanoma. The median PFS was 9.9 months in the combination group and 6.2 months in the control group. The interim analysis showed a 9-month survival rate of 81% in the combination group and 73% in the control group, with no significantly higher incidence of AEs in either arm.59 A longer follow-up will be needed to assess the OS benefit with the combination.

Encorafenib, a selective BRAF inhibitor, has been studied in a phase 1 trial in combination with binimetinib.60 This trial has paved the way to the initiation of a currently ongoing phase 3 trial (NCT01909453) comparing the combination with vemurafenib or encorafenib alone.

C-KIT Inhibitors

Mutations of c-KIT are seen more commonly in chronic sun damage-induced cutaneous melanomas, along with acral and mucosal melanomas.61,62 Earlier trials involving patients without selection for c-KIT mutation positivity failed to show benefit with imatinib. A single-arm, phase 2 trial of imatinib mesylate in patients with metastatic melanoma harboring the c-KIT mutation, an ORR of 23% was achieved, with a median PFS of 3.5 months.63 Imatinib showed an ORR of 29% in a phase 2 trial of mucosal, acral, and in chronic sun damage-induced melanoma patients with c-KIT amplifications and/or mutations. It was demonstrated that c-KIT amplification alone is not as responsive to imatinib compared with c-KIT mutation, suggesting that all patients with these specific melanomas should be tested for KIT mutation status.64

A second-generation c-KIT inhibitor, nilotinib, has shown some promising results with a favorable AE profile in small phase 2 trials.65,66 However, more clinical research will be needed before definite recommendations on its use in cutaneous melanomas can be made. Currently, its role seems to be limited to the management of acral, mucosal, and chronic sun damage-related melanomas with c-KIT mutations.

Future Directions

Angiogenesis promoters, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor, fibroblast growth factor, and interleukin-8, are overexpressed in melanoma. Bevacizumab, an anti-VEGF antibody, has been shown to have some benefit in combination with carboplatin and paclitaxel as a triple therapy.67 However, grade 3 AEs were seen in a portion of patients.

The phosphatidylinositol-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway has also been studied as a target for melanoma therapy. Everolimus, an mTOR inhibitor, was studied in a phase 2 trial in combination with bevacizumab for treatment of metastatic melanoma. The combination showed improved median PFS and OS with the combination (4 months and 8.6 months, respectively), with 43% of patients alive after 12 months of follow-up.68 This study points to the direction of possible benefits with the combination of anti-VEGF and immunotherapy. A recent study failed to show survival advantage with combination of bevacizumab and temozolomide.69

Buparlisib (BKM120), a PI3K inhibitor, has been shown to have activity in vivo and in vitro against melanoma brain metastases.70 More studies need to be done to assess the possible combination with other established therapies.

Oblimersen is an antisense oligonucleotide that suppresses B-cell lymphoma-2, thereby suppressing its anti-apoptotic effect. The triple combination of oblimersen with temozolomide and albumin-bound paclitaxel has shown to be safe and efficacious in a phase 1 trial, thereby creating a need for further clinical trials.71

Treatment Approach

Systemic therapy for metastatic melanoma depends on several factors, including BRAF mutation status, functional status of the patient, disease burden, and severity of symptoms. Assessing the BRAF mutation status has become an important component in the management of patients with metastatic melanoma. It can help recognize patients who will benefit from molecular targeted therapy. In case of a BRAF-positive melanoma, treatment can be initiated with either immunotherapy or BRAF inhibitors. There are no randomized studies comparing immunotherapy to molecular targeted therapy.

Patients who have good PS and lymph node metastases can be treated initially with IL-2, which has the advantage of inducing cure in a minority of patients but should only be considered in patients with well-preserved organ function who can be monitored in an intensive care setting. On the other hand, patients who have bulky, symptomatic disease and poor PS should be treated initially with BRAF inhibitors. Combination of BRAF and MEK inhibitors can also be used and has an improved PFS and OS with potential to cause early tumor regression. There are studies to suggest suboptimal outcomes in patients who are treated with ipilimumab after progression on a BRAF inhibitor compared with initial treatment with ipilimumab followed by a BRAF inhibitor.72-74 However, all these studies are retrospective and there is no prospective data to suggest the above. BRAF mutation-positive patients who progress on a BRAF inhibitor
can be treated with PD-1 inhibitors.

Patients who do not have a BRAF mutation are unlikely to benefit from a BRAF inhibitor and primarily receive immunotherapy with ipilimumab or IL-2. Whenever possible, such patients should be enrolled in a clinical trial, as they have a poor prognosis. Patients who progress on ipilimumab can be treated with one of the PD-1 inhibitors (pembrolizumab, nivolumab). These PD-L1 inhibitors are still being investigated for use in such situations.

The role of chemotherapy in the management of metastatic melanoma has been limited by numerous studies showing significantly better survival with immunotherapy and molecular targeted therapy. Dacarbazine is the only FDA-approved drug for the treatment of melanoma. Its use is reserved mainly for patients who are not candidates for any of the other therapies available, including enrollment in a clinical trial.

Conclusion

Therapies for metastatic melanoma are in a state of flux. In the past decade, several new therapeutic agents have been introduced for the management of this potentially lethal disease. The treatment of metastatic melanoma has gradually shifted from cytotoxic chemotherapy toward a more individualized treatment that has a definite survival advantage over traditional counterparts. The advent of novel therapies has led to initiation of further studies to determine their role in the treatment of advanced melanoma, singly or in combination with other agents. In addition to evaluating new agents, more studies are needed to compare existing treatment modalities so that definitive treatment protocols can be formulated.

Acknowledgement
The authors would like to thank Felicia Ratnaraj, MD, for her assistance in creating the figures.

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

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

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Click here to read the digital edition.

Melanoma is the most aggressive form of skin cancer, contributing to about 76,000 new cases and more than 9,000 deaths in 2014.1 Depending on the stage of the disease, 5-year melanoma survival can range from 15% to 97%. Patients with local and distant metastases have a 5-year survival of about 60% and 15%, respectively.2

The incidence of melanoma is rising, partly because of the increasing number of skin biopsies being performed.3 If melanoma is diagnosed early, surgical excision is the treatment of choice. In patients with oligometastatic disease (cancer that has spread, but only to 1 or a small number of sites), complete surgical excision of the metastases may provide prolonged overall survival (OS) and delay the need to use systemic therapy.4

Recently, many new drug therapies have shown promising results in clinical trials, which may improve the prognosis of metastatic disease. This article reviews currently available systemic treatment options for the management of metastatic melanoma, the role of cytotoxic chemotherapy and interleukin-2 (IL-2), and the latest therapies available, including immune checkpoint inhibitors.

Cytotoxic Chemotherapy and Interleukin-2

Cytotoxic chemotherapy does not have an established role in the initial treatment of metastatic melanoma. Currently, cytotoxic chemotherapy is used in patients who have not responded to immunotherapy or molecular targeted therapy. The most commonly used drugs include dacarbazine and its prodrug, temozolomide. Several studies have failed to demonstrate a survival benefit using a single-agent chemotherapy with either dacarbazine or temozolomide.5,6

Other agents used in metastatic melanoma include nitrosoureas (fotemustine), platinum compounds (cisplatin, carboplatin), vinca alkaloids (vincristine),
and taxanes (paclitaxel). None of these agents provide a survival benefit, but an objective response may be seen in a minority of cases. Combination chemotherapy regimens have not shown an advantage over singleagent dacarbazine or temozolomide.7,8

High-dose IL-2 has been used in cases of metastatic melanoma with good performance status (PS) and organ function. Studies have shown a complete response rate of 3% to 7% and a prolonged disease-free survival in a minority of patients.9-11 The use of highdose IL-2, however, is limited by the high incidence of adverse effects (AEs), which include bacterial sepsis, pulmonary edema, arrhythmias, fever, and on some occasions, death due to complications.10 The use of  IL-2 requires admission of the patient to a specialized unit for AE monitoring and management. Because of its ability to “cure” a minority of patients, a role still exists for IL-2 therapy in the treatment of younger, healthy patients with no evidence of organ dysfunction at baseline.

Immune Checkpoint Inhibitors

Checkpoint inhibitors are a class of drugs that unmask the immune system to fight against cancer cells. This class of drugs has shown significant activity and survival advantage in recent phase 2 and 3 trials. The class includes the anticytotoxic T-lymphocyte antigen 4 (CTLA-4) antibody ipilimumab and monoclonal antibodies targeting the programmed death 1 protein (PD-1) or its ligand (PD-L1).

Anti-CTLA-4 Antibodies: Ipilimumab

Cytotoxic T-lymphocyte antigen 4 is the antigen responsible for inhibition of cytotoxic T-cell-mediated immunity against foreign antigens presented by the antigen presenting cells (APCs). The APCs cause activation of the T cells when peptide fragments of intracellular proteins are presented in combination with mixed histocompatibility complex molecules. This step requires interaction of a costimulatory molecule (B7) on the APCs with a cluster of differentiation 28 protein (CD28) receptor located on T cells. CTLA-4 competes with CD28 to bind with the B7 molecule, thereby inhibiting the activation of the cytotoxic T cells (Figure 1). This pathway is thought to help with development of tolerance to host tissue antigens. Ipilimumab is a human monoclonal antibody that inhibits this CTLA-4 molecule and facilitates T-cell mediated antitumor activity.12 By blocking the CTLA-4 molecule, ipilimumab also mediates its autoimmune AEs on the host tissues.

Hodi and colleagues conducted a phase 3 trial of ipilimumab, including 676 patients who progressed after prior treatment for stage III or IV melanoma, and found that median OS was significantly better in the ipilimumab groups: 10 months in the ipilimumab plus gp100 peptide vaccine group vs 6.4 months in the gp100 vaccine alone group; 10.1 months in the ipilimumab alone group vs 6.4 months in the gp100 vaccine alone group.13 In another phase 3 trial comparing ipilimumab plus dacarbazine to dacarbazine alone, the ipilimumab group had a significantly improved OS (11.2 months vs 9.1 months).1 Survival rates with ipilimumab were prolonged for up to 3 years compared with the dacarbazine plus placebo group. However, the combination was associated with increased incidence of hepatotoxicity, thereby limiting its use.

A long-term survival analysis of 10 prospective and 2 retrospective studies of ipilimumab showed a median OS of 11.4 months and a long-term survival that began at 3 years with a plateau at 10 years of 21%, which was independent of prior therapy or ipilimumab dose.14 The immune-related AEs of ipilimumab are secondary to its activity against the host antigens and include dermatitis, enterocolitis, hepatitis, and endocrinopathies.15

A recent phase 2 trial studied the combination of ipilimumab with granulocyte-macrophage colonystimulating factor in 245 patients with stage III and IV melanoma. Median OS after 13 months was significantly higher with the combination compared with ipilimumab alone. The 1-year survival rate was 69% with
the combination and 53% with ipilimumab alone. There was no difference in the overall response rate (ORR) or progression-free survival (PFS) between the 2 groups. However, the AEs were significantly reduced with the combination (45% vs 58%).16 The dose of ipilimumab used in the trial was higher than the approved dose, making it difficult to apply the results in practice without further studies on the combination.

Anti-PD-1 Antibodies

Programmed death 1 ligands (PD-L1 and PD-L2) are expressed by tumor or stromal cells to inhibit the T-cell mediated antitumor activity. These ligands bind to the PD-1 protein on the surface of activated T cells to mediate their immunosuppressive effects. Interruption of this interaction by either anti-PD-1 antibodies or anti-PD-L1 antibodies facilitates tumor cell killing by activated T cells.17

Pembrozilumab and nivolumab are the 2 anti-PD-1 monoclonal antibodies that have been approved for treatment of metastatic melanoma. In a phase 1 trial
of pembrolizumab, 411 patients with advanced melanoma (consisting of both ipilimumab-naïve [IPI-N] and ipilimumab-treated [IPI-T] patients), ORR was 40% in IPI-N and 28% in IPI-T patients with a 1-year OS of 71% in all patients. Median PFS was 24 weeks in IPI-N and 23 weeks in IPI-T pts.18 There was no difference in outcomes and safety profiles across the various dosing regimens.18,19 Of note, pembrolizumab had antitumor activity irrespective of the PS, lactate dehydrogenase levels, BRAF (B-Raf proto-oncogene, serine/threonine kinase) gene mutation, metastatic stage, and number and type of prior therapy. In a subgroup analysis, 173 patients who had progression after treatment with ipilimumab were randomly assigned to pembrolizumab 2 mg/kg every 3 weeks (q3w) or 10 mg/kg q3w dosing regimens. Both groups had no significant difference in the ORR (26% in both) and safety profiles.20

In the 2012 KEYNOTE-002 clinical trial, a randomized phase 2 trial involving 540 patients with ipilimumab-refractory advanced melanoma, patients were randomized 1:1:1 to pembrolizumab 2 mg/kg or 10 mg/kg q3w or investigator-choice chemotherapy (control arm consisting of carboplatin plus paclitaxel, carboplatin, paclitaxel, dacarbazine, or temozolomide). The 6-month PFS was significantly improved with pembrolizumab (34% and 38% for pembrolizumab 2 mg/kg and 10 mg/kg, respectively) compared with 16% with chemotherapy. The ORR was significantly better with pembrolizumab (21% at 2 mg/kg, 25% at 10 mg/kg) compared with the control arm (4%).21 These findings led to the approval of pembrolizumab by the FDA for treatment of patients with advanced melanoma who have progressed on ipilimumab. Pembrolizumab is generally well tolerated. The most common AEs include fatigue, pruritus, and rash.

Nivolumab was studied in a recent phase 1 trial in which 107 patients with previously treated advanced melanoma were treated with escalated doses every
2 weeks.22 The 2-year and 3-year OS rates were 48% and 41%, respectively. Objective responses were seen in 32% of the patients. The median response duration was 23 months.23

The first phase 3 trial was conducted in 418 patients with previously untreated metastatic melanoma BRAF mutation. Patients were randomized to receive either nivolumab or dacarbazine. The PFS and OS were significantly better with nivolumab compared with dacarbazine (PFS 5.1 months vs 2.2 months; OS 73% vs 42% at 1 year).24 The AE profile of nivolumab is similar to pembrolizumab and includes lung, skin, endocrine, renal, and gastrointestinal tract toxicities.

Preliminary results of another phase 3 trial were presented at the European Society of Medical Oncology 2014 meeting. Patients with previously treated metastatic melanoma (ipilimumab or BRAF inhibitor) were randomized in a 2:1 ratio to receive either nivolumab or investigators’ choice chemotherapy (dacarbazine or carboplatin plus paclitaxel). The ORR was significantly better with nivolumab (32% vs 11%), and 95% of patients were still responding after 6 months. The nivolumab group showed a complete remission in 3% of the patients with 34% of the responses lasting ≥ 6 months.25 This led to the recent approval of nivolumab for patients with metastatic melanoma with a BRAF mutation who have advanced on ipilimumab. In the phase 3 NCT01844505 trial patients are being randomized to receive ipilimumab, nivolumab, or both.

A newer PD-1 inhibitor, pidilizumab, was studied in a phase 2 trial that included 103 patients with metastatic melanoma, 51% of whom had received therapy with ipilimumab. The ORR in the study group was relatively lower (6%), but the OS at 1 year was 64.5%.26 Further studies are underway to evaluate the role of this drug in metastatic melanoma.

The response with both nivolumab and pembrolizumab is durable as well as sustained, even after discontinuation of therapy. None of the deaths in the aforementioned studies were atributed to drug-related toxicities. As evidenced by current data, these 2 drugs hold a great promise for the management of patients who progress after therapy with anti-CTLA-4 antibodies.

Anti-PD-L1 Antibodies

The anti-PD-L1 monoclonal antibodies work in a similar way to the PD-1 inhibitors and block the interaction between the PD-1 and its ligand, PD-L1. This causes sustained activation of cytotoxic T cells and facilitates their antitumor activity. Two of PD-L1 inhibitors have shown clinical activity against metastatic melanoma.

BMS-936559, the first PD-L1 antibody, is being studied in a phase 1 trial that includes 55 patients with advanced melanoma along with 152 patients with other solid malignancies. Three patients achieved a complete response, and 5 patients had an objective response lasting 1 year. The ORR for melanoma was 17%, with disease stabilization of ≥ 24 weeks in 27% of the patients.27 Common AEs included infusion reactions, diarrhea, fatigue, rash, hypothyroidism, and hepatitis.

The second PD-L1 antibody, MPDL3280A, was studied in a phase 1 trial of 45 patients with metastatic melanoma. An ORR of 29% was observed, along with a 24-week PFS of 43%.28 Commonly noted AEs included hyperglycemia and elevated liver aminotransferases.

A newer PD-L1 inhibitor, MEDI4736, is being studied for advanced malignancies in 8 patients with melanoma. In preliminary analysis, MEDI4736 demonstrated a partial response in 1 out of 8 melanoma patients with a disease control rate of 46%.29 Although the PD-L1 inhibitors seem promising, more information will help discern their role in the management of metastatic melanoma.

Combined Anti-CTLA-4 Plus Anti-PD-1 Antibody

The combination of ipilimumab and the PD-1 inhibitor nivolumab was tested in a phase 1 trial in which both drugs were used concurrently as well as sequentially in metastatic melanoma.30 The 1- and 2-year OS in patients who were treated concurrently was 82% and 75%, respectively. Complete remission was seen in 17% of the patients, and the responses were seen irrespective of the BRAF mutation status. The responses were durable, and about 64% of the objective responses remain in remission at last follow-up.31 Grade 3 to grade 4 AEs were noted in 53% of the patients, with 11 patients requiring discontinuation of the medications. More studies are required to ascertain the optimum dosage of the combination prior to its approval for use in metastatic melanoma.

Molecular Targeted Therapy

The RAS-RAF–mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway is activated in almost 90% of patients
with melanoma.32 This pathway is normally required for the growth and survival of nonmalignant cells. In malignant transformation, mutations and/or overexpression is seen at various levels including KIT, NRAS, BRAF, and the MEK protein. This leads to activation of serine and threonine protein kinases, which lead to uncontrolled cell proliferation and survival.33

Novel therapeutic approaches have tried inhibiting one or more of these pathways for melanoma treatment. The most important mediator of tumorigenesis is BRAF, which is a downstream receptor of NRAS, and is mutated in almost 50% of melanoma cases.34 NRAS mutations are seen in 15% to 20% of cutaneous melanomas.35,36 After its activation, the RAF enzyme—coded by the BRAF gene—causes phosphorylation of the MEK protein, which activates ERK. This ERK activation leads to growth signaling and is the final pathway in several malignancies (Figure 2).37,38

BRAF Inhibitors

BRAF is the first mediator whose inhibition led to clinically significant outcomes in patients with melanoma. The most common BRAF mutation consists of the
substitution of glutamic acid for valine at amino acid 600 (V600E mutation) with majority of the remainder consisting of an alternate substitution (V600V or V600K).34 Vemurafenib and dabrafenib are the 2 BRAF inhibitors that have been shown to improve tumor regression, PFS, and OS considerably, especially in combination with a MEK protein inhibitor. In the phase 3 BRIM-3 trial, the vemurafenib group had a significantly prolonged PFS and OS compared with dacarbazine (13.6 months vs 9.7 months; 6.9 months vs 1.6 months, respectively). It was the first study to show improved survival with vemurafenib in both the V600E and V600K BRAF mutant melanomas.39

Another BRAF inhibitor, dabrafenib, was approved by the FDA for treatment of advanced melanoma with BRAF V600E mutation. It was tested in a phase 3 trial in which it was compared with dacarbazine in patients with advanced melanoma. Median OS in the dabrafenib arm was > 18 months and in dacarbazine arm > 15 months.40 Fifty-seven percent of the patients in dacarbazine arm were crossed over to the dabrafenib arm, thereby confounding the survival data for the former group. Another multicenter, phase 2 trial showed dabrafenib to have activity in melanoma patients with brain metastases, irrespective of previous therapy for the brain metastases.41 The long-term analysis of the BREAK-2 trial, which included 92 patients with metastatic melanoma treated with dabrafenib, showed a median OS of 12.9 months in BRAF V600K group and 13.1 months in BRAF V600E group.42

Adverse effects associated with BRAF inhibition include fatigue, rash, arthralgia, and photosensitivity reactions.43 Dermatologic complications may also include squamous cell carcinoma (SCC) (19%-26%), with keratoacanthoma being the most common subtype.44 These are believed to be likely secondary to the paradoxical activation of the MAPK signaling, since most of these lesions are found to have mutations in the RAS molecule.45 Other specific AEs of dabrafenib include hyperkeratosis (33%) and pyrexia (29%).42

Most patients treated with a BRAF inhibitor eventually have disease progression, likely secondary to reactivation of the MAPK pathway.46,47 This result has led to a heightened interest in combination therapies in an effort to improve outcomes. Combination therapy with ipilimumab and vemurafenib was studied and resulted in a higher incidence of hepatotoxicity (50%).48 However, no hepatotoxicity was seen in a phase 1 trial of combined dabrafenib and ipilimumab.49

Some studies have also suggested that extended BRAF inhibition after progression on a BRAF inhibitor may prolong survival.50,51 The phase 2 trial NCT01983124 is being conducted to evaluate the survival benefit with a combination of vemurafenib and a nitrosourea alkylating agent, fotemustine, in patients who have progressed on vemurafenib alone.

MEK Inhibitors

The inhibition of MEK can halt cell proliferation and induce apoptosis. The phase 3 METRIC trial, which compared the oral MEK inhibitor (trametinib) with chemotherapy, was conducted in 322 patients who had metastatic melanoma with a V600E or V600K BRAF mutation. The PFS and 6-month OS were significantly better with trametinib (4.8 months vs 1.5 months, 81% vs 66%) despite the crossover between the 2 groups.52 The AEs associated with trametinib included rash, diarrhea, and peripheral edema. Another phase 2 trial of trametinib including patients pretreated with a BRAF inhibitor showed no confirmed objective responses, 28% patients with stable disease, and minimal improvement in PFS (2 months). Among patients treated with prior chemotherapy and/or immunotherapy, trametinib showed significant improvement in complete responses, partial responses, stable disease, and the median PFS (2%, 23%, 51%, 4 months, respectively).53

The second MEK inhibitor, binimetinib, was studied in a phase 2 trial of advanced melanoma cases harboring a BRAF V600E or NRAS. Bimetinib demonstrated a PR in 20% cases of both the BRAF and NRAS mutant melanomas. Durable disease control was seen in 43% of the NRAS group and 32% of the BRAF group.54 The AE profile was similar to that seen with trametinib. Bimetinib is being studied in phase 1 and 2 trials with the CDK4/6 inhibitor as well as in the phase 3 trial NCT01763164 compared with dacarbazine in NRAS mutation positive melanomas.55

Selumetinib is a MEK inhibitor that has been compared with dacarbazine and temozolomide with no significant OS advantage. A novel highly specific inhibitor of MEK, cobimetinib, is currently being studied in combination with BRAF inhibitors.

Combined BRAF and MEK Inhibition

A randomized, double-blind, phase 3 study comparing the combination of dabrafenib and trametinib with dabrafenib and placebo in patients with advanced melanoma with a BRAF V600E mutation was presented at the 2014 American Society of Clinical Oncology meeting. Researchers found that after a median follow-up period of 9 months, there was a significant improvement with the combination in the PFS (9.3 months vs 8.8 months) and the ORR (67% vs 51%), with a similar incidence of AEs.56 The combination therapy group had fewer incidences of SCC of the skin but more incidence of pyrexia.

The combination of dabrafenib and trametinib was compared with vemurafenib monotherapy in a recent randomized phase 3 trial among 704 metastatic melanoma patients with a BRAF V600 mutation. Median PFS and ORR were significantly better with combination therapy compared with vemurafenib alone (11.4 months vs 7.3 months, 64% vs 51%, respectively). Overall survival rate at 1 year was significantly improved in the combination group as well (72% vs 65%).57 The incidence of SCC and keratoacanthoma was less in the combination (1%) compared with vemurafenib alone (18%). Another study investigating the coadministration and sequential administration of vemurafenib and trametinib is underway.58

The vemurafenib and cobimetinib combination was studied in a phase 3 trial of previously untreated unresectable locally advanced or metastatic BRAF V600
mutation-positive melanoma. The median PFS was 9.9 months in the combination group and 6.2 months in the control group. The interim analysis showed a 9-month survival rate of 81% in the combination group and 73% in the control group, with no significantly higher incidence of AEs in either arm.59 A longer follow-up will be needed to assess the OS benefit with the combination.

Encorafenib, a selective BRAF inhibitor, has been studied in a phase 1 trial in combination with binimetinib.60 This trial has paved the way to the initiation of a currently ongoing phase 3 trial (NCT01909453) comparing the combination with vemurafenib or encorafenib alone.

C-KIT Inhibitors

Mutations of c-KIT are seen more commonly in chronic sun damage-induced cutaneous melanomas, along with acral and mucosal melanomas.61,62 Earlier trials involving patients without selection for c-KIT mutation positivity failed to show benefit with imatinib. A single-arm, phase 2 trial of imatinib mesylate in patients with metastatic melanoma harboring the c-KIT mutation, an ORR of 23% was achieved, with a median PFS of 3.5 months.63 Imatinib showed an ORR of 29% in a phase 2 trial of mucosal, acral, and in chronic sun damage-induced melanoma patients with c-KIT amplifications and/or mutations. It was demonstrated that c-KIT amplification alone is not as responsive to imatinib compared with c-KIT mutation, suggesting that all patients with these specific melanomas should be tested for KIT mutation status.64

A second-generation c-KIT inhibitor, nilotinib, has shown some promising results with a favorable AE profile in small phase 2 trials.65,66 However, more clinical research will be needed before definite recommendations on its use in cutaneous melanomas can be made. Currently, its role seems to be limited to the management of acral, mucosal, and chronic sun damage-related melanomas with c-KIT mutations.

Future Directions

Angiogenesis promoters, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor, fibroblast growth factor, and interleukin-8, are overexpressed in melanoma. Bevacizumab, an anti-VEGF antibody, has been shown to have some benefit in combination with carboplatin and paclitaxel as a triple therapy.67 However, grade 3 AEs were seen in a portion of patients.

The phosphatidylinositol-3 kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway has also been studied as a target for melanoma therapy. Everolimus, an mTOR inhibitor, was studied in a phase 2 trial in combination with bevacizumab for treatment of metastatic melanoma. The combination showed improved median PFS and OS with the combination (4 months and 8.6 months, respectively), with 43% of patients alive after 12 months of follow-up.68 This study points to the direction of possible benefits with the combination of anti-VEGF and immunotherapy. A recent study failed to show survival advantage with combination of bevacizumab and temozolomide.69

Buparlisib (BKM120), a PI3K inhibitor, has been shown to have activity in vivo and in vitro against melanoma brain metastases.70 More studies need to be done to assess the possible combination with other established therapies.

Oblimersen is an antisense oligonucleotide that suppresses B-cell lymphoma-2, thereby suppressing its anti-apoptotic effect. The triple combination of oblimersen with temozolomide and albumin-bound paclitaxel has shown to be safe and efficacious in a phase 1 trial, thereby creating a need for further clinical trials.71

Treatment Approach

Systemic therapy for metastatic melanoma depends on several factors, including BRAF mutation status, functional status of the patient, disease burden, and severity of symptoms. Assessing the BRAF mutation status has become an important component in the management of patients with metastatic melanoma. It can help recognize patients who will benefit from molecular targeted therapy. In case of a BRAF-positive melanoma, treatment can be initiated with either immunotherapy or BRAF inhibitors. There are no randomized studies comparing immunotherapy to molecular targeted therapy.

Patients who have good PS and lymph node metastases can be treated initially with IL-2, which has the advantage of inducing cure in a minority of patients but should only be considered in patients with well-preserved organ function who can be monitored in an intensive care setting. On the other hand, patients who have bulky, symptomatic disease and poor PS should be treated initially with BRAF inhibitors. Combination of BRAF and MEK inhibitors can also be used and has an improved PFS and OS with potential to cause early tumor regression. There are studies to suggest suboptimal outcomes in patients who are treated with ipilimumab after progression on a BRAF inhibitor compared with initial treatment with ipilimumab followed by a BRAF inhibitor.72-74 However, all these studies are retrospective and there is no prospective data to suggest the above. BRAF mutation-positive patients who progress on a BRAF inhibitor
can be treated with PD-1 inhibitors.

Patients who do not have a BRAF mutation are unlikely to benefit from a BRAF inhibitor and primarily receive immunotherapy with ipilimumab or IL-2. Whenever possible, such patients should be enrolled in a clinical trial, as they have a poor prognosis. Patients who progress on ipilimumab can be treated with one of the PD-1 inhibitors (pembrolizumab, nivolumab). These PD-L1 inhibitors are still being investigated for use in such situations.

The role of chemotherapy in the management of metastatic melanoma has been limited by numerous studies showing significantly better survival with immunotherapy and molecular targeted therapy. Dacarbazine is the only FDA-approved drug for the treatment of melanoma. Its use is reserved mainly for patients who are not candidates for any of the other therapies available, including enrollment in a clinical trial.

Conclusion

Therapies for metastatic melanoma are in a state of flux. In the past decade, several new therapeutic agents have been introduced for the management of this potentially lethal disease. The treatment of metastatic melanoma has gradually shifted from cytotoxic chemotherapy toward a more individualized treatment that has a definite survival advantage over traditional counterparts. The advent of novel therapies has led to initiation of further studies to determine their role in the treatment of advanced melanoma, singly or in combination with other agents. In addition to evaluating new agents, more studies are needed to compare existing treatment modalities so that definitive treatment protocols can be formulated.

Acknowledgement
The authors would like to thank Felicia Ratnaraj, MD, for her assistance in creating the figures.

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

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

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Click here to read the digital edition.

References

1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin.2014;64(1):9-29.

2. Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27(36):6199-6206.

3. Welch HG, Woloshin S, Schwartz LM. Skin biopsy rates and incidence of melanoma:
population based ecological study. BMJ. 2005;331(7515):481.

4. Sosman JA, Moon J, Tuthill RJ, et al. A phase 2 trial of complete resection for stage IV melanoma: results of Southwest Oncology Group Clinical Trial S9430. Cancer. 2011;117(20):4740-4746.

5. Atkins MB. The role of cytotoxic chemotherapeutic agents either alone or in combination with biological response modifiers. In: Kirkwood JK, ed. Molecular Diagnosis, Prevention, & Therapy of Melanoma. New York, NY: Marcel Dekker;1997:219-225.

6. Patel PM, Suciu S, Mortier L, et al. Extended schedule, escalated dose temozolomide versus dacarbazine in stage IV melanoma: final results of a randomised phase III study (EORTC 18032). Eur J Cancer. 2011;47(10):1476-1483.

7. Chapman PB, Einhorn LH, Meyers ML, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 1999;17(9):2745-2751.

8. Flaherty KT, Lee SJ, Zhao F, et al. Phase III trial of carboplatin and paclitaxel with
or without sorafenib in metastatic melanoma. J Clin Oncol. 2013;31(3):373-379.

9. Rosenberg SA, Yang JC, Topalian SL, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. 1994;271(12):907-913.

10. Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol. 1999;17(7):2105-2116.

11. Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am. 2000;6(suppl 1):S11-S14.

12. Hoos A, Ibrahim R, Korman A, et al. Development of ipilimumab: contribution to a new paradigm for cancer immunotherapy. Semin Oncol. 2010;37(5):533-546.

13. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723.

14. Schadendorf D, Hodi FS, Robert C, et. al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma [published online ahead of print February 9, 2015]. J Clin Oncol. pii:JCO.2014.56.2736.

15. Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691-2697.

16. Hodi FS, Lee S, McDermott DF, et al. Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: a randomized clinical trial. JAMA. 2014;312(17):1744-1753.

17. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443-2454.

18. Ribas A, Hodi FS, Kefford R, et al. Efficacy and safety of the anti-PD-1 monoclonal antibody pembrolizumab (MK-3475) in 411 patients (pts) with melanoma (MEL) (Abstract LBA9000). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

19. Hamid O, Robert C, Ribas A, et al. Randomized comparison of two doses of the anti-PD-1 monoclonal antibody MK-3475 for ipilimumab-refractory (IPI-R) and IPI-naive (IPI-N) melanoma (MEL) (abstract 3000). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

20. Robert C, Ribas A, Wolchok JD, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014; 384(9948):1109-1117.

21. Dummer R, Daud A, Puzanov I, et. al. A randomized controlled comparison of pembrolizumab and chemotherapy in patients with ipilimumab-refractory melanoma. J Transl Med. 2015;13(suppl 1):O5.

22. Topalian SL, Sznol M, McDermott DF, et. al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020-1030.

23. Hodi FS, Sznol M, Kluger HM, et al. Long-term survival of ipilimumab-naive patients with advanced melanoma (MEL) treated with nivolumab (anti-PD-1, BMS-936558, ONO-4538) in a phase I trial (abstract 9002). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

24. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320-330.

25. Weber J, D’Angelo S, Gutzmer R, et al. A phase 3 randomized, open-label study of nivolumab versus investigator’s choice of chemotherapy in patients with advanced melanoma after prior anti-CTLA4 therapy (abstract LBA3). Paper presented at: European Society of Medical Oncology 2014 meeting; September 2014; Madrid, Spain.

26. Atkins MB, Kudchadkar RR, Sznol M, et al. Phase 2, multicenter, safety and efficacy study of pidilizumab in patients with metastatic melanoma (abstract 9001). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

27. Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455-2465.

28. Hamid O, Sosman JA, Lawrence DP, et. al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). J Clin Oncol. 2013;31(15)(suppl): Abstract 9010.

29. Lutzky J, Antonia SJ, Blake-Haskins A, et. al. A phase 1 study of MEDI4736, an anti–PD-L1 antibody, in patients with advanced solid tumors. J Clin Oncol. 2014;32(15)(suppl): Abstract 3001.

30. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced
melanoma. N Engl J Med. 2013;369(2):122-133.

31. Sznol M, Kluger HM, Callahan MK, et al. Survival, response duration, and activity by BRAF mutation (MT) status of nivolumab (NIVO, anti-PD-1, BMS-936558, ONO-4538) and ipilimumab (IPI) concurrent therapy in advanced melanoma (MEL) (abstract LBA9003). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

32. Omholt K, Platz A, Kanter L, Ringborg U, Hansson J. NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progression. Clin Cancer Res. 2003;9(17):6483-6488.

33. Wellbrock C, Hurlstone A. BRAF as therapeutic target in melanoma. Biochem Pharmacol. 2010;80(5):561-567.

34. Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol. 2011;29(10):1239-1246.

35. Ball NJ, Yohn JJ, Morelli JG, et al. Ras mutations in human melanoma: a marker of malignant progression. J Invest Dermatol. 1994;102(3):285-290.

36. Platz A, Ringborg U, Brahme EM, Lagerlöf B. Melanoma metastases from patients with hereditary cutaneous malignant melanoma contain a high frequency of N-ras activating mutations. Melanoma Res. 1994;4(3):169-177.

37. Beeram M, Patnaik A, Rowinsky EK. Raf: a strategic target for therapeutic development against cancer. J Clin Oncol. 2005;23(27):6771-6790.

38. Terai K, Matsuda M. The amino-terminal B-Raf-specific region mediates calcium-dependent homo- and hetero-dimerization of Raf. EMBO J. 2006;25(15):3556-3564.

39. McArthur GA, Chapman PB, Robert C, et al. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014;15(3):323-332.

40. Hauschild A, Grob JJ, Demidov LV, et al. An update on BREAK-3, a phase III, randomized trial: dabrafenib versus dacarbazine in patients with BRAF V600E-positive mutation metastatic melanoma (Abstract 9013). Paper presented at: American Society of Clinical Oncology 2013 meeting; May-June 2013; Chicago, IL.

41. Long GV, Trefzer U, Davies MA, et al. Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(11):1087-1095.

42. Ascierto PA, Minor DR, Ribas A, et. al., Long-term safety and overall survival update for BREAK-2, a phase 2, single-arm, open-label study of dabrafenib in previously treated metastatic melanoma (NCT01153763). J Clin Oncol. 2014;32(15)(suppl): Abstract 9034.

43. Larkin J, Del Vecchio M, Ascierto PA, et al. Vemurafenib in patients with
BRAF(V600) mutated metastatic melanoma: an open-label, multicentre, safety
study. Lancet Oncol. 2014;15(4):436-444.

44. Lacouture ME, Duvic M, Hauschild A, et al. Analysis of dermatologic events in vemurafenib-treated patients with melanoma. Oncologist. 2013;18(3):314-322.

45. Su F, Viros A, Milagre C, et al. RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors. N Engl J Med. 2012;366(3):207-215.

46. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364(26):2507-2516.

47. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380(9839):358-365.

48. Ribas A, Hodi FS, Callahan M, et. al. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med. 2014;368(14):1365-1366.

49. Linette GP, Puzanov I, Callahan MK, et al. Phase 1 study of the BRAF inhibitor dabrafenib (D) with or without the MEK inhibitor trametinib (T) in combination with ipilimumab (Ipi) for V600E/K mutation–positive unresectable or metastatic melanoma (MM). J Clin Oncol. 2014;32(15)(suppl): Abstract 2511.

50. Chan MMK, Haydu LE, Menzies AM, et al. The nature and management of metastatic melanoma after progression on BRAF inhibitors: effects of extended BRAF inhibition. Cancer. 2014;120(20):3142-3153.

51. Carlino MS, Gowrishankar K, Saunders CAB, et al. Antiproliferative effects of continued mitogen-activated protein kinase pathway inhibition following acquired resistance to BRAF and/or MEK inhibition in melanoma. Mol Cancer Ther. 2013;12(7):1332-1342.

52. Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367(2):107-114.

53. Kim KB, Kefford R, Pavlick AC, et. al. Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol. 2013;31(1):482-489.

54. Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol. 2013;14(3):249-256.

55. Sosman JA, Kittaneh M, Lolkema MP, et al. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: early encouraging clinical activity (abstract 9009). Paper presented at: 2014 American Society of Clinical Oncology meeting ; May-June 2014; Chicago, IL.

56. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. 2014;371(20):1877-1888.

57. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30-39.

58. Gogas H, Schadendorf D, Dummer R. Vemurafenib treatment in patients with BRAF-mutated melanoma failing MEK inhibition with trametinib. J Clin Oncol. 2014;32(15)(suppl): Abstract 9061.

59. Larkin J, Ascierto PA, Dréno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371(20):1867-1876.

60. Kefford R, Miller WH, Tan DS, et al. Preliminary results from a phase Ib/II, openlabel, dose-escalation study of the oral BRAF inhibitor LGX818 in combination with the oral MEK1/2 inhibitor MEK162 in BRAF V600-dependent advanced solid tumors (abstract 9019). Paper presented at: 2013 American Society of Clinical Oncology meeting; May-June 2014; Chicago, IL.

61. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct
subtypes of melanoma. J Clin Oncol. 2006;24(26):4340-4346.

62. Jin SA, Chun SM, Choi YD, et al. BRAF mutations and KIT aberrations and their clinicopathological correlation in 202 Korean melanomas. J Invest Dermatol. 2013;133(2):579-582.

63. Guo J, Si L, Kong Y et. al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol. 2011;29(21):2904-2909.

64. Hodi FS, Corless CL, Giobbie-Hurder A, et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol. 2013;31(26):3182-3190.

65. Cho JH, Kim KM, Kwon M, Kim JH, Lee J. Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Invest New Drugs. 2012;30(5): 2008-2014.

66. Lebbe C, Chevret S, Jouary T, et. al. Phase II multicentric uncontrolled national trial assessing the efficacy of nilotinib in the treatment of advanced melanomas with c-KIT mutation or amplification. J Clin Oncol. 2014;32(15)(suppl): Abstract 9032.

67. Perez DG, Suman VJ, Fitch TR, et al. Phase 2 trial of carboplatin, weekly paclitaxel, and biweekly bevacizumab in patients with unresectable stage IV melanoma: a North Central Cancer Treatment Group study, N047A. Cancer. 2009;115(1):119-127.

68. Hainsworth JD, Infante JR, Spigel DR, et al. Bevacizumab and everolimus in the treatment of patients with metastatic melanoma. Cancer. 2010;116(17): 4122-4129.

69. Dronca RS, Allred JB, Perez DG, et. al. Phase II study of temozolomide (TMZ) and everolimus (RAD001) therapy for metastatic melanoma: a North Central Cancer Treatment Group study, N0675. Am J Clin Oncol. 2014;37(4):369-376.

70. Meier FE, Niessner H, Schmitz J, et al. The PI3K inhibitor BKM120 has potent antitumor activity in melanoma brain metastases in vitro and in vivo. J Clin Oncol. 2013;31(15)(suppl): Abstract e20050.

71. Ott PA, Chang J, Madden K, et al. Oblimersen in combination with temozolomide and albumin-bound paclitaxel in patients with advanced melanoma: a phase I trial. Cancer Chemother Pharmacol. 2013;71(1);183-191.

72. Ackerman A, Klein O, McDermott DF, et al. Outcomes of patients with metastatic
melanoma treated with immunotherapy prior to or after BRAF inhibitors. Cancer. 2014;120(11):1695-1701.

73. Ascierto PA, Margolin K. Ipilimumab before BRAF inhibitor treatment may be
more beneficial than vice versa for the majority of patients with advanced melanoma.
Cancer. 2014;120(11):1617-1619.

74. Ascierto PA, Simeone E, Sileni VC, et al. Sequential treatment with ipilimumab and BRAF inhibitors in patients with metastatic melanoma: data from the Italian cohort of the ipilimumab expanded access program. Cancer Invest. 2014;32(4):144-149.

References

1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin.2014;64(1):9-29.

2. Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27(36):6199-6206.

3. Welch HG, Woloshin S, Schwartz LM. Skin biopsy rates and incidence of melanoma:
population based ecological study. BMJ. 2005;331(7515):481.

4. Sosman JA, Moon J, Tuthill RJ, et al. A phase 2 trial of complete resection for stage IV melanoma: results of Southwest Oncology Group Clinical Trial S9430. Cancer. 2011;117(20):4740-4746.

5. Atkins MB. The role of cytotoxic chemotherapeutic agents either alone or in combination with biological response modifiers. In: Kirkwood JK, ed. Molecular Diagnosis, Prevention, & Therapy of Melanoma. New York, NY: Marcel Dekker;1997:219-225.

6. Patel PM, Suciu S, Mortier L, et al. Extended schedule, escalated dose temozolomide versus dacarbazine in stage IV melanoma: final results of a randomised phase III study (EORTC 18032). Eur J Cancer. 2011;47(10):1476-1483.

7. Chapman PB, Einhorn LH, Meyers ML, et al. Phase III multicenter randomized trial of the Dartmouth regimen versus dacarbazine in patients with metastatic melanoma. J Clin Oncol. 1999;17(9):2745-2751.

8. Flaherty KT, Lee SJ, Zhao F, et al. Phase III trial of carboplatin and paclitaxel with
or without sorafenib in metastatic melanoma. J Clin Oncol. 2013;31(3):373-379.

9. Rosenberg SA, Yang JC, Topalian SL, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA. 1994;271(12):907-913.

10. Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993. J Clin Oncol. 1999;17(7):2105-2116.

11. Atkins MB, Kunkel L, Sznol M, Rosenberg SA. High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sci Am. 2000;6(suppl 1):S11-S14.

12. Hoos A, Ibrahim R, Korman A, et al. Development of ipilimumab: contribution to a new paradigm for cancer immunotherapy. Semin Oncol. 2010;37(5):533-546.

13. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711-723.

14. Schadendorf D, Hodi FS, Robert C, et. al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma [published online ahead of print February 9, 2015]. J Clin Oncol. pii:JCO.2014.56.2736.

15. Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691-2697.

16. Hodi FS, Lee S, McDermott DF, et al. Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: a randomized clinical trial. JAMA. 2014;312(17):1744-1753.

17. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443-2454.

18. Ribas A, Hodi FS, Kefford R, et al. Efficacy and safety of the anti-PD-1 monoclonal antibody pembrolizumab (MK-3475) in 411 patients (pts) with melanoma (MEL) (Abstract LBA9000). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

19. Hamid O, Robert C, Ribas A, et al. Randomized comparison of two doses of the anti-PD-1 monoclonal antibody MK-3475 for ipilimumab-refractory (IPI-R) and IPI-naive (IPI-N) melanoma (MEL) (abstract 3000). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

20. Robert C, Ribas A, Wolchok JD, et al. Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. Lancet. 2014; 384(9948):1109-1117.

21. Dummer R, Daud A, Puzanov I, et. al. A randomized controlled comparison of pembrolizumab and chemotherapy in patients with ipilimumab-refractory melanoma. J Transl Med. 2015;13(suppl 1):O5.

22. Topalian SL, Sznol M, McDermott DF, et. al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32(10):1020-1030.

23. Hodi FS, Sznol M, Kluger HM, et al. Long-term survival of ipilimumab-naive patients with advanced melanoma (MEL) treated with nivolumab (anti-PD-1, BMS-936558, ONO-4538) in a phase I trial (abstract 9002). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

24. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med. 2015;372(4):320-330.

25. Weber J, D’Angelo S, Gutzmer R, et al. A phase 3 randomized, open-label study of nivolumab versus investigator’s choice of chemotherapy in patients with advanced melanoma after prior anti-CTLA4 therapy (abstract LBA3). Paper presented at: European Society of Medical Oncology 2014 meeting; September 2014; Madrid, Spain.

26. Atkins MB, Kudchadkar RR, Sznol M, et al. Phase 2, multicenter, safety and efficacy study of pidilizumab in patients with metastatic melanoma (abstract 9001). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

27. Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366(26):2455-2465.

28. Hamid O, Sosman JA, Lawrence DP, et. al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic melanoma (mM). J Clin Oncol. 2013;31(15)(suppl): Abstract 9010.

29. Lutzky J, Antonia SJ, Blake-Haskins A, et. al. A phase 1 study of MEDI4736, an anti–PD-L1 antibody, in patients with advanced solid tumors. J Clin Oncol. 2014;32(15)(suppl): Abstract 3001.

30. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in advanced
melanoma. N Engl J Med. 2013;369(2):122-133.

31. Sznol M, Kluger HM, Callahan MK, et al. Survival, response duration, and activity by BRAF mutation (MT) status of nivolumab (NIVO, anti-PD-1, BMS-936558, ONO-4538) and ipilimumab (IPI) concurrent therapy in advanced melanoma (MEL) (abstract LBA9003). Paper presented at: 2014 American Society of Clinical Oncology (ASCO) meeting; May-June 2014; Chicago, IL.

32. Omholt K, Platz A, Kanter L, Ringborg U, Hansson J. NRAS and BRAF mutations arise early during melanoma pathogenesis and are preserved throughout tumor progression. Clin Cancer Res. 2003;9(17):6483-6488.

33. Wellbrock C, Hurlstone A. BRAF as therapeutic target in melanoma. Biochem Pharmacol. 2010;80(5):561-567.

34. Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol. 2011;29(10):1239-1246.

35. Ball NJ, Yohn JJ, Morelli JG, et al. Ras mutations in human melanoma: a marker of malignant progression. J Invest Dermatol. 1994;102(3):285-290.

36. Platz A, Ringborg U, Brahme EM, Lagerlöf B. Melanoma metastases from patients with hereditary cutaneous malignant melanoma contain a high frequency of N-ras activating mutations. Melanoma Res. 1994;4(3):169-177.

37. Beeram M, Patnaik A, Rowinsky EK. Raf: a strategic target for therapeutic development against cancer. J Clin Oncol. 2005;23(27):6771-6790.

38. Terai K, Matsuda M. The amino-terminal B-Raf-specific region mediates calcium-dependent homo- and hetero-dimerization of Raf. EMBO J. 2006;25(15):3556-3564.

39. McArthur GA, Chapman PB, Robert C, et al. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol. 2014;15(3):323-332.

40. Hauschild A, Grob JJ, Demidov LV, et al. An update on BREAK-3, a phase III, randomized trial: dabrafenib versus dacarbazine in patients with BRAF V600E-positive mutation metastatic melanoma (Abstract 9013). Paper presented at: American Society of Clinical Oncology 2013 meeting; May-June 2013; Chicago, IL.

41. Long GV, Trefzer U, Davies MA, et al. Dabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13(11):1087-1095.

42. Ascierto PA, Minor DR, Ribas A, et. al., Long-term safety and overall survival update for BREAK-2, a phase 2, single-arm, open-label study of dabrafenib in previously treated metastatic melanoma (NCT01153763). J Clin Oncol. 2014;32(15)(suppl): Abstract 9034.

43. Larkin J, Del Vecchio M, Ascierto PA, et al. Vemurafenib in patients with
BRAF(V600) mutated metastatic melanoma: an open-label, multicentre, safety
study. Lancet Oncol. 2014;15(4):436-444.

44. Lacouture ME, Duvic M, Hauschild A, et al. Analysis of dermatologic events in vemurafenib-treated patients with melanoma. Oncologist. 2013;18(3):314-322.

45. Su F, Viros A, Milagre C, et al. RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors. N Engl J Med. 2012;366(3):207-215.

46. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. 2011;364(26):2507-2516.

47. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2012;380(9839):358-365.

48. Ribas A, Hodi FS, Callahan M, et. al. Hepatotoxicity with combination of vemurafenib and ipilimumab. N Engl J Med. 2014;368(14):1365-1366.

49. Linette GP, Puzanov I, Callahan MK, et al. Phase 1 study of the BRAF inhibitor dabrafenib (D) with or without the MEK inhibitor trametinib (T) in combination with ipilimumab (Ipi) for V600E/K mutation–positive unresectable or metastatic melanoma (MM). J Clin Oncol. 2014;32(15)(suppl): Abstract 2511.

50. Chan MMK, Haydu LE, Menzies AM, et al. The nature and management of metastatic melanoma after progression on BRAF inhibitors: effects of extended BRAF inhibition. Cancer. 2014;120(20):3142-3153.

51. Carlino MS, Gowrishankar K, Saunders CAB, et al. Antiproliferative effects of continued mitogen-activated protein kinase pathway inhibition following acquired resistance to BRAF and/or MEK inhibition in melanoma. Mol Cancer Ther. 2013;12(7):1332-1342.

52. Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367(2):107-114.

53. Kim KB, Kefford R, Pavlick AC, et. al. Phase II study of the MEK1/MEK2 inhibitor Trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitor. J Clin Oncol. 2013;31(1):482-489.

54. Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol. 2013;14(3):249-256.

55. Sosman JA, Kittaneh M, Lolkema MP, et al. A phase 1b/2 study of LEE011 in combination with binimetinib (MEK162) in patients with NRAS-mutant melanoma: early encouraging clinical activity (abstract 9009). Paper presented at: 2014 American Society of Clinical Oncology meeting ; May-June 2014; Chicago, IL.

56. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. 2014;371(20):1877-1888.

57. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N Engl J Med. 2015;372(1):30-39.

58. Gogas H, Schadendorf D, Dummer R. Vemurafenib treatment in patients with BRAF-mutated melanoma failing MEK inhibition with trametinib. J Clin Oncol. 2014;32(15)(suppl): Abstract 9061.

59. Larkin J, Ascierto PA, Dréno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371(20):1867-1876.

60. Kefford R, Miller WH, Tan DS, et al. Preliminary results from a phase Ib/II, openlabel, dose-escalation study of the oral BRAF inhibitor LGX818 in combination with the oral MEK1/2 inhibitor MEK162 in BRAF V600-dependent advanced solid tumors (abstract 9019). Paper presented at: 2013 American Society of Clinical Oncology meeting; May-June 2014; Chicago, IL.

61. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct
subtypes of melanoma. J Clin Oncol. 2006;24(26):4340-4346.

62. Jin SA, Chun SM, Choi YD, et al. BRAF mutations and KIT aberrations and their clinicopathological correlation in 202 Korean melanomas. J Invest Dermatol. 2013;133(2):579-582.

63. Guo J, Si L, Kong Y et. al. Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol. 2011;29(21):2904-2909.

64. Hodi FS, Corless CL, Giobbie-Hurder A, et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J Clin Oncol. 2013;31(26):3182-3190.

65. Cho JH, Kim KM, Kwon M, Kim JH, Lee J. Nilotinib in patients with metastatic melanoma harboring KIT gene aberration. Invest New Drugs. 2012;30(5): 2008-2014.

66. Lebbe C, Chevret S, Jouary T, et. al. Phase II multicentric uncontrolled national trial assessing the efficacy of nilotinib in the treatment of advanced melanomas with c-KIT mutation or amplification. J Clin Oncol. 2014;32(15)(suppl): Abstract 9032.

67. Perez DG, Suman VJ, Fitch TR, et al. Phase 2 trial of carboplatin, weekly paclitaxel, and biweekly bevacizumab in patients with unresectable stage IV melanoma: a North Central Cancer Treatment Group study, N047A. Cancer. 2009;115(1):119-127.

68. Hainsworth JD, Infante JR, Spigel DR, et al. Bevacizumab and everolimus in the treatment of patients with metastatic melanoma. Cancer. 2010;116(17): 4122-4129.

69. Dronca RS, Allred JB, Perez DG, et. al. Phase II study of temozolomide (TMZ) and everolimus (RAD001) therapy for metastatic melanoma: a North Central Cancer Treatment Group study, N0675. Am J Clin Oncol. 2014;37(4):369-376.

70. Meier FE, Niessner H, Schmitz J, et al. The PI3K inhibitor BKM120 has potent antitumor activity in melanoma brain metastases in vitro and in vivo. J Clin Oncol. 2013;31(15)(suppl): Abstract e20050.

71. Ott PA, Chang J, Madden K, et al. Oblimersen in combination with temozolomide and albumin-bound paclitaxel in patients with advanced melanoma: a phase I trial. Cancer Chemother Pharmacol. 2013;71(1);183-191.

72. Ackerman A, Klein O, McDermott DF, et al. Outcomes of patients with metastatic
melanoma treated with immunotherapy prior to or after BRAF inhibitors. Cancer. 2014;120(11):1695-1701.

73. Ascierto PA, Margolin K. Ipilimumab before BRAF inhibitor treatment may be
more beneficial than vice versa for the majority of patients with advanced melanoma.
Cancer. 2014;120(11):1617-1619.

74. Ascierto PA, Simeone E, Sileni VC, et al. Sequential treatment with ipilimumab and BRAF inhibitors in patients with metastatic melanoma: data from the Italian cohort of the ipilimumab expanded access program. Cancer Invest. 2014;32(4):144-149.

Issue
Federal Practitioner - 32(4)s
Issue
Federal Practitioner - 32(4)s
Publications
Publications
Topics
Article Type
Sections
Citation Override
Fed Pract. 2015 May;32(suppl 4):57S-65S
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME

Automating venous thromboembolism risk calculation using electronic health record data upon hospital admission: The automated Padua Prediction Score

Article Type
Changed
Fri, 12/14/2018 - 08:25
Display Headline
Automating venous thromboembolism risk calculation using electronic health record data upon hospital admission: The automated Padua Prediction Score

Hospital-acquired venous thromboembolism (VTE) continues to be a critical quality challenge for U.S. hospitals,1 and high-risk patients are often not adequately prophylaxed. Use of VTE prophylaxis (VTEP) varies as widely as 26% to 85% of patients in various studies, as does patient outcomes and care expenditures.2-6 The 9th edition of the American College of Chest Physicians (CHEST) guidelines7 recommend the Padua Prediction Score (PPS) to select individual patients who may be at high risk for venous thromboembolism (VTE) and could benefit from thromboprophylaxis. Use of the manually calculated PPS to select patients for thromboprophylaxis has been shown to help decrease 30-day and 90-day mortality associated with VTE events after hospitalization to medical services.8 However, the PPS requires time-consuming manual calculation by a provider, who may be focused on more immediate aspects of patient care and several other risk scores competing for his attention, potentially decreasing its use.

Other risk scores that use only discrete scalar data, such as vital signs and lab results to predict early recognition of sepsis, have been successfully automated and implemented within electronic health records (EHRs).9-11 Successful automation of scores requiring input of diagnoses, recent medical events, and current clinical status such as the PPS remains difficult.12 Data representing these characteristics are more prone to error, and harder to translate clearly into a single data field than discrete elements like heart rate, potentially impacting validity of the calculated result.13 To improve usage of guideline based VTE risk assessment and decrease physician burden, we developed an algorithm called Automated Padua Prediction Score (APPS) that automatically calculates the PPS using only EHR data available within prior encounters and the first 4 hours of admission, a similar timeframe to when admitting providers would be entering orders. Our goal was to assess if an automatically calculated version of the PPS, a score that depends on criteria more complex than vital signs and labs, would accurately assess risk for hospital-acquired VTE when compared to traditional manual calculation of the Padua Prediction Score by a provider.

METHODS

Site Description and Ethics

The study was conducted at University of California, San Francisco Medical Center, a 790-bed academic hospital; its Institutional Review Board approved the study and collection of data via chart review. Handling of patient information complied with the Health Insurance Portability and Accountability Act of 1996.

 

 

Patient Inclusion

Adult patients admitted to a medical or surgical service between July 1, 2012 and April 1, 2014 were included in the study if they were candidates for VTEP, defined as: length of stay (LOS) greater than 2 days, not on hospice care, not pregnant at admission, no present on admission VTE diagnosis, no known contraindications to prophylaxis (eg, gastrointestinal bleed), and were not receiving therapeutic doses of warfarin, low molecular weight heparins, heparin, or novel anticoagulants prior to admission.

Data Sources

Clinical variables were extracted from the EHR’s enterprise data warehouse (EDW) by SQL Server query (Microsoft, Redmond, Washington) and deposited in a secure database. Chart review was conducted by a trained researcher (Mr. Jacolbia) using the EHR and a standardized protocol. Findings were recorded using REDCap (REDCap Consortium, Vanderbilt University, Nashville, Tennessee). The specific ICD-9, procedure, and lab codes used to determine each criterion of APPS are available in the Appendix.

Creation of the Automated Padua Prediction Score (APPS)

We developed APPS from the original 11 criteria that comprise the Padua Prediction Score: active cancer, previous VTE (excluding superficial vein thrombosis), reduced mobility, known thrombophilic condition, recent (1 month or less) trauma and/or surgery, age 70 years or older, heart and/or respiratory failure, acute myocardial infarction and/or ischemic stroke, acute infection and/or rheumatologic disorder, body mass index (BMI) 30 or higher, and ongoing hormonal treatment.13 APPS has the same scoring methodology as PPS: criteria are weighted from 1 to 3 points and summed with a maximum score of 20, representing highest risk of VTE. To automate the score calculation from data routinely available in the EHR, APPS checks pre-selected structured data fields for specific values within laboratory results, orders, nursing flowsheets and claims. Claims data included all ICD-9 and procedure codes used for billing purposes. If any of the predetermined data elements are found, then the specific criterion is considered positive; otherwise, it is scored as negative. The creators of the PPS were consulted in the generation of these data queries to replicate the original standards for deeming a criterion positive. The automated calculation required no use of natural language processing.

Characterization of Study Population

We recorded patient demographics (age, race, gender, BMI), LOS, and rate of hospital-acquired VTE. These patients were separated into 2 cohorts determined by the VTE prophylaxis they received. The risk profile of patients who received pharmacologic prophylaxis was hypothesized to be inherently different from those who had not. To evaluate APPS within this heterogeneous cohort, patients were divided into 2 major categories: pharmacologic vs. no pharmacologic prophylaxis. If they had a completed order or medication administration record on the institution’s approved formulary for pharmacologic VTEP, they were considered to have received pharmacologic prophylaxis. If they had only a completed order for usage of mechanical prophylaxis (sequential compression devices) or no evidence of any form of VTEP, they were considered to have received no pharmacologic prophylaxis. Patients with evidence of both pharmacologic and mechanical were placed in the pharmacologic prophylaxis group. To ensure that automated designation of prophylaxis group was accurate, we reviewed 40 randomly chosen charts because prior researchers were able to achieve sensitivity and specificity greater than 90% with that sample size.14

The primary outcome of hospital-acquired VTE was defined as an ICD-9 code for VTE (specific codes are found in the Appendix) paired with a “present on admission = no” flag on that encounter’s hospital billing data, abstracted from the EDW. A previous study at this institution used the same methodology and found 212/226 (94%) of patients with a VTE ICD-9 code on claim had evidence of a hospital-acquired VTE event upon chart review.14 Chart review was also completed to ensure that the primary outcome of newly discovered hospital-acquired VTE was differentiated from chronic VTE or history of VTE. Theoretically, ICD-9 codes and other data elements treat chronic VTE, history of VTE, and hospital-acquired VTE as distinct diagnoses, but it was unclear if this was true in our dataset. For 75 randomly selected cases of presumed hospital-acquired VTE, charts were reviewed for evidence that confirmed newly found VTE during that encounter.

Validation of APPS through Comparison to Manual Calculation of the Original PPS

To compare our automated calculation to standard clinical practice, we manually calculated the PPS through chart review within the first 2 days of admission on 300 random patients, a subsample of the entire study cohort. The largest study we could find had manually calculated the PPS of 1,080 hospitalized patients with a mean PPS of 4.86 (standard deviation [SD], 2.26).15 One researcher (Mr. Jacolbia) accessed the EHR with all patient information available to physicians, including admission notes, orders, labs, flowsheets, past medical history, and all prior encounters to calculate and record the PPS. To limit potential score bias, 2 authors (Drs. Elias and Davies) assessed 30 randomly selected charts from the cohort of 300. The standardized chart review protocol mimicked a physician’s approach to determine if a patient met a criterion, such as concluding if he/she had active cancer by examining medication lists for chemotherapy, procedure notes for radiation, and recent diagnoses on problem lists. After the original PPS was manually calculated, APPS was automatically calculated for the same 300 patients. We intended to characterize similarities and differences between APPS and manual calculation prior to investigating APPS’ predictive capacity for the entire study population, because it would not be feasible to manually calculate the PPS for all 30,726 patients.

 

 

Statistical Analysis

For the 75 randomly selected cases of presumed hospital-acquired VTE, the number of cases was chosen by powering our analysis to find a difference in proportion of 20% with 90% power, α = 0.05 (two-sided). We conducted χ2 tests on the entire study cohort to determine if there were significant differences in demographics, LOS, and incidence of hospital-acquired VTE by prophylaxis received. For both the pharmacologic and the no pharmacologic prophylaxis groups, we conducted 2-sample Student t tests to determine significant differences in demographics and LOS between patients who experienced a hospital-acquired VTE and those who did not.

For the comparison of our automated calculation to standard clinical practice, we manually calculated the PPS through chart review within the first 2 days of admission on a subsample of 300 random patients. We powered our analysis to detect a difference in mean PPS from 4.86 to 4.36, enough to alter the point value, with 90% power and α = 0.05 (two-sided) and found 300 patients to be comfortably above the required sample size. We compared APPS and manual calculation in the 300-patient cohort using: 2-sample Student t tests to compare mean scores, χ2 tests to compare the frequency with which criteria were positive, and receiver operating characteristic (ROC) curves to determine capacity to predict a hospital-acquired VTE event. Pearson’s correlation was also completed to assess score agreement between APPS and manual calculation on a per-patient basis. After comparing automated calculation of APPS to manual chart review on the same 300 patients, we used APPS to calculate scores for the entire study cohort (n = 30,726). We calculated the mean of APPS by prophylaxis group and whether hospital-acquired VTE had occurred. We analyzed APPS’ ROC curve statistics by prophylaxis group to determine its overall predictive capacity in our study population. Lastly, we computed the time required to calculate APPS per patient. Statistical analyses were conducted using SPSS Statistics (IBM, Armonk, New York) and Python 2.7 (Python Software Foundation, Beaverton, Oregon); 95% confidence intervals (CI) and (SD) were reported when appropriate.

RESULTS

Among the 30,726 unique patients in our entire cohort (all patients admitted during the time period who met the study criteria), we found 6574 (21.4%) on pharmacologic (with or without mechanical) prophylaxis, 13,511 (44.0%) on mechanical only, and 10,641 (34.6%) on no prophylaxis. χ2 tests found no significant differences in demographics, LOS, or incidence of hospital-acquired VTE between the patients who received mechanical prophylaxis only and those who received no prophylaxis (Table 1). Similarly, there were no differences in these characteristics in patients receiving pharmacologic prophylaxis with or without the addition of mechanical prophylaxis. Designation of prophylaxis group by manual chart review vs. our automated process was found to agree in categorization for 39/40 (97.5%) sampled encounters. When comparing the cohort that received pharmacologic prophylaxis against the cohort that did not, there were significant differences in racial distribution, sex, BMI, and average LOS as shown in Table 1. Those who received pharmacologic prophylaxis were found to be significantly older than those who did not (62.7 years versus 53.2 years, P < 0.001), more likely to be male (50.6% vs, 42.4%, P < 0.001), more likely to have hospital-acquired VTE (2.2% vs. 0.5%, P < 0.001), and to have a shorter LOS (7.1 days vs. 9.8, P < 0.001).

Distribution of Patient Characteristics in Cohort
Table 1

Within the cohort group receiving pharmacologic prophylaxis (n = 6574), hospital-acquired VTE occurred in patients who were significantly younger (58.2 years vs. 62.8 years, P = 0.003) with a greater LOS (23.8 days vs. 6.7, P < 0.001) than those without. Within the group receiving no pharmacologic prophylaxis (n = 24,152), hospital-acquired VTE occurred in patients who were significantly older (57.1 years vs. 53.2 years, P = 0.014) with more than twice the LOS (20.2 days vs. 9.7 days, P < 0.001) compared to those without. Sixty-six of 75 (88%) randomly selected patients in which new VTE was identified by the automated electronic query had this diagnosis confirmed during manual chart review.

As shown in Table 2, automated calculation on a subsample of 300 randomly selected patients using APPS had a mean of 5.5 (SD, 2.9) while manual calculation of the original PPS on the same patients had a mean of 5.1 (SD, 2.6). There was no significant difference in mean between manual calculation and APPS (P = 0.073). There were, however, significant differences in how often individual criteria were considered present. The largest contributors to the difference in scores between APPS and manual calculation were “prior VTE” (positive, 16% vs. 8.3%, respectively) and “reduced mobility” (positive, 74.3% vs. 66%, respectively) as shown in Table 2. In the subsample, there were a total of 6 (2.0%) hospital-acquired VTE events. APPS’ automated calculation had an AUC = 0.79 (CI, 0.63-0.95) that was significant (P = 0.016) with a cutoff value of 5. Chart review’s manual calculation of the PPS had an AUC = 0.76 (CI 0.61-0.91) that was also significant (P = 0.029).

Distribution of Patient Characteristics in Cohort

Comparison of APPS to Manual Calculation of PPS
Table 2


Our entire cohort of 30,726 unique patients admitted during the study period included 260 (0.8%) who experienced hospital-acquired VTEs (Table 3). In patients receiving no pharmacologic prophylaxis, the average APPS was 4.0 (SD, 2.4) for those without VTE and 7.1 (SD, 2.3) for those with VTE. In patients who had received pharmacologic prophylaxis, those without hospital-acquired VTE had an average APPS of 4.9 (SD, 2.6) and those with hospital-acquired VTE averaged 7.7 (SD, 2.6). APPS’ ROC curves for “no pharmacologic prophylaxis” had an AUC = 0.81 (CI, 0.79 – 0.83) that was significant (P < 0.001) with a cutoff value of 5. There was similar performance in the pharmacologic prophylaxis group with an AUC = 0.79 (CI, 0.76 – 0.82) and cutoff value of 5, as shown in the Figure. Over the entire cohort, APPS had a sensitivity of 85.4%, specificity of 53.3%, positive predictive value (PPV) of 1.5%, and a negative predictive value (NPV) of 99.8% when using a cutoff of 5. The average APPS calculation time was 0.03 seconds per encounter. Additional information on individual criteria can be found in Table 3.

ROC curves and predictive characteristics of the APPS
Figure

 

 

DISCUSSION

Automated calculation of APPS using EHR data from prior encounters and the first 4 hours of admission was predictive of in-hospital VTE. APPS performed as well as traditional manual score calculation of the PPS. It was able to do so with no physician input, significantly lessening the burden of calculation and potentially increasing frequency of data-driven VTE risk assessment.

While automated calculation of certain scores is becoming more common, risk calculators that require data beyond vital signs and lab results have lagged,16-19 in part because of uncertainty about 2 issues. The first is whether EHR data accurately represent the current clinical picture. The second is if a machine-interpretable algorithm to determine a clinical status (eg, “active cancer”) would be similar to a doctor’s perception of that same concept. We attempted to better understand these 2 challenges through developing APPS. Concerning accuracy, EHR data correctly represent the clinical scenario: designations of VTEP and hospital-acquired VTE were accurate in approximately 90% of reviewed cases. Regarding the second concern, when comparing APPS to manual calculation, we found significant differences (P < 0.001) in how often 8 of the 11 criteria were positive, yet no significant difference in overall score and similar predictive capacity. Manual calculation appeared more likely to find data in the index encounter or in structured data. For example, “active cancer” may be documented only in a physician’s note, easily accounted for during a physician’s calculation but missed by APPS looking only for structured data. In contrast, automated calculation found historic criteria, such as “prior VTE” or “known thrombophilic condition,” positive more often. If the patient is being admitted for a problem unrelated to blood clots, the physician may have little time or interest to look through hundreds of EHR documents to discover a 2-year-old VTE. As patients’ records become larger and denser, more historic data can become buried and forgotten. While the 2 scores differ on individual criteria, they are similarly predictive and able to bifurcate the at-risk population to those who should and should not receive pharmacologic prophylaxis.

APPS Criteria by Prophylaxis and VTE Occurrence
Table 3

The APPS was found to have near-equal performance in the pharmacologic vs. no pharmacologic prophylaxis cohorts. This finding agrees with a study that found no significant difference in predicting 90-day VTE when looking at 86 risk factors vs. the most significant 4, none of which related to prescribed prophylaxis.18 The original PPS had a reported sensitivity of 94.6%, specificity 62%, PPV 7.5%, and NPV 99.7% in its derivation cohort.13 We matched APPS to the ratio of sensitivity to specificity, using 5 as the cutoff value. APPS performed slightly worse with sensitivity of 85.4%, specificity 53.3%, PPV 1.5%, and NPV 99.8%. This difference may have resulted from the original PPS study’s use of 90-day follow-up to determine VTE occurrence, whereas we looked only until the end of current hospitalization, an average of 9.2 days. Furthermore, the PPS had significantly poorer performance (AUC = 0.62) than that seen in the original derivation cohort in a separate study that manually calculated the score on more than 1000 patients.15

There are important limitations to our study. It was done at a single academic institution using a dataset of VTE-associated, validated research that was well-known to the researchers.20 Another major limitation is the dependence of the algorithm on data available within the first 4 hours of admission and earlier; thus, previous encounters may frequently play an important role. Patients presenting to our health system for the first time would have significantly fewer data available at the time of calculation. Additionally, our data could not reliably tell us the total doses of pharmacologic prophylaxis that a patient received. While most patients will maintain a consistent VTEP regimen once initiated in the hospital, 2 patients with the same LOS may have received differing amounts of pharmacologic prophylaxis. This research study did not assess how much time automatic calculation of VTE risk might save providers, because we did not record the time for each manual abstraction; however, from discussion with the main abstracter, chart review and manual calculation for this study took from 2 to 14 minutes per patient, depending on the number of previous interactions with the health system. Finally, although we chose data elements that are likely to exist at most institutions using an EHR, many institutions’ EHRs do not have EDW capabilities nor programmers who can assist with an automated risk score.

The EHR interventions to assist providers in determining appropriate VTEP have been able to increase rates of VTEP and decrease VTE-associated mortality.16,21 In addition to automating the calculation of guideline-adherent risk scores, there is a need for wider adoption for clinical decision support for VTE. For this reason, we chose only structured data fields from some of the most common elements within our EHR’s data warehouse to derive APPS (Appendix 1). Our study supports the idea that automated calculation of scores requiring input of more complex data such as diagnoses, recent medical events, and current clinical status remains predictive of hospital-acquired VTE risk. Because it is calculated automatically in the background while the clinician completes his or her assessment, the APPS holds the potential to significantly reduce the burden on providers while making guideline-adherent risk assessment more readily accessible. Further research is required to determine the exact amount of time automatic calculation saves, and, more important, if the relatively high predictive capacity we observed using APPS would be reproducible across institutions and could reduce incidence of hospital-acquired VTE.

 

 

Disclosures

Dr. Auerbach was supported by NHLBI K24HL098372 during the period of this study. Dr. Khanna, who is an implementation scientist at the University of California San Francisco Center for Digital Health Innovation, is the principal inventor of CareWeb, and may benefit financially from its commercialization. The other authors report no financial conflicts of interest.

Files
References

1. Galson S. The Surgeon General’s call to action to prevent deep vein thrombosis and pulmonary embolism. 2008. https://www.ncbi.nlm.nih.gov/books/NBK44178/. Accessed February 11, 2016. PubMed
2. Borch KH, Nyegaard C, Hansen JB, et al. Joint effects of obesity and body height on the risk of venous thromboembolism: the Tromsø study. Arterioscler Thromb Vasc Biol. 2011;31(6):1439-44. PubMed
3. Braekkan SK, Borch KH, Mathiesen EB, Njølstad I, Wilsgaard T, Hansen JB.. Body height and risk of venous thromboembolism: the Tromsø Study. Am J Epidemiol. 2010;171(10):1109-1115. PubMed
4. Bounameaux H, Rosendaal FR. Venous thromboembolism: why does ethnicity matter? Circulation. 2011;123(200:2189-2191. PubMed
5. Spyropoulos AC, Anderson FA Jr, Fitzgerald G, et al; IMPROVE Investigators. Predictive and associative models to identify hospitalized medical patients at risk for VTE. Chest. 2011;140(3):706-714. PubMed
6. Rothberg MB, Lindenauer PK, Lahti M, Pekow PS, Selker HP. Risk factor model to predict venous thromboembolism in hospitalized medical patients. J Hosp Med. 2011;6(4):202-209. PubMed
7. Perioperative Management of Antithrombotic Therapy: Prevention of VTE in Nonsurgical Patients: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(6):1645.
8. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521-526. PubMed
9. Alvarez CA, Clark CA, Zhang S, et al. Predicting out of intensive care unit cardiopulmonary arrest or death using electronic medical record data. BMC Med Inform Decis Mak. 2013;13:28. PubMed
10. Escobar GJ, LaGuardia JC, Turk BJ, Ragins A, Kipnis P, Draper D. Early detection of impending physiologic deterioration among patients who are not in intensive care: development of predictive models using data from an automated electronic medical record. J Hosp Med. 2012;7(5):388-395. PubMed
11. Umscheid CA, Hanish A, Chittams J, Weiner MG, Hecht TE. Effectiveness of a novel and scalable clinical decision support intervention to improve venous thromboembolism prophylaxis: a quasi-experimental study. BMC Med Inform Decis Mak. 2012;12:92. PubMed
12. Tepas JJ 3rd, Rimar JM, Hsiao AL, Nussbaum MS. Automated analysis of electronic medical record data reflects the pathophysiology of operative complications. Surgery. 2013;154(4):918-924. PubMed
13. Barbar S, Noventa F, Rossetto V, et al. A risk assessment model for the identification of hospitalized medical patients at risk for venous thromboembolism: the Padua Prediction Score. J Thromb Haemost. 2010; 8(11):2450-2457. PubMed
14. Khanna R, Maynard G, Sadeghi B, et al. Incidence of hospital-acquired venous thromboembolic codes in medical patients hospitalized in academic medical centers. J Hosp Med. 2014; 9(4):221-225. PubMed
15. Vardi M, Ghanem-Zoubi NO, Zidan R, Yurin V, Bitterman H. Venous thromboembolism and the utility of the Padua Prediction Score in patients with sepsis admitted to internal medicine departments. J Thromb Haemost. 2013;11(3):467-473. PubMed
16. Samama MM, Dahl OE, Mismetti P, et al. An electronic tool for venous thromboembolism prevention in medical and surgical patients. Haematologica. 2006;91(1):64-70. PubMed
17. Mann DM, Kannry JL, Edonyabo D, et al. Rationale, design, and implementation protocol of an electronic health record integrated clinical prediction rule (iCPR) randomized trial in primary care. Implement Sci. 2011;6:109. PubMed
18. Woller SC, Stevens SM, Jones JP, et al. Derivation and validation of a simple model to identify venous thromboembolism risk in medical patients. Am J Med. 2011;124(10):947-954. PubMed
19. Huang W, Anderson FA, Spencer FA, Gallus A, Goldberg RJ. Risk-assessment models for predicting venous thromboembolism among hospitalized non-surgical patients: a systematic review. J Thromb Thrombolysis. 2013;35(1):67-80. PubMed
20. Khanna RR, Kim SB, Jenkins I, et al. Predictive value of the present-on-admission indicator for hospital-acquired venous thromboembolism. Med Care. 2015;53(4):e31-e36. PubMed
21. Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous thromboembolism a
mong hospitalized patients. N Engl J Med. 2005;352(10):969-977. PubMed

Article PDF
Issue
Journal of Hospital Medicine 12(4)
Topics
Page Number
231-237
Sections
Files
Files
Article PDF
Article PDF

Hospital-acquired venous thromboembolism (VTE) continues to be a critical quality challenge for U.S. hospitals,1 and high-risk patients are often not adequately prophylaxed. Use of VTE prophylaxis (VTEP) varies as widely as 26% to 85% of patients in various studies, as does patient outcomes and care expenditures.2-6 The 9th edition of the American College of Chest Physicians (CHEST) guidelines7 recommend the Padua Prediction Score (PPS) to select individual patients who may be at high risk for venous thromboembolism (VTE) and could benefit from thromboprophylaxis. Use of the manually calculated PPS to select patients for thromboprophylaxis has been shown to help decrease 30-day and 90-day mortality associated with VTE events after hospitalization to medical services.8 However, the PPS requires time-consuming manual calculation by a provider, who may be focused on more immediate aspects of patient care and several other risk scores competing for his attention, potentially decreasing its use.

Other risk scores that use only discrete scalar data, such as vital signs and lab results to predict early recognition of sepsis, have been successfully automated and implemented within electronic health records (EHRs).9-11 Successful automation of scores requiring input of diagnoses, recent medical events, and current clinical status such as the PPS remains difficult.12 Data representing these characteristics are more prone to error, and harder to translate clearly into a single data field than discrete elements like heart rate, potentially impacting validity of the calculated result.13 To improve usage of guideline based VTE risk assessment and decrease physician burden, we developed an algorithm called Automated Padua Prediction Score (APPS) that automatically calculates the PPS using only EHR data available within prior encounters and the first 4 hours of admission, a similar timeframe to when admitting providers would be entering orders. Our goal was to assess if an automatically calculated version of the PPS, a score that depends on criteria more complex than vital signs and labs, would accurately assess risk for hospital-acquired VTE when compared to traditional manual calculation of the Padua Prediction Score by a provider.

METHODS

Site Description and Ethics

The study was conducted at University of California, San Francisco Medical Center, a 790-bed academic hospital; its Institutional Review Board approved the study and collection of data via chart review. Handling of patient information complied with the Health Insurance Portability and Accountability Act of 1996.

 

 

Patient Inclusion

Adult patients admitted to a medical or surgical service between July 1, 2012 and April 1, 2014 were included in the study if they were candidates for VTEP, defined as: length of stay (LOS) greater than 2 days, not on hospice care, not pregnant at admission, no present on admission VTE diagnosis, no known contraindications to prophylaxis (eg, gastrointestinal bleed), and were not receiving therapeutic doses of warfarin, low molecular weight heparins, heparin, or novel anticoagulants prior to admission.

Data Sources

Clinical variables were extracted from the EHR’s enterprise data warehouse (EDW) by SQL Server query (Microsoft, Redmond, Washington) and deposited in a secure database. Chart review was conducted by a trained researcher (Mr. Jacolbia) using the EHR and a standardized protocol. Findings were recorded using REDCap (REDCap Consortium, Vanderbilt University, Nashville, Tennessee). The specific ICD-9, procedure, and lab codes used to determine each criterion of APPS are available in the Appendix.

Creation of the Automated Padua Prediction Score (APPS)

We developed APPS from the original 11 criteria that comprise the Padua Prediction Score: active cancer, previous VTE (excluding superficial vein thrombosis), reduced mobility, known thrombophilic condition, recent (1 month or less) trauma and/or surgery, age 70 years or older, heart and/or respiratory failure, acute myocardial infarction and/or ischemic stroke, acute infection and/or rheumatologic disorder, body mass index (BMI) 30 or higher, and ongoing hormonal treatment.13 APPS has the same scoring methodology as PPS: criteria are weighted from 1 to 3 points and summed with a maximum score of 20, representing highest risk of VTE. To automate the score calculation from data routinely available in the EHR, APPS checks pre-selected structured data fields for specific values within laboratory results, orders, nursing flowsheets and claims. Claims data included all ICD-9 and procedure codes used for billing purposes. If any of the predetermined data elements are found, then the specific criterion is considered positive; otherwise, it is scored as negative. The creators of the PPS were consulted in the generation of these data queries to replicate the original standards for deeming a criterion positive. The automated calculation required no use of natural language processing.

Characterization of Study Population

We recorded patient demographics (age, race, gender, BMI), LOS, and rate of hospital-acquired VTE. These patients were separated into 2 cohorts determined by the VTE prophylaxis they received. The risk profile of patients who received pharmacologic prophylaxis was hypothesized to be inherently different from those who had not. To evaluate APPS within this heterogeneous cohort, patients were divided into 2 major categories: pharmacologic vs. no pharmacologic prophylaxis. If they had a completed order or medication administration record on the institution’s approved formulary for pharmacologic VTEP, they were considered to have received pharmacologic prophylaxis. If they had only a completed order for usage of mechanical prophylaxis (sequential compression devices) or no evidence of any form of VTEP, they were considered to have received no pharmacologic prophylaxis. Patients with evidence of both pharmacologic and mechanical were placed in the pharmacologic prophylaxis group. To ensure that automated designation of prophylaxis group was accurate, we reviewed 40 randomly chosen charts because prior researchers were able to achieve sensitivity and specificity greater than 90% with that sample size.14

The primary outcome of hospital-acquired VTE was defined as an ICD-9 code for VTE (specific codes are found in the Appendix) paired with a “present on admission = no” flag on that encounter’s hospital billing data, abstracted from the EDW. A previous study at this institution used the same methodology and found 212/226 (94%) of patients with a VTE ICD-9 code on claim had evidence of a hospital-acquired VTE event upon chart review.14 Chart review was also completed to ensure that the primary outcome of newly discovered hospital-acquired VTE was differentiated from chronic VTE or history of VTE. Theoretically, ICD-9 codes and other data elements treat chronic VTE, history of VTE, and hospital-acquired VTE as distinct diagnoses, but it was unclear if this was true in our dataset. For 75 randomly selected cases of presumed hospital-acquired VTE, charts were reviewed for evidence that confirmed newly found VTE during that encounter.

Validation of APPS through Comparison to Manual Calculation of the Original PPS

To compare our automated calculation to standard clinical practice, we manually calculated the PPS through chart review within the first 2 days of admission on 300 random patients, a subsample of the entire study cohort. The largest study we could find had manually calculated the PPS of 1,080 hospitalized patients with a mean PPS of 4.86 (standard deviation [SD], 2.26).15 One researcher (Mr. Jacolbia) accessed the EHR with all patient information available to physicians, including admission notes, orders, labs, flowsheets, past medical history, and all prior encounters to calculate and record the PPS. To limit potential score bias, 2 authors (Drs. Elias and Davies) assessed 30 randomly selected charts from the cohort of 300. The standardized chart review protocol mimicked a physician’s approach to determine if a patient met a criterion, such as concluding if he/she had active cancer by examining medication lists for chemotherapy, procedure notes for radiation, and recent diagnoses on problem lists. After the original PPS was manually calculated, APPS was automatically calculated for the same 300 patients. We intended to characterize similarities and differences between APPS and manual calculation prior to investigating APPS’ predictive capacity for the entire study population, because it would not be feasible to manually calculate the PPS for all 30,726 patients.

 

 

Statistical Analysis

For the 75 randomly selected cases of presumed hospital-acquired VTE, the number of cases was chosen by powering our analysis to find a difference in proportion of 20% with 90% power, α = 0.05 (two-sided). We conducted χ2 tests on the entire study cohort to determine if there were significant differences in demographics, LOS, and incidence of hospital-acquired VTE by prophylaxis received. For both the pharmacologic and the no pharmacologic prophylaxis groups, we conducted 2-sample Student t tests to determine significant differences in demographics and LOS between patients who experienced a hospital-acquired VTE and those who did not.

For the comparison of our automated calculation to standard clinical practice, we manually calculated the PPS through chart review within the first 2 days of admission on a subsample of 300 random patients. We powered our analysis to detect a difference in mean PPS from 4.86 to 4.36, enough to alter the point value, with 90% power and α = 0.05 (two-sided) and found 300 patients to be comfortably above the required sample size. We compared APPS and manual calculation in the 300-patient cohort using: 2-sample Student t tests to compare mean scores, χ2 tests to compare the frequency with which criteria were positive, and receiver operating characteristic (ROC) curves to determine capacity to predict a hospital-acquired VTE event. Pearson’s correlation was also completed to assess score agreement between APPS and manual calculation on a per-patient basis. After comparing automated calculation of APPS to manual chart review on the same 300 patients, we used APPS to calculate scores for the entire study cohort (n = 30,726). We calculated the mean of APPS by prophylaxis group and whether hospital-acquired VTE had occurred. We analyzed APPS’ ROC curve statistics by prophylaxis group to determine its overall predictive capacity in our study population. Lastly, we computed the time required to calculate APPS per patient. Statistical analyses were conducted using SPSS Statistics (IBM, Armonk, New York) and Python 2.7 (Python Software Foundation, Beaverton, Oregon); 95% confidence intervals (CI) and (SD) were reported when appropriate.

RESULTS

Among the 30,726 unique patients in our entire cohort (all patients admitted during the time period who met the study criteria), we found 6574 (21.4%) on pharmacologic (with or without mechanical) prophylaxis, 13,511 (44.0%) on mechanical only, and 10,641 (34.6%) on no prophylaxis. χ2 tests found no significant differences in demographics, LOS, or incidence of hospital-acquired VTE between the patients who received mechanical prophylaxis only and those who received no prophylaxis (Table 1). Similarly, there were no differences in these characteristics in patients receiving pharmacologic prophylaxis with or without the addition of mechanical prophylaxis. Designation of prophylaxis group by manual chart review vs. our automated process was found to agree in categorization for 39/40 (97.5%) sampled encounters. When comparing the cohort that received pharmacologic prophylaxis against the cohort that did not, there were significant differences in racial distribution, sex, BMI, and average LOS as shown in Table 1. Those who received pharmacologic prophylaxis were found to be significantly older than those who did not (62.7 years versus 53.2 years, P < 0.001), more likely to be male (50.6% vs, 42.4%, P < 0.001), more likely to have hospital-acquired VTE (2.2% vs. 0.5%, P < 0.001), and to have a shorter LOS (7.1 days vs. 9.8, P < 0.001).

Distribution of Patient Characteristics in Cohort
Table 1

Within the cohort group receiving pharmacologic prophylaxis (n = 6574), hospital-acquired VTE occurred in patients who were significantly younger (58.2 years vs. 62.8 years, P = 0.003) with a greater LOS (23.8 days vs. 6.7, P < 0.001) than those without. Within the group receiving no pharmacologic prophylaxis (n = 24,152), hospital-acquired VTE occurred in patients who were significantly older (57.1 years vs. 53.2 years, P = 0.014) with more than twice the LOS (20.2 days vs. 9.7 days, P < 0.001) compared to those without. Sixty-six of 75 (88%) randomly selected patients in which new VTE was identified by the automated electronic query had this diagnosis confirmed during manual chart review.

As shown in Table 2, automated calculation on a subsample of 300 randomly selected patients using APPS had a mean of 5.5 (SD, 2.9) while manual calculation of the original PPS on the same patients had a mean of 5.1 (SD, 2.6). There was no significant difference in mean between manual calculation and APPS (P = 0.073). There were, however, significant differences in how often individual criteria were considered present. The largest contributors to the difference in scores between APPS and manual calculation were “prior VTE” (positive, 16% vs. 8.3%, respectively) and “reduced mobility” (positive, 74.3% vs. 66%, respectively) as shown in Table 2. In the subsample, there were a total of 6 (2.0%) hospital-acquired VTE events. APPS’ automated calculation had an AUC = 0.79 (CI, 0.63-0.95) that was significant (P = 0.016) with a cutoff value of 5. Chart review’s manual calculation of the PPS had an AUC = 0.76 (CI 0.61-0.91) that was also significant (P = 0.029).

Distribution of Patient Characteristics in Cohort

Comparison of APPS to Manual Calculation of PPS
Table 2


Our entire cohort of 30,726 unique patients admitted during the study period included 260 (0.8%) who experienced hospital-acquired VTEs (Table 3). In patients receiving no pharmacologic prophylaxis, the average APPS was 4.0 (SD, 2.4) for those without VTE and 7.1 (SD, 2.3) for those with VTE. In patients who had received pharmacologic prophylaxis, those without hospital-acquired VTE had an average APPS of 4.9 (SD, 2.6) and those with hospital-acquired VTE averaged 7.7 (SD, 2.6). APPS’ ROC curves for “no pharmacologic prophylaxis” had an AUC = 0.81 (CI, 0.79 – 0.83) that was significant (P < 0.001) with a cutoff value of 5. There was similar performance in the pharmacologic prophylaxis group with an AUC = 0.79 (CI, 0.76 – 0.82) and cutoff value of 5, as shown in the Figure. Over the entire cohort, APPS had a sensitivity of 85.4%, specificity of 53.3%, positive predictive value (PPV) of 1.5%, and a negative predictive value (NPV) of 99.8% when using a cutoff of 5. The average APPS calculation time was 0.03 seconds per encounter. Additional information on individual criteria can be found in Table 3.

ROC curves and predictive characteristics of the APPS
Figure

 

 

DISCUSSION

Automated calculation of APPS using EHR data from prior encounters and the first 4 hours of admission was predictive of in-hospital VTE. APPS performed as well as traditional manual score calculation of the PPS. It was able to do so with no physician input, significantly lessening the burden of calculation and potentially increasing frequency of data-driven VTE risk assessment.

While automated calculation of certain scores is becoming more common, risk calculators that require data beyond vital signs and lab results have lagged,16-19 in part because of uncertainty about 2 issues. The first is whether EHR data accurately represent the current clinical picture. The second is if a machine-interpretable algorithm to determine a clinical status (eg, “active cancer”) would be similar to a doctor’s perception of that same concept. We attempted to better understand these 2 challenges through developing APPS. Concerning accuracy, EHR data correctly represent the clinical scenario: designations of VTEP and hospital-acquired VTE were accurate in approximately 90% of reviewed cases. Regarding the second concern, when comparing APPS to manual calculation, we found significant differences (P < 0.001) in how often 8 of the 11 criteria were positive, yet no significant difference in overall score and similar predictive capacity. Manual calculation appeared more likely to find data in the index encounter or in structured data. For example, “active cancer” may be documented only in a physician’s note, easily accounted for during a physician’s calculation but missed by APPS looking only for structured data. In contrast, automated calculation found historic criteria, such as “prior VTE” or “known thrombophilic condition,” positive more often. If the patient is being admitted for a problem unrelated to blood clots, the physician may have little time or interest to look through hundreds of EHR documents to discover a 2-year-old VTE. As patients’ records become larger and denser, more historic data can become buried and forgotten. While the 2 scores differ on individual criteria, they are similarly predictive and able to bifurcate the at-risk population to those who should and should not receive pharmacologic prophylaxis.

APPS Criteria by Prophylaxis and VTE Occurrence
Table 3

The APPS was found to have near-equal performance in the pharmacologic vs. no pharmacologic prophylaxis cohorts. This finding agrees with a study that found no significant difference in predicting 90-day VTE when looking at 86 risk factors vs. the most significant 4, none of which related to prescribed prophylaxis.18 The original PPS had a reported sensitivity of 94.6%, specificity 62%, PPV 7.5%, and NPV 99.7% in its derivation cohort.13 We matched APPS to the ratio of sensitivity to specificity, using 5 as the cutoff value. APPS performed slightly worse with sensitivity of 85.4%, specificity 53.3%, PPV 1.5%, and NPV 99.8%. This difference may have resulted from the original PPS study’s use of 90-day follow-up to determine VTE occurrence, whereas we looked only until the end of current hospitalization, an average of 9.2 days. Furthermore, the PPS had significantly poorer performance (AUC = 0.62) than that seen in the original derivation cohort in a separate study that manually calculated the score on more than 1000 patients.15

There are important limitations to our study. It was done at a single academic institution using a dataset of VTE-associated, validated research that was well-known to the researchers.20 Another major limitation is the dependence of the algorithm on data available within the first 4 hours of admission and earlier; thus, previous encounters may frequently play an important role. Patients presenting to our health system for the first time would have significantly fewer data available at the time of calculation. Additionally, our data could not reliably tell us the total doses of pharmacologic prophylaxis that a patient received. While most patients will maintain a consistent VTEP regimen once initiated in the hospital, 2 patients with the same LOS may have received differing amounts of pharmacologic prophylaxis. This research study did not assess how much time automatic calculation of VTE risk might save providers, because we did not record the time for each manual abstraction; however, from discussion with the main abstracter, chart review and manual calculation for this study took from 2 to 14 minutes per patient, depending on the number of previous interactions with the health system. Finally, although we chose data elements that are likely to exist at most institutions using an EHR, many institutions’ EHRs do not have EDW capabilities nor programmers who can assist with an automated risk score.

The EHR interventions to assist providers in determining appropriate VTEP have been able to increase rates of VTEP and decrease VTE-associated mortality.16,21 In addition to automating the calculation of guideline-adherent risk scores, there is a need for wider adoption for clinical decision support for VTE. For this reason, we chose only structured data fields from some of the most common elements within our EHR’s data warehouse to derive APPS (Appendix 1). Our study supports the idea that automated calculation of scores requiring input of more complex data such as diagnoses, recent medical events, and current clinical status remains predictive of hospital-acquired VTE risk. Because it is calculated automatically in the background while the clinician completes his or her assessment, the APPS holds the potential to significantly reduce the burden on providers while making guideline-adherent risk assessment more readily accessible. Further research is required to determine the exact amount of time automatic calculation saves, and, more important, if the relatively high predictive capacity we observed using APPS would be reproducible across institutions and could reduce incidence of hospital-acquired VTE.

 

 

Disclosures

Dr. Auerbach was supported by NHLBI K24HL098372 during the period of this study. Dr. Khanna, who is an implementation scientist at the University of California San Francisco Center for Digital Health Innovation, is the principal inventor of CareWeb, and may benefit financially from its commercialization. The other authors report no financial conflicts of interest.

Hospital-acquired venous thromboembolism (VTE) continues to be a critical quality challenge for U.S. hospitals,1 and high-risk patients are often not adequately prophylaxed. Use of VTE prophylaxis (VTEP) varies as widely as 26% to 85% of patients in various studies, as does patient outcomes and care expenditures.2-6 The 9th edition of the American College of Chest Physicians (CHEST) guidelines7 recommend the Padua Prediction Score (PPS) to select individual patients who may be at high risk for venous thromboembolism (VTE) and could benefit from thromboprophylaxis. Use of the manually calculated PPS to select patients for thromboprophylaxis has been shown to help decrease 30-day and 90-day mortality associated with VTE events after hospitalization to medical services.8 However, the PPS requires time-consuming manual calculation by a provider, who may be focused on more immediate aspects of patient care and several other risk scores competing for his attention, potentially decreasing its use.

Other risk scores that use only discrete scalar data, such as vital signs and lab results to predict early recognition of sepsis, have been successfully automated and implemented within electronic health records (EHRs).9-11 Successful automation of scores requiring input of diagnoses, recent medical events, and current clinical status such as the PPS remains difficult.12 Data representing these characteristics are more prone to error, and harder to translate clearly into a single data field than discrete elements like heart rate, potentially impacting validity of the calculated result.13 To improve usage of guideline based VTE risk assessment and decrease physician burden, we developed an algorithm called Automated Padua Prediction Score (APPS) that automatically calculates the PPS using only EHR data available within prior encounters and the first 4 hours of admission, a similar timeframe to when admitting providers would be entering orders. Our goal was to assess if an automatically calculated version of the PPS, a score that depends on criteria more complex than vital signs and labs, would accurately assess risk for hospital-acquired VTE when compared to traditional manual calculation of the Padua Prediction Score by a provider.

METHODS

Site Description and Ethics

The study was conducted at University of California, San Francisco Medical Center, a 790-bed academic hospital; its Institutional Review Board approved the study and collection of data via chart review. Handling of patient information complied with the Health Insurance Portability and Accountability Act of 1996.

 

 

Patient Inclusion

Adult patients admitted to a medical or surgical service between July 1, 2012 and April 1, 2014 were included in the study if they were candidates for VTEP, defined as: length of stay (LOS) greater than 2 days, not on hospice care, not pregnant at admission, no present on admission VTE diagnosis, no known contraindications to prophylaxis (eg, gastrointestinal bleed), and were not receiving therapeutic doses of warfarin, low molecular weight heparins, heparin, or novel anticoagulants prior to admission.

Data Sources

Clinical variables were extracted from the EHR’s enterprise data warehouse (EDW) by SQL Server query (Microsoft, Redmond, Washington) and deposited in a secure database. Chart review was conducted by a trained researcher (Mr. Jacolbia) using the EHR and a standardized protocol. Findings were recorded using REDCap (REDCap Consortium, Vanderbilt University, Nashville, Tennessee). The specific ICD-9, procedure, and lab codes used to determine each criterion of APPS are available in the Appendix.

Creation of the Automated Padua Prediction Score (APPS)

We developed APPS from the original 11 criteria that comprise the Padua Prediction Score: active cancer, previous VTE (excluding superficial vein thrombosis), reduced mobility, known thrombophilic condition, recent (1 month or less) trauma and/or surgery, age 70 years or older, heart and/or respiratory failure, acute myocardial infarction and/or ischemic stroke, acute infection and/or rheumatologic disorder, body mass index (BMI) 30 or higher, and ongoing hormonal treatment.13 APPS has the same scoring methodology as PPS: criteria are weighted from 1 to 3 points and summed with a maximum score of 20, representing highest risk of VTE. To automate the score calculation from data routinely available in the EHR, APPS checks pre-selected structured data fields for specific values within laboratory results, orders, nursing flowsheets and claims. Claims data included all ICD-9 and procedure codes used for billing purposes. If any of the predetermined data elements are found, then the specific criterion is considered positive; otherwise, it is scored as negative. The creators of the PPS were consulted in the generation of these data queries to replicate the original standards for deeming a criterion positive. The automated calculation required no use of natural language processing.

Characterization of Study Population

We recorded patient demographics (age, race, gender, BMI), LOS, and rate of hospital-acquired VTE. These patients were separated into 2 cohorts determined by the VTE prophylaxis they received. The risk profile of patients who received pharmacologic prophylaxis was hypothesized to be inherently different from those who had not. To evaluate APPS within this heterogeneous cohort, patients were divided into 2 major categories: pharmacologic vs. no pharmacologic prophylaxis. If they had a completed order or medication administration record on the institution’s approved formulary for pharmacologic VTEP, they were considered to have received pharmacologic prophylaxis. If they had only a completed order for usage of mechanical prophylaxis (sequential compression devices) or no evidence of any form of VTEP, they were considered to have received no pharmacologic prophylaxis. Patients with evidence of both pharmacologic and mechanical were placed in the pharmacologic prophylaxis group. To ensure that automated designation of prophylaxis group was accurate, we reviewed 40 randomly chosen charts because prior researchers were able to achieve sensitivity and specificity greater than 90% with that sample size.14

The primary outcome of hospital-acquired VTE was defined as an ICD-9 code for VTE (specific codes are found in the Appendix) paired with a “present on admission = no” flag on that encounter’s hospital billing data, abstracted from the EDW. A previous study at this institution used the same methodology and found 212/226 (94%) of patients with a VTE ICD-9 code on claim had evidence of a hospital-acquired VTE event upon chart review.14 Chart review was also completed to ensure that the primary outcome of newly discovered hospital-acquired VTE was differentiated from chronic VTE or history of VTE. Theoretically, ICD-9 codes and other data elements treat chronic VTE, history of VTE, and hospital-acquired VTE as distinct diagnoses, but it was unclear if this was true in our dataset. For 75 randomly selected cases of presumed hospital-acquired VTE, charts were reviewed for evidence that confirmed newly found VTE during that encounter.

Validation of APPS through Comparison to Manual Calculation of the Original PPS

To compare our automated calculation to standard clinical practice, we manually calculated the PPS through chart review within the first 2 days of admission on 300 random patients, a subsample of the entire study cohort. The largest study we could find had manually calculated the PPS of 1,080 hospitalized patients with a mean PPS of 4.86 (standard deviation [SD], 2.26).15 One researcher (Mr. Jacolbia) accessed the EHR with all patient information available to physicians, including admission notes, orders, labs, flowsheets, past medical history, and all prior encounters to calculate and record the PPS. To limit potential score bias, 2 authors (Drs. Elias and Davies) assessed 30 randomly selected charts from the cohort of 300. The standardized chart review protocol mimicked a physician’s approach to determine if a patient met a criterion, such as concluding if he/she had active cancer by examining medication lists for chemotherapy, procedure notes for radiation, and recent diagnoses on problem lists. After the original PPS was manually calculated, APPS was automatically calculated for the same 300 patients. We intended to characterize similarities and differences between APPS and manual calculation prior to investigating APPS’ predictive capacity for the entire study population, because it would not be feasible to manually calculate the PPS for all 30,726 patients.

 

 

Statistical Analysis

For the 75 randomly selected cases of presumed hospital-acquired VTE, the number of cases was chosen by powering our analysis to find a difference in proportion of 20% with 90% power, α = 0.05 (two-sided). We conducted χ2 tests on the entire study cohort to determine if there were significant differences in demographics, LOS, and incidence of hospital-acquired VTE by prophylaxis received. For both the pharmacologic and the no pharmacologic prophylaxis groups, we conducted 2-sample Student t tests to determine significant differences in demographics and LOS between patients who experienced a hospital-acquired VTE and those who did not.

For the comparison of our automated calculation to standard clinical practice, we manually calculated the PPS through chart review within the first 2 days of admission on a subsample of 300 random patients. We powered our analysis to detect a difference in mean PPS from 4.86 to 4.36, enough to alter the point value, with 90% power and α = 0.05 (two-sided) and found 300 patients to be comfortably above the required sample size. We compared APPS and manual calculation in the 300-patient cohort using: 2-sample Student t tests to compare mean scores, χ2 tests to compare the frequency with which criteria were positive, and receiver operating characteristic (ROC) curves to determine capacity to predict a hospital-acquired VTE event. Pearson’s correlation was also completed to assess score agreement between APPS and manual calculation on a per-patient basis. After comparing automated calculation of APPS to manual chart review on the same 300 patients, we used APPS to calculate scores for the entire study cohort (n = 30,726). We calculated the mean of APPS by prophylaxis group and whether hospital-acquired VTE had occurred. We analyzed APPS’ ROC curve statistics by prophylaxis group to determine its overall predictive capacity in our study population. Lastly, we computed the time required to calculate APPS per patient. Statistical analyses were conducted using SPSS Statistics (IBM, Armonk, New York) and Python 2.7 (Python Software Foundation, Beaverton, Oregon); 95% confidence intervals (CI) and (SD) were reported when appropriate.

RESULTS

Among the 30,726 unique patients in our entire cohort (all patients admitted during the time period who met the study criteria), we found 6574 (21.4%) on pharmacologic (with or without mechanical) prophylaxis, 13,511 (44.0%) on mechanical only, and 10,641 (34.6%) on no prophylaxis. χ2 tests found no significant differences in demographics, LOS, or incidence of hospital-acquired VTE between the patients who received mechanical prophylaxis only and those who received no prophylaxis (Table 1). Similarly, there were no differences in these characteristics in patients receiving pharmacologic prophylaxis with or without the addition of mechanical prophylaxis. Designation of prophylaxis group by manual chart review vs. our automated process was found to agree in categorization for 39/40 (97.5%) sampled encounters. When comparing the cohort that received pharmacologic prophylaxis against the cohort that did not, there were significant differences in racial distribution, sex, BMI, and average LOS as shown in Table 1. Those who received pharmacologic prophylaxis were found to be significantly older than those who did not (62.7 years versus 53.2 years, P < 0.001), more likely to be male (50.6% vs, 42.4%, P < 0.001), more likely to have hospital-acquired VTE (2.2% vs. 0.5%, P < 0.001), and to have a shorter LOS (7.1 days vs. 9.8, P < 0.001).

Distribution of Patient Characteristics in Cohort
Table 1

Within the cohort group receiving pharmacologic prophylaxis (n = 6574), hospital-acquired VTE occurred in patients who were significantly younger (58.2 years vs. 62.8 years, P = 0.003) with a greater LOS (23.8 days vs. 6.7, P < 0.001) than those without. Within the group receiving no pharmacologic prophylaxis (n = 24,152), hospital-acquired VTE occurred in patients who were significantly older (57.1 years vs. 53.2 years, P = 0.014) with more than twice the LOS (20.2 days vs. 9.7 days, P < 0.001) compared to those without. Sixty-six of 75 (88%) randomly selected patients in which new VTE was identified by the automated electronic query had this diagnosis confirmed during manual chart review.

As shown in Table 2, automated calculation on a subsample of 300 randomly selected patients using APPS had a mean of 5.5 (SD, 2.9) while manual calculation of the original PPS on the same patients had a mean of 5.1 (SD, 2.6). There was no significant difference in mean between manual calculation and APPS (P = 0.073). There were, however, significant differences in how often individual criteria were considered present. The largest contributors to the difference in scores between APPS and manual calculation were “prior VTE” (positive, 16% vs. 8.3%, respectively) and “reduced mobility” (positive, 74.3% vs. 66%, respectively) as shown in Table 2. In the subsample, there were a total of 6 (2.0%) hospital-acquired VTE events. APPS’ automated calculation had an AUC = 0.79 (CI, 0.63-0.95) that was significant (P = 0.016) with a cutoff value of 5. Chart review’s manual calculation of the PPS had an AUC = 0.76 (CI 0.61-0.91) that was also significant (P = 0.029).

Distribution of Patient Characteristics in Cohort

Comparison of APPS to Manual Calculation of PPS
Table 2


Our entire cohort of 30,726 unique patients admitted during the study period included 260 (0.8%) who experienced hospital-acquired VTEs (Table 3). In patients receiving no pharmacologic prophylaxis, the average APPS was 4.0 (SD, 2.4) for those without VTE and 7.1 (SD, 2.3) for those with VTE. In patients who had received pharmacologic prophylaxis, those without hospital-acquired VTE had an average APPS of 4.9 (SD, 2.6) and those with hospital-acquired VTE averaged 7.7 (SD, 2.6). APPS’ ROC curves for “no pharmacologic prophylaxis” had an AUC = 0.81 (CI, 0.79 – 0.83) that was significant (P < 0.001) with a cutoff value of 5. There was similar performance in the pharmacologic prophylaxis group with an AUC = 0.79 (CI, 0.76 – 0.82) and cutoff value of 5, as shown in the Figure. Over the entire cohort, APPS had a sensitivity of 85.4%, specificity of 53.3%, positive predictive value (PPV) of 1.5%, and a negative predictive value (NPV) of 99.8% when using a cutoff of 5. The average APPS calculation time was 0.03 seconds per encounter. Additional information on individual criteria can be found in Table 3.

ROC curves and predictive characteristics of the APPS
Figure

 

 

DISCUSSION

Automated calculation of APPS using EHR data from prior encounters and the first 4 hours of admission was predictive of in-hospital VTE. APPS performed as well as traditional manual score calculation of the PPS. It was able to do so with no physician input, significantly lessening the burden of calculation and potentially increasing frequency of data-driven VTE risk assessment.

While automated calculation of certain scores is becoming more common, risk calculators that require data beyond vital signs and lab results have lagged,16-19 in part because of uncertainty about 2 issues. The first is whether EHR data accurately represent the current clinical picture. The second is if a machine-interpretable algorithm to determine a clinical status (eg, “active cancer”) would be similar to a doctor’s perception of that same concept. We attempted to better understand these 2 challenges through developing APPS. Concerning accuracy, EHR data correctly represent the clinical scenario: designations of VTEP and hospital-acquired VTE were accurate in approximately 90% of reviewed cases. Regarding the second concern, when comparing APPS to manual calculation, we found significant differences (P < 0.001) in how often 8 of the 11 criteria were positive, yet no significant difference in overall score and similar predictive capacity. Manual calculation appeared more likely to find data in the index encounter or in structured data. For example, “active cancer” may be documented only in a physician’s note, easily accounted for during a physician’s calculation but missed by APPS looking only for structured data. In contrast, automated calculation found historic criteria, such as “prior VTE” or “known thrombophilic condition,” positive more often. If the patient is being admitted for a problem unrelated to blood clots, the physician may have little time or interest to look through hundreds of EHR documents to discover a 2-year-old VTE. As patients’ records become larger and denser, more historic data can become buried and forgotten. While the 2 scores differ on individual criteria, they are similarly predictive and able to bifurcate the at-risk population to those who should and should not receive pharmacologic prophylaxis.

APPS Criteria by Prophylaxis and VTE Occurrence
Table 3

The APPS was found to have near-equal performance in the pharmacologic vs. no pharmacologic prophylaxis cohorts. This finding agrees with a study that found no significant difference in predicting 90-day VTE when looking at 86 risk factors vs. the most significant 4, none of which related to prescribed prophylaxis.18 The original PPS had a reported sensitivity of 94.6%, specificity 62%, PPV 7.5%, and NPV 99.7% in its derivation cohort.13 We matched APPS to the ratio of sensitivity to specificity, using 5 as the cutoff value. APPS performed slightly worse with sensitivity of 85.4%, specificity 53.3%, PPV 1.5%, and NPV 99.8%. This difference may have resulted from the original PPS study’s use of 90-day follow-up to determine VTE occurrence, whereas we looked only until the end of current hospitalization, an average of 9.2 days. Furthermore, the PPS had significantly poorer performance (AUC = 0.62) than that seen in the original derivation cohort in a separate study that manually calculated the score on more than 1000 patients.15

There are important limitations to our study. It was done at a single academic institution using a dataset of VTE-associated, validated research that was well-known to the researchers.20 Another major limitation is the dependence of the algorithm on data available within the first 4 hours of admission and earlier; thus, previous encounters may frequently play an important role. Patients presenting to our health system for the first time would have significantly fewer data available at the time of calculation. Additionally, our data could not reliably tell us the total doses of pharmacologic prophylaxis that a patient received. While most patients will maintain a consistent VTEP regimen once initiated in the hospital, 2 patients with the same LOS may have received differing amounts of pharmacologic prophylaxis. This research study did not assess how much time automatic calculation of VTE risk might save providers, because we did not record the time for each manual abstraction; however, from discussion with the main abstracter, chart review and manual calculation for this study took from 2 to 14 minutes per patient, depending on the number of previous interactions with the health system. Finally, although we chose data elements that are likely to exist at most institutions using an EHR, many institutions’ EHRs do not have EDW capabilities nor programmers who can assist with an automated risk score.

The EHR interventions to assist providers in determining appropriate VTEP have been able to increase rates of VTEP and decrease VTE-associated mortality.16,21 In addition to automating the calculation of guideline-adherent risk scores, there is a need for wider adoption for clinical decision support for VTE. For this reason, we chose only structured data fields from some of the most common elements within our EHR’s data warehouse to derive APPS (Appendix 1). Our study supports the idea that automated calculation of scores requiring input of more complex data such as diagnoses, recent medical events, and current clinical status remains predictive of hospital-acquired VTE risk. Because it is calculated automatically in the background while the clinician completes his or her assessment, the APPS holds the potential to significantly reduce the burden on providers while making guideline-adherent risk assessment more readily accessible. Further research is required to determine the exact amount of time automatic calculation saves, and, more important, if the relatively high predictive capacity we observed using APPS would be reproducible across institutions and could reduce incidence of hospital-acquired VTE.

 

 

Disclosures

Dr. Auerbach was supported by NHLBI K24HL098372 during the period of this study. Dr. Khanna, who is an implementation scientist at the University of California San Francisco Center for Digital Health Innovation, is the principal inventor of CareWeb, and may benefit financially from its commercialization. The other authors report no financial conflicts of interest.

References

1. Galson S. The Surgeon General’s call to action to prevent deep vein thrombosis and pulmonary embolism. 2008. https://www.ncbi.nlm.nih.gov/books/NBK44178/. Accessed February 11, 2016. PubMed
2. Borch KH, Nyegaard C, Hansen JB, et al. Joint effects of obesity and body height on the risk of venous thromboembolism: the Tromsø study. Arterioscler Thromb Vasc Biol. 2011;31(6):1439-44. PubMed
3. Braekkan SK, Borch KH, Mathiesen EB, Njølstad I, Wilsgaard T, Hansen JB.. Body height and risk of venous thromboembolism: the Tromsø Study. Am J Epidemiol. 2010;171(10):1109-1115. PubMed
4. Bounameaux H, Rosendaal FR. Venous thromboembolism: why does ethnicity matter? Circulation. 2011;123(200:2189-2191. PubMed
5. Spyropoulos AC, Anderson FA Jr, Fitzgerald G, et al; IMPROVE Investigators. Predictive and associative models to identify hospitalized medical patients at risk for VTE. Chest. 2011;140(3):706-714. PubMed
6. Rothberg MB, Lindenauer PK, Lahti M, Pekow PS, Selker HP. Risk factor model to predict venous thromboembolism in hospitalized medical patients. J Hosp Med. 2011;6(4):202-209. PubMed
7. Perioperative Management of Antithrombotic Therapy: Prevention of VTE in Nonsurgical Patients: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(6):1645.
8. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521-526. PubMed
9. Alvarez CA, Clark CA, Zhang S, et al. Predicting out of intensive care unit cardiopulmonary arrest or death using electronic medical record data. BMC Med Inform Decis Mak. 2013;13:28. PubMed
10. Escobar GJ, LaGuardia JC, Turk BJ, Ragins A, Kipnis P, Draper D. Early detection of impending physiologic deterioration among patients who are not in intensive care: development of predictive models using data from an automated electronic medical record. J Hosp Med. 2012;7(5):388-395. PubMed
11. Umscheid CA, Hanish A, Chittams J, Weiner MG, Hecht TE. Effectiveness of a novel and scalable clinical decision support intervention to improve venous thromboembolism prophylaxis: a quasi-experimental study. BMC Med Inform Decis Mak. 2012;12:92. PubMed
12. Tepas JJ 3rd, Rimar JM, Hsiao AL, Nussbaum MS. Automated analysis of electronic medical record data reflects the pathophysiology of operative complications. Surgery. 2013;154(4):918-924. PubMed
13. Barbar S, Noventa F, Rossetto V, et al. A risk assessment model for the identification of hospitalized medical patients at risk for venous thromboembolism: the Padua Prediction Score. J Thromb Haemost. 2010; 8(11):2450-2457. PubMed
14. Khanna R, Maynard G, Sadeghi B, et al. Incidence of hospital-acquired venous thromboembolic codes in medical patients hospitalized in academic medical centers. J Hosp Med. 2014; 9(4):221-225. PubMed
15. Vardi M, Ghanem-Zoubi NO, Zidan R, Yurin V, Bitterman H. Venous thromboembolism and the utility of the Padua Prediction Score in patients with sepsis admitted to internal medicine departments. J Thromb Haemost. 2013;11(3):467-473. PubMed
16. Samama MM, Dahl OE, Mismetti P, et al. An electronic tool for venous thromboembolism prevention in medical and surgical patients. Haematologica. 2006;91(1):64-70. PubMed
17. Mann DM, Kannry JL, Edonyabo D, et al. Rationale, design, and implementation protocol of an electronic health record integrated clinical prediction rule (iCPR) randomized trial in primary care. Implement Sci. 2011;6:109. PubMed
18. Woller SC, Stevens SM, Jones JP, et al. Derivation and validation of a simple model to identify venous thromboembolism risk in medical patients. Am J Med. 2011;124(10):947-954. PubMed
19. Huang W, Anderson FA, Spencer FA, Gallus A, Goldberg RJ. Risk-assessment models for predicting venous thromboembolism among hospitalized non-surgical patients: a systematic review. J Thromb Thrombolysis. 2013;35(1):67-80. PubMed
20. Khanna RR, Kim SB, Jenkins I, et al. Predictive value of the present-on-admission indicator for hospital-acquired venous thromboembolism. Med Care. 2015;53(4):e31-e36. PubMed
21. Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous thromboembolism a
mong hospitalized patients. N Engl J Med. 2005;352(10):969-977. PubMed

References

1. Galson S. The Surgeon General’s call to action to prevent deep vein thrombosis and pulmonary embolism. 2008. https://www.ncbi.nlm.nih.gov/books/NBK44178/. Accessed February 11, 2016. PubMed
2. Borch KH, Nyegaard C, Hansen JB, et al. Joint effects of obesity and body height on the risk of venous thromboembolism: the Tromsø study. Arterioscler Thromb Vasc Biol. 2011;31(6):1439-44. PubMed
3. Braekkan SK, Borch KH, Mathiesen EB, Njølstad I, Wilsgaard T, Hansen JB.. Body height and risk of venous thromboembolism: the Tromsø Study. Am J Epidemiol. 2010;171(10):1109-1115. PubMed
4. Bounameaux H, Rosendaal FR. Venous thromboembolism: why does ethnicity matter? Circulation. 2011;123(200:2189-2191. PubMed
5. Spyropoulos AC, Anderson FA Jr, Fitzgerald G, et al; IMPROVE Investigators. Predictive and associative models to identify hospitalized medical patients at risk for VTE. Chest. 2011;140(3):706-714. PubMed
6. Rothberg MB, Lindenauer PK, Lahti M, Pekow PS, Selker HP. Risk factor model to predict venous thromboembolism in hospitalized medical patients. J Hosp Med. 2011;6(4):202-209. PubMed
7. Perioperative Management of Antithrombotic Therapy: Prevention of VTE in Nonsurgical Patients: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(6):1645.
8. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521-526. PubMed
9. Alvarez CA, Clark CA, Zhang S, et al. Predicting out of intensive care unit cardiopulmonary arrest or death using electronic medical record data. BMC Med Inform Decis Mak. 2013;13:28. PubMed
10. Escobar GJ, LaGuardia JC, Turk BJ, Ragins A, Kipnis P, Draper D. Early detection of impending physiologic deterioration among patients who are not in intensive care: development of predictive models using data from an automated electronic medical record. J Hosp Med. 2012;7(5):388-395. PubMed
11. Umscheid CA, Hanish A, Chittams J, Weiner MG, Hecht TE. Effectiveness of a novel and scalable clinical decision support intervention to improve venous thromboembolism prophylaxis: a quasi-experimental study. BMC Med Inform Decis Mak. 2012;12:92. PubMed
12. Tepas JJ 3rd, Rimar JM, Hsiao AL, Nussbaum MS. Automated analysis of electronic medical record data reflects the pathophysiology of operative complications. Surgery. 2013;154(4):918-924. PubMed
13. Barbar S, Noventa F, Rossetto V, et al. A risk assessment model for the identification of hospitalized medical patients at risk for venous thromboembolism: the Padua Prediction Score. J Thromb Haemost. 2010; 8(11):2450-2457. PubMed
14. Khanna R, Maynard G, Sadeghi B, et al. Incidence of hospital-acquired venous thromboembolic codes in medical patients hospitalized in academic medical centers. J Hosp Med. 2014; 9(4):221-225. PubMed
15. Vardi M, Ghanem-Zoubi NO, Zidan R, Yurin V, Bitterman H. Venous thromboembolism and the utility of the Padua Prediction Score in patients with sepsis admitted to internal medicine departments. J Thromb Haemost. 2013;11(3):467-473. PubMed
16. Samama MM, Dahl OE, Mismetti P, et al. An electronic tool for venous thromboembolism prevention in medical and surgical patients. Haematologica. 2006;91(1):64-70. PubMed
17. Mann DM, Kannry JL, Edonyabo D, et al. Rationale, design, and implementation protocol of an electronic health record integrated clinical prediction rule (iCPR) randomized trial in primary care. Implement Sci. 2011;6:109. PubMed
18. Woller SC, Stevens SM, Jones JP, et al. Derivation and validation of a simple model to identify venous thromboembolism risk in medical patients. Am J Med. 2011;124(10):947-954. PubMed
19. Huang W, Anderson FA, Spencer FA, Gallus A, Goldberg RJ. Risk-assessment models for predicting venous thromboembolism among hospitalized non-surgical patients: a systematic review. J Thromb Thrombolysis. 2013;35(1):67-80. PubMed
20. Khanna RR, Kim SB, Jenkins I, et al. Predictive value of the present-on-admission indicator for hospital-acquired venous thromboembolism. Med Care. 2015;53(4):e31-e36. PubMed
21. Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous thromboembolism a
mong hospitalized patients. N Engl J Med. 2005;352(10):969-977. PubMed

Issue
Journal of Hospital Medicine 12(4)
Issue
Journal of Hospital Medicine 12(4)
Page Number
231-237
Page Number
231-237
Topics
Article Type
Display Headline
Automating venous thromboembolism risk calculation using electronic health record data upon hospital admission: The automated Padua Prediction Score
Display Headline
Automating venous thromboembolism risk calculation using electronic health record data upon hospital admission: The automated Padua Prediction Score
Sections
Article Source

© 2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Pierre Elias, MD, Columbia University-New York Presbyterian Hospital, 622 West 168th Street, VC-205, New York, NY 10032; Telephone: 212-305-6354; Fax: 212-305-6279; E-mail: pae9043@nyp.org.
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Use ProPublica
Article PDF Media
Media Files

Impact of a Connected Care model on 30-day readmission rates from skilled nursing facilities

Article Type
Changed
Wed, 04/26/2017 - 13:38
Display Headline
Impact of a Connected Care model on 30-day readmission rates from skilled nursing facilities

Approximately 20% of hospitalized Medicare beneficiaries in the U.S. are discharged to skilled nursing facilities (SNFs) for post-acute care,1,2 and 23.5% of these patients are readmitted within 30 days.3 Because hospital readmissions are costly and associated with worse outcomes,4,5 30-day readmission rates are considered a quality indicator,6 and there are financial penalties for hospitals with higher than expected rates.7 As a result, hospitals invest substantial resources in programs to reduce readmissions.8-10 The SNFs represent an attractive target for readmission reduction efforts, since SNFs contribute a disproportionate share of readmissions.3,4 Because SNF patients are in a monitored environment with high medication adherence, risk factors for readmission likely differ between patients discharged to SNFs and those sent home. For example, 1 study showed that among heart failure patients with cognitive impairment, those discharged to SNFs had lower readmissions during the first 20 days, likely due to better medication adherence.11 Patients discharged to SNFs generally have more complex illnesses, lower functional status, and higher 1-year mortality than patients discharged to the community.12,13 Despite this, SNF patients might have infrequent contact with physicians. Federal regulations require only that patients discharged to SNFs need to be seen within 30 days and then at least once every 30 days thereafter.14 According to the 2014 Office of Inspector General report, one-third of Medicare beneficiaries in SNFs experience adverse events from substandard treatment, inadequate resident monitoring and failure or delay of necessary care, most of which are thought to be preventable.15

To address this issue, the Cleveland Clinic developed a program called “Connected Care SNF,” in which hospital-employed physicians and advanced practice professionals visit patients in selected SNFs 4 to 5 times per week, for the purpose of reducing preventable readmissions. The aim of this study was to assess whether the program reduced 30-day readmissions, and to identify which patients benefited most from the program.

METHODS

Setting and Intervention

The Cleveland Clinic main campus is a tertiary academic medical center with 1400 beds and approximately 50,000 admissions per year. In late 2012, the Cleveland Clinic implemented the Connected Care SNF program, wherein Cleveland Clinic physicians regularly visited patients who were discharged from the Cleveland Clinic main campus to 7 regional SNFs. Beginning in December 2012, these 7 high-volume referral SNFs that were not part of the Cleveland Clinic Health System (CCHS) agreed to participate in the program, which focused on reducing avoidable hospital readmissions and delivering quality care (Table 1). The Connected Care team, comprised of 2 geriatricians (1 of whom was also a palliative medicine specialist), 1 internist, 1 family physician, and 5 advanced practice professionals (nurse practitioners and physician assistants), provided medical services at the participating SNFs. These providers aimed to see patients 4 to 5 times per week, were available on site during working hours, and provided telephone coverage at nights and on weekends. All providers had access to hospital electronic medical records and could communicate with the discharging physician and with specialists familiar with the patient as needed. Prior to the admission, providers were informed about patient arrival and, at the time of admission to the SNF, providers reviewed medications and discussed goals of care with patients and their families. In the SNF, providers worked closely with staff members to deliver medications and timely treatment. They also met monthly with multidisciplinary teams for continuous quality improvement and to review outcomes. Patients at Connected Care SNFs who had their own physicians, including most long-stay and some short-stay residents, did not receive the Connected Care intervention. They constituted less than 10% of the patients discharged from Cleveland Clinic main campus.

Connected Care SNF Program
Table 1

 

 

Study Design and Population

We reviewed administrative and clinical data from a retrospective cohort of patients discharged to SNF from the Cleveland Clinic main campus from January 1, 2011 to December 31, 2014. We included all patients who were discharged to an SNF during the study period. Our main outcome measure was 30-day all-cause readmissions to any hospital in the Cleveland Clinic Health System (CCHS), including the main campus and 8 regional community hospitals. Study patients were followed until January 30, 2015 to capture 30-day readmissions. According to 2012 Medicare data, of CCHS patients who were readmitted within 30 days, 83% of pneumonia, 81% of major joint replacement, 72% of heart failure and 57% of acute myocardial infarction patients were readmitted to a CCHS facility. As the Cleveland Clinic main campus attracts cardiac patients from a 100+-mile radius, they may be more likely to seek care readmission near home and are not reflective of CCHS patients overall. Because we did not have access to readmissions data from non-CCHS hospitals, we excluded patients who were discharged to SNFs beyond a 25-mile radius from the main campus, where they may be more likely to utilize non-CCHS hospitals for acute hospitalization. We also excluded patients discharged to non-CCHS hospital-based SNFs, which may refer readmissions to their own hospital system. Because the Connected Care program began in December 2012, the years 2011-2012 served as the baseline period. The intervention was conducted at 7 SNFs. All other SNFs within the 25-mile radius were included as controls, except for 3 hospital-based SNFs that would be unlikely to admit patients to CCHS. We compared the change in all-cause 30-day readmission rates after implementation of Connected Care, using all patients discharged to SNFs within 25 miles to control for temporal changes in local readmission rates. Discharge to specific SNFs was determined solely by patient choice.

Data Collection

For each patient, we collected the following data that has been shown to be associated with readmissions:16-18 demographics (age, race, sex, ZIP code), lab values on discharge (hemoglobin and sodium); hemodialysis status; medicine or surgical service; elective surgery or nonelective surgery; details of the index admission index (diagnosis-related group [DRG], Medicare severity-diagnosis-related groups [MS-DRG] weight, primary diagnosis code; principal procedure code; admission date; discharge date, length of stay, and post-acute care provider); and common comorbidities, as listed in Table 2. We also calculated each patient’s HOSPITAL19,20 score. The HOSPITAL score was developed to predict risk of preventable 30-day readmissions,19 but it has also been validated to predict 30-day all-cause readmission rates for patients discharged to SNF.21 The model contains 7 elements (hemoglobin, oncology service, sodium, procedure, index type, admissions within the last year, length of stay) (supplemental Table).Patients with a high score (7 or higher) have a 41% chance of readmission, while those with a low score (4 or lower) have only a 15% chance. 21 We assessed all cause 30-day readmission status from CCHS administrative data. Observation patients and outpatient same-day surgeries were not considered to be admissions. For patients with multiple admissions, each admission was counted as a separate index hospitalization. Cleveland Clinic’s Institutional Review Board approved the study.

Characteristics of Patients Discharged in 2011-2012 vs. 2013-2014 to 7 Intervention SNFs and 103 Usual-Care SNFs
Table 2

Statistical Analysis

For the 7 intervention SNFs, patient characteristics were summarized as means and standard deviations or frequencies and percentages for the periods of 2011-2012 and 2013-2014, respectively, and the 2 periods were compared using the Student t test or χ2 test as appropriate.

Mixed-effects logistic regression models were used to model 30-day readmission rates. Since the intervention was implemented in the last quarter of 2012, we examined the difference in readmission rates before and after that time point. The model included the following fixed effects: SNF type (intervention or usual care), time points (quarters of 2011-2014), whether the time is pre- or postintervention (binary), and the 3-way interaction between SNF type, pre- or postintervention and time points, and patient characteristics. The model also contained a Gaussian random effect at the SNF level to account for possible correlations among the outcomes of patients from the same SNF. For each quarter, the mean adjusted readmission rates of 2 types of SNFs were calculated from the fitted mixed models and plotted over time. Furthermore, we compared the mean readmission rates of the 2 groups in the pre- and postintervention periods. Subgroup analyses were performed for medical and surgical patients, and for patients in the low, intermediate and high HOSPITAL score groups.

All analyses were performed using RStudio (Boston, Massachusetts). Statistical significance was established with 2-sided P values less than 0.05.

RESULTS

 

 

We identified 119 SNFs within a 25-mile radius of the hospital. Of these, 6 did not receive any referrals. Three non-CCHS hospital-based SNFs were excluded, leaving a total of 110 SNFs in the study sample: 7 intervention SNFs and 103 usual-care SNFs. Between January 2011 and December 2014, there were 23,408 SNF discharges from Cleveland Clinic main campus, including 13,544 who were discharged to study SNFs (Supplemental Figure). Of these, 3334 were discharged to 7 intervention SNFs and 10,210 were discharged to usual care SNFs. Characteristics of patients in both periods appear in Table 2. At baseline, patients in the intervention and control SNFs varied in a number of ways. Patients at intervention SNFs were older (75.6 vs. 70.2 years; P < 0.001), more likely to be African American (45.5% vs. 35.9%; P < 0.001), female (61% vs. 55.4%; P < 0.001) and to be insured by Medicare (85.2% vs. 71.4%; P < 0.001). Both groups had similar proportions of patients with high, intermediate, and low readmission risk as measured by HOSPITAL score. Compared to the 2011-2012 pre-intervention period, during the 2013-2014 intervention period, there were more surgeries (34.3% vs. 41.9%; P < 0.001), more elective surgeries (21.8% vs. 25.5%; P = 0.01), fewer medical patients (65.7% vs. 58.1%; P < 0.001), and an increase in comorbidities, including myocardial infarction, peripheral vascular disease, and liver disease (Table 2).

Adjusted 30-day Readmission Rates, 2011-2012 vs. 2013-2014 from 7 Intervention SNFs and 103 Usual-Care SNFs
Table 3

Table 3 shows adjusted 30-day readmissions rates, before and during the intervention period at the intervention and usual care SNFs. Compared to the pre-intervention period, 30-day all-cause adjusted readmission rates declined in the intervention SNFs (28.1% to 21.7%, P < 0.001), while it increased slightly at control sites (27.1% to 28.5%, P < 0.001). The Figure shows the adjusted 30-day readmission rates by quarter throughout the study period.

Adjusted 30-day readmission rates on 7 intervention SNF discharged patients
Figure

Declines in 30-day readmission rates were greater for medical patients (31.0% to 24.6%, P < 0.001) than surgical patients (22.4% to 17.7%, P < 0.001). Patients with high HOSPITAL scores had the greatest decline, while those with low HOSPITAL scores had smaller declines.

DISCUSSION

In this retrospective study of 4 years of discharges to 110 SNFs, we report on the impact of a Connected Care program, in which a physician visited patients on admission to the SNF and 4 to 5 times per week during their stay. Introduction of the program was followed by a 6.8% absolute reduction in all-cause 30-day readmission rates compared to usual care. The absolute reductions ranged from 4.6% for patients at low risk for readmission to 9.1% for patients at high risk, and medical patients benefited more than surgical patients.

Most studies of interventions to reduce hospital readmissions have focused on patients discharged to the community setting.7-9 Interventions have centered on discharge planning, medication reconciliation, and close follow-up to assess for medication adherence and early signs of deterioration. Because patients in SNFs have their medications administered by staff and are under frequent surveillance, such interventions are unlikely to be helpful in this population. We found no studies that focus on short-stay or skilled patients discharged to SNF. Two studies have demonstrated that interventions can reduce hospitalization from nursing homes.22,23 Neither study included readmissions. The Evercare model consisted of nurse practitioners providing active primary care services within the nursing home, as well as offering incentive payments to nursing homes for not hospitalizing patients.22 During a 2-year period, long term residents who enrolled in Evercare had an almost 50% reduction in incident hospitalizations compared to those who did not.22 INTERACT II was a quality improvement intervention that provided tools, education, and strategies to help identify and manage acute conditions proactively.23 In 25 nursing homes employing INTERACT II, there was a 17% reduction in self-reported hospital admissions during the 6-month project, with higher rates of reduction among nursing homes rated as more engaged in the process.23 Although nursing homes may serve some short-stay or skilled patients, they generally serve long-term populations, and studies have shown that short-stay patients are at higher risk for 30-day readmissions.24

There are a number of reasons that short-term SNF patients are at higher risk for readmission. Although prior to admission, they were considered hospital level of care and received a physician visit daily, on transfer to the SNF, relatively little medical care is available. Current federal regulations regarding physician services at a SNF require the resident to be seen by a physician at least once every 30 days for the first 90 days after admission, and at least once every 60 days thereafter.25

The Connected Care program physicians provided a smooth transition of care from hospital to SNF as well as frequent reassessment. Physicians were alerted prior to hospital discharge and performed an initial comprehensive visit generally on the day of admission to the SNF and always within 48 hours. The initial evaluation is important because miscommunication during the handoff from hospital to SNF may result in incorrect medication regimens or inaccurate assessments. By performing prompt medication reconciliation and periodic reassessments of a patient’s medical condition, the Connected Care providers recreate some of the essential elements of successful outpatient readmissions prevention programs.

They also worked together with each SNF’s interdisciplinary team to deliver quality care. There were monthly meetings at each participating Connected Care SNF. Physicians reviewed monthly 30-day readmissions and performed root-cause analysis. When they discovered challenges to timely medication and treatment delivery during daily rounds, they provided in-services to SNF nurses.

In addition, Connected Care providers discussed goals of care—something that is often overlooked on admission to a SNF. This is particularly important because patients with chronic illnesses who are discharged to SNF often have poor prognoses. For example, Medicare patients with heart failure who are discharged to SNFs have 1-year mortality in excess of 50%.13 By implementing a plan of care consistent with patient and family goals, inappropriate readmissions for terminal patients may be avoided.

Reducing readmissions is important for hospitals because under the Hospital Readmissions Reduction Program, hospitals now face substantial penalties for higher than expected readmissions rates. Hospitals involved in bundled payments or other total cost-of-care arrangements have additional incentive to avoid readmissions. Beginning in 2019, SNFs will also receive incentive payments based on their 30-day all-cause hospital readmissions as part of the Skilled Nursing Facility Value-Based Purchasing program.25 The Connected Care model offers 1 means of achieving this goal through partnership between hospitals and SNFs.

Our study has several limitations. First, our study was observational in nature, so the observed reduction in readmissions could have been due to temporal trends unrelated to the intervention. However, no significant reduction was noted during the same time period in other area SNFs. There was also little change in the characteristics of patients admitted to the intervention SNFs. Importantly, the HOSPITAL score, which can predict 30-day readmission rates,20 did not change throughout the study period. Second, the results reflect patients discharged from a single hospital and may not be generalizable to other geographic areas. However, because the program included 7 SNFs, we believe it could be reproduced in other settings. Third, our readmissions measure included only those patients who returned to a CCHS facility. Although we may have missed some readmissions to other hospital systems, such leakage is uncommon—more than 80% of CCHS patients are readmitted to CCHS facilities—and would be unlikely to differ across the short duration of the study. Finally, at the intervention SNFs, most long-stay and some short-stay residents did not receive the Connected Care intervention because they were cared for by their own physicians who did not participate in Connected Care. Had these patients’ readmissions been excluded from our results, the intervention might appear even more effective.

 

 

CONCLUSION

A Connected Care intervention reduced 30-day readmission rates among patients discharged to SNFs from a tertiary academic center. While all subgroups had substantial reductions in readmissions following the implementation of the intervention, patients who are at the highest risk of readmission benefited the most. Further study is necessary to know whether Connected Care can be reproduced in other health care systems and whether it reduces overall costs.

Acknowledgments

The authors would like to thank Michael Felver, MD, and teams for their clinical care of patients; Michael Felver, MD, William Zafirau, MD, Dan Blechschmid, MHA, and Kathy Brezine, and Seth Vilensky, MBA, for their administrative support; and Brad Souder, MPT, for assistance with data collection.

Disclosure

Nothing to report.

Files
References

1. Medicare Payment Advisory Commission. Report to the Congress: Medicare Payment Policy. Chapter 8. Skilled Nursing Facility Services. March 2013. http://www.medpac.gov/docs/default-source/reports/mar13_entirereport.pdf?sfvrsn=0. Accessed March 1, 2017.
2. Kim DG, Messinger-Rapport BJ. Clarion call for a dedicated clinical and research approach to post-acute care. J Am Med Dir Assoc. 2014;15(8):607. e1-e3. PubMed
3. Mor V, Intrator O, Feng Z, Grabowski D. The revolving door of rehospitalization from skilled nursing facilities. Health Aff. 2010;29(1):57-64. PubMed
4. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service program. N Engl J Med. 2009;360(14):1418-1428. PubMed
5. Creditor MC. Hazards of hospitalization of the elderly. Ann Intern Med 1993;118(3):219-223. PubMed
6. Van Walraven C, Bennett C, Jennings A, Austin PC, Forester AJ. Proportion of hospital readmissions deemed avoidable: a systematic review. CMAJ. 2011;183(7):E391-E402. PubMed
7. Brenson RA, Paulus RA, Kalman NS. Medicare’s readmissions-reduction program – a positive alternative. N Engl J Med 2012;366(15):1364-1366. PubMed
8. Jack BW, Chetty VK, Anthony D, et al. A reengineered hospital discharge program to decrease rehospitalization: a randomized trial. Ann Intern Med. 2009;150(3):178-187. PubMed
9. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow-up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613-620. PubMed
10. Coleman EA, Parry C, Chalmers S, Min SJ. The care transition intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):1822-1828. PubMed
11. Patel A, Parikh R, Howell EH, Hsich E, Landers SH, Gorodeski EZ. Mini-cog performance: novel marker of post discharge risk among patients hospitalized for heart failure. Circ Heart Fail. 2015;8(1):8-16. PubMed
12. Walter LC, Brand RJ, Counsell SR, et al. Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization. JAMA. 2001;285(23):2987-2994. PubMed
13. Allen LA, Hernandez AF, Peterson ED, et al. Discharge to a skilled nursing facility and subsequent clinical outcomes among older patients hospitalized for heart failure. Circ Heart Fail. 2011;4(3):293-300. PubMed
14. 42 CFR 483.40 – Physician services. US government Publishing Office. https://www.gpo.gov/fdsys/granule/CFR-2011-title42-vol5/CFR-2011-title42-vol5-sec483-40. Published October 1, 2011. Accessed August 31, 2016.
15. Office of Inspector General. Adverse Events in Skilled Nursing Facilities: National Incidence among Medicare Beneficiaries. OEI-06-11-00370. February 2014. http://oig.hhs.gov/oei/reports/oei-06-11-00370.pdf. Accessed March 22, 2016.
16. Hasan O, Meltzer DO, Shaykevich SA, et al. Hospital readmission in general medicine patients: a prediction model. J Gen Intern Med. 2010;25(3):211-219. PubMed
17. Boult C, Dowd B, McCaffrey D, Boult L, Hernandez R, Krulewitch H. Screening elders for risk of hospital admission. J Am Geriatr Soc. 1993;41(8):811-817. PubMed
18. Silverstein MD, Qin H, Mercer SQ, Fong J, Haydar Z. Risk factors for 30-day hospital readmission in patients ≥65 years of age. Proc (Bayl Univ Med Cent). 2008;21(4):363-372. PubMed
19. Donzé J, Aujesky D, Williams D, Schnipper JL. Potentially avoidable 30-day hospital readmissions in medical patients: derivation and validation of a prediction model. JAMA Intern Med. 2013;173(8):632-638. PubMed
20. Donzé JD, Williams MV, Robinson EJ, et al. International validity of the HOSPITAL score to predict 30-day potentially avoidable hospital readmissions. JAMA Intern Med. 2016;176(4):496-502. PubMed
21. Kim LD, Kou L, Messinger-Rapport BJ, Rothberg MB. Validation of the HOSPITAL score for 30-day all-cause readmissions of patients discharged to skilled nursing facilities. J Am Med Dir Assoc. 2016;17(9):e15-e18. PubMed
22. Kane RL, Keckhafer G, Flood S, Bershardsky B, Siadaty MS. The effect of Evercare on hospital use. J Am Geriatr Soc. 2003;51(10):1427-1434. PubMed
23. Ouslander JG, Lamb G, Tappen R, et al. Interventions to reduce hospitalizations from nursing homes: Evaluation of the INTERACT II collaboration quality improvement project. J Am Geriatr Soc. 2011;59(4):745-753. PubMed
24. Cost drivers for dually eligible beneficiaries: Potentially avoidable hospitalizations from nursing facility, skilled nursing facility, and home and community based service waiver programs. http://www.cms.gov/Research-Statistics-Data-and-Systems/Statistics-Trends-and-Reports/Reports/downloads/costdriverstask2.pdf. Accessed August 31, 2016.
25. H.R. 4302 (113th), Section 215, Protecting Access to Medicare Act of 2014 (PAMA). April 2, 2014. https://www.govtrack.us/congress/bills/113/hr4302/text. Accessed August 31, 2016.

Article PDF
Issue
Journal of Hospital Medicine 12(4)
Topics
Page Number
238-244
Sections
Files
Files
Article PDF
Article PDF

Approximately 20% of hospitalized Medicare beneficiaries in the U.S. are discharged to skilled nursing facilities (SNFs) for post-acute care,1,2 and 23.5% of these patients are readmitted within 30 days.3 Because hospital readmissions are costly and associated with worse outcomes,4,5 30-day readmission rates are considered a quality indicator,6 and there are financial penalties for hospitals with higher than expected rates.7 As a result, hospitals invest substantial resources in programs to reduce readmissions.8-10 The SNFs represent an attractive target for readmission reduction efforts, since SNFs contribute a disproportionate share of readmissions.3,4 Because SNF patients are in a monitored environment with high medication adherence, risk factors for readmission likely differ between patients discharged to SNFs and those sent home. For example, 1 study showed that among heart failure patients with cognitive impairment, those discharged to SNFs had lower readmissions during the first 20 days, likely due to better medication adherence.11 Patients discharged to SNFs generally have more complex illnesses, lower functional status, and higher 1-year mortality than patients discharged to the community.12,13 Despite this, SNF patients might have infrequent contact with physicians. Federal regulations require only that patients discharged to SNFs need to be seen within 30 days and then at least once every 30 days thereafter.14 According to the 2014 Office of Inspector General report, one-third of Medicare beneficiaries in SNFs experience adverse events from substandard treatment, inadequate resident monitoring and failure or delay of necessary care, most of which are thought to be preventable.15

To address this issue, the Cleveland Clinic developed a program called “Connected Care SNF,” in which hospital-employed physicians and advanced practice professionals visit patients in selected SNFs 4 to 5 times per week, for the purpose of reducing preventable readmissions. The aim of this study was to assess whether the program reduced 30-day readmissions, and to identify which patients benefited most from the program.

METHODS

Setting and Intervention

The Cleveland Clinic main campus is a tertiary academic medical center with 1400 beds and approximately 50,000 admissions per year. In late 2012, the Cleveland Clinic implemented the Connected Care SNF program, wherein Cleveland Clinic physicians regularly visited patients who were discharged from the Cleveland Clinic main campus to 7 regional SNFs. Beginning in December 2012, these 7 high-volume referral SNFs that were not part of the Cleveland Clinic Health System (CCHS) agreed to participate in the program, which focused on reducing avoidable hospital readmissions and delivering quality care (Table 1). The Connected Care team, comprised of 2 geriatricians (1 of whom was also a palliative medicine specialist), 1 internist, 1 family physician, and 5 advanced practice professionals (nurse practitioners and physician assistants), provided medical services at the participating SNFs. These providers aimed to see patients 4 to 5 times per week, were available on site during working hours, and provided telephone coverage at nights and on weekends. All providers had access to hospital electronic medical records and could communicate with the discharging physician and with specialists familiar with the patient as needed. Prior to the admission, providers were informed about patient arrival and, at the time of admission to the SNF, providers reviewed medications and discussed goals of care with patients and their families. In the SNF, providers worked closely with staff members to deliver medications and timely treatment. They also met monthly with multidisciplinary teams for continuous quality improvement and to review outcomes. Patients at Connected Care SNFs who had their own physicians, including most long-stay and some short-stay residents, did not receive the Connected Care intervention. They constituted less than 10% of the patients discharged from Cleveland Clinic main campus.

Connected Care SNF Program
Table 1

 

 

Study Design and Population

We reviewed administrative and clinical data from a retrospective cohort of patients discharged to SNF from the Cleveland Clinic main campus from January 1, 2011 to December 31, 2014. We included all patients who were discharged to an SNF during the study period. Our main outcome measure was 30-day all-cause readmissions to any hospital in the Cleveland Clinic Health System (CCHS), including the main campus and 8 regional community hospitals. Study patients were followed until January 30, 2015 to capture 30-day readmissions. According to 2012 Medicare data, of CCHS patients who were readmitted within 30 days, 83% of pneumonia, 81% of major joint replacement, 72% of heart failure and 57% of acute myocardial infarction patients were readmitted to a CCHS facility. As the Cleveland Clinic main campus attracts cardiac patients from a 100+-mile radius, they may be more likely to seek care readmission near home and are not reflective of CCHS patients overall. Because we did not have access to readmissions data from non-CCHS hospitals, we excluded patients who were discharged to SNFs beyond a 25-mile radius from the main campus, where they may be more likely to utilize non-CCHS hospitals for acute hospitalization. We also excluded patients discharged to non-CCHS hospital-based SNFs, which may refer readmissions to their own hospital system. Because the Connected Care program began in December 2012, the years 2011-2012 served as the baseline period. The intervention was conducted at 7 SNFs. All other SNFs within the 25-mile radius were included as controls, except for 3 hospital-based SNFs that would be unlikely to admit patients to CCHS. We compared the change in all-cause 30-day readmission rates after implementation of Connected Care, using all patients discharged to SNFs within 25 miles to control for temporal changes in local readmission rates. Discharge to specific SNFs was determined solely by patient choice.

Data Collection

For each patient, we collected the following data that has been shown to be associated with readmissions:16-18 demographics (age, race, sex, ZIP code), lab values on discharge (hemoglobin and sodium); hemodialysis status; medicine or surgical service; elective surgery or nonelective surgery; details of the index admission index (diagnosis-related group [DRG], Medicare severity-diagnosis-related groups [MS-DRG] weight, primary diagnosis code; principal procedure code; admission date; discharge date, length of stay, and post-acute care provider); and common comorbidities, as listed in Table 2. We also calculated each patient’s HOSPITAL19,20 score. The HOSPITAL score was developed to predict risk of preventable 30-day readmissions,19 but it has also been validated to predict 30-day all-cause readmission rates for patients discharged to SNF.21 The model contains 7 elements (hemoglobin, oncology service, sodium, procedure, index type, admissions within the last year, length of stay) (supplemental Table).Patients with a high score (7 or higher) have a 41% chance of readmission, while those with a low score (4 or lower) have only a 15% chance. 21 We assessed all cause 30-day readmission status from CCHS administrative data. Observation patients and outpatient same-day surgeries were not considered to be admissions. For patients with multiple admissions, each admission was counted as a separate index hospitalization. Cleveland Clinic’s Institutional Review Board approved the study.

Characteristics of Patients Discharged in 2011-2012 vs. 2013-2014 to 7 Intervention SNFs and 103 Usual-Care SNFs
Table 2

Statistical Analysis

For the 7 intervention SNFs, patient characteristics were summarized as means and standard deviations or frequencies and percentages for the periods of 2011-2012 and 2013-2014, respectively, and the 2 periods were compared using the Student t test or χ2 test as appropriate.

Mixed-effects logistic regression models were used to model 30-day readmission rates. Since the intervention was implemented in the last quarter of 2012, we examined the difference in readmission rates before and after that time point. The model included the following fixed effects: SNF type (intervention or usual care), time points (quarters of 2011-2014), whether the time is pre- or postintervention (binary), and the 3-way interaction between SNF type, pre- or postintervention and time points, and patient characteristics. The model also contained a Gaussian random effect at the SNF level to account for possible correlations among the outcomes of patients from the same SNF. For each quarter, the mean adjusted readmission rates of 2 types of SNFs were calculated from the fitted mixed models and plotted over time. Furthermore, we compared the mean readmission rates of the 2 groups in the pre- and postintervention periods. Subgroup analyses were performed for medical and surgical patients, and for patients in the low, intermediate and high HOSPITAL score groups.

All analyses were performed using RStudio (Boston, Massachusetts). Statistical significance was established with 2-sided P values less than 0.05.

RESULTS

 

 

We identified 119 SNFs within a 25-mile radius of the hospital. Of these, 6 did not receive any referrals. Three non-CCHS hospital-based SNFs were excluded, leaving a total of 110 SNFs in the study sample: 7 intervention SNFs and 103 usual-care SNFs. Between January 2011 and December 2014, there were 23,408 SNF discharges from Cleveland Clinic main campus, including 13,544 who were discharged to study SNFs (Supplemental Figure). Of these, 3334 were discharged to 7 intervention SNFs and 10,210 were discharged to usual care SNFs. Characteristics of patients in both periods appear in Table 2. At baseline, patients in the intervention and control SNFs varied in a number of ways. Patients at intervention SNFs were older (75.6 vs. 70.2 years; P < 0.001), more likely to be African American (45.5% vs. 35.9%; P < 0.001), female (61% vs. 55.4%; P < 0.001) and to be insured by Medicare (85.2% vs. 71.4%; P < 0.001). Both groups had similar proportions of patients with high, intermediate, and low readmission risk as measured by HOSPITAL score. Compared to the 2011-2012 pre-intervention period, during the 2013-2014 intervention period, there were more surgeries (34.3% vs. 41.9%; P < 0.001), more elective surgeries (21.8% vs. 25.5%; P = 0.01), fewer medical patients (65.7% vs. 58.1%; P < 0.001), and an increase in comorbidities, including myocardial infarction, peripheral vascular disease, and liver disease (Table 2).

Adjusted 30-day Readmission Rates, 2011-2012 vs. 2013-2014 from 7 Intervention SNFs and 103 Usual-Care SNFs
Table 3

Table 3 shows adjusted 30-day readmissions rates, before and during the intervention period at the intervention and usual care SNFs. Compared to the pre-intervention period, 30-day all-cause adjusted readmission rates declined in the intervention SNFs (28.1% to 21.7%, P < 0.001), while it increased slightly at control sites (27.1% to 28.5%, P < 0.001). The Figure shows the adjusted 30-day readmission rates by quarter throughout the study period.

Adjusted 30-day readmission rates on 7 intervention SNF discharged patients
Figure

Declines in 30-day readmission rates were greater for medical patients (31.0% to 24.6%, P < 0.001) than surgical patients (22.4% to 17.7%, P < 0.001). Patients with high HOSPITAL scores had the greatest decline, while those with low HOSPITAL scores had smaller declines.

DISCUSSION

In this retrospective study of 4 years of discharges to 110 SNFs, we report on the impact of a Connected Care program, in which a physician visited patients on admission to the SNF and 4 to 5 times per week during their stay. Introduction of the program was followed by a 6.8% absolute reduction in all-cause 30-day readmission rates compared to usual care. The absolute reductions ranged from 4.6% for patients at low risk for readmission to 9.1% for patients at high risk, and medical patients benefited more than surgical patients.

Most studies of interventions to reduce hospital readmissions have focused on patients discharged to the community setting.7-9 Interventions have centered on discharge planning, medication reconciliation, and close follow-up to assess for medication adherence and early signs of deterioration. Because patients in SNFs have their medications administered by staff and are under frequent surveillance, such interventions are unlikely to be helpful in this population. We found no studies that focus on short-stay or skilled patients discharged to SNF. Two studies have demonstrated that interventions can reduce hospitalization from nursing homes.22,23 Neither study included readmissions. The Evercare model consisted of nurse practitioners providing active primary care services within the nursing home, as well as offering incentive payments to nursing homes for not hospitalizing patients.22 During a 2-year period, long term residents who enrolled in Evercare had an almost 50% reduction in incident hospitalizations compared to those who did not.22 INTERACT II was a quality improvement intervention that provided tools, education, and strategies to help identify and manage acute conditions proactively.23 In 25 nursing homes employing INTERACT II, there was a 17% reduction in self-reported hospital admissions during the 6-month project, with higher rates of reduction among nursing homes rated as more engaged in the process.23 Although nursing homes may serve some short-stay or skilled patients, they generally serve long-term populations, and studies have shown that short-stay patients are at higher risk for 30-day readmissions.24

There are a number of reasons that short-term SNF patients are at higher risk for readmission. Although prior to admission, they were considered hospital level of care and received a physician visit daily, on transfer to the SNF, relatively little medical care is available. Current federal regulations regarding physician services at a SNF require the resident to be seen by a physician at least once every 30 days for the first 90 days after admission, and at least once every 60 days thereafter.25

The Connected Care program physicians provided a smooth transition of care from hospital to SNF as well as frequent reassessment. Physicians were alerted prior to hospital discharge and performed an initial comprehensive visit generally on the day of admission to the SNF and always within 48 hours. The initial evaluation is important because miscommunication during the handoff from hospital to SNF may result in incorrect medication regimens or inaccurate assessments. By performing prompt medication reconciliation and periodic reassessments of a patient’s medical condition, the Connected Care providers recreate some of the essential elements of successful outpatient readmissions prevention programs.

They also worked together with each SNF’s interdisciplinary team to deliver quality care. There were monthly meetings at each participating Connected Care SNF. Physicians reviewed monthly 30-day readmissions and performed root-cause analysis. When they discovered challenges to timely medication and treatment delivery during daily rounds, they provided in-services to SNF nurses.

In addition, Connected Care providers discussed goals of care—something that is often overlooked on admission to a SNF. This is particularly important because patients with chronic illnesses who are discharged to SNF often have poor prognoses. For example, Medicare patients with heart failure who are discharged to SNFs have 1-year mortality in excess of 50%.13 By implementing a plan of care consistent with patient and family goals, inappropriate readmissions for terminal patients may be avoided.

Reducing readmissions is important for hospitals because under the Hospital Readmissions Reduction Program, hospitals now face substantial penalties for higher than expected readmissions rates. Hospitals involved in bundled payments or other total cost-of-care arrangements have additional incentive to avoid readmissions. Beginning in 2019, SNFs will also receive incentive payments based on their 30-day all-cause hospital readmissions as part of the Skilled Nursing Facility Value-Based Purchasing program.25 The Connected Care model offers 1 means of achieving this goal through partnership between hospitals and SNFs.

Our study has several limitations. First, our study was observational in nature, so the observed reduction in readmissions could have been due to temporal trends unrelated to the intervention. However, no significant reduction was noted during the same time period in other area SNFs. There was also little change in the characteristics of patients admitted to the intervention SNFs. Importantly, the HOSPITAL score, which can predict 30-day readmission rates,20 did not change throughout the study period. Second, the results reflect patients discharged from a single hospital and may not be generalizable to other geographic areas. However, because the program included 7 SNFs, we believe it could be reproduced in other settings. Third, our readmissions measure included only those patients who returned to a CCHS facility. Although we may have missed some readmissions to other hospital systems, such leakage is uncommon—more than 80% of CCHS patients are readmitted to CCHS facilities—and would be unlikely to differ across the short duration of the study. Finally, at the intervention SNFs, most long-stay and some short-stay residents did not receive the Connected Care intervention because they were cared for by their own physicians who did not participate in Connected Care. Had these patients’ readmissions been excluded from our results, the intervention might appear even more effective.

 

 

CONCLUSION

A Connected Care intervention reduced 30-day readmission rates among patients discharged to SNFs from a tertiary academic center. While all subgroups had substantial reductions in readmissions following the implementation of the intervention, patients who are at the highest risk of readmission benefited the most. Further study is necessary to know whether Connected Care can be reproduced in other health care systems and whether it reduces overall costs.

Acknowledgments

The authors would like to thank Michael Felver, MD, and teams for their clinical care of patients; Michael Felver, MD, William Zafirau, MD, Dan Blechschmid, MHA, and Kathy Brezine, and Seth Vilensky, MBA, for their administrative support; and Brad Souder, MPT, for assistance with data collection.

Disclosure

Nothing to report.

Approximately 20% of hospitalized Medicare beneficiaries in the U.S. are discharged to skilled nursing facilities (SNFs) for post-acute care,1,2 and 23.5% of these patients are readmitted within 30 days.3 Because hospital readmissions are costly and associated with worse outcomes,4,5 30-day readmission rates are considered a quality indicator,6 and there are financial penalties for hospitals with higher than expected rates.7 As a result, hospitals invest substantial resources in programs to reduce readmissions.8-10 The SNFs represent an attractive target for readmission reduction efforts, since SNFs contribute a disproportionate share of readmissions.3,4 Because SNF patients are in a monitored environment with high medication adherence, risk factors for readmission likely differ between patients discharged to SNFs and those sent home. For example, 1 study showed that among heart failure patients with cognitive impairment, those discharged to SNFs had lower readmissions during the first 20 days, likely due to better medication adherence.11 Patients discharged to SNFs generally have more complex illnesses, lower functional status, and higher 1-year mortality than patients discharged to the community.12,13 Despite this, SNF patients might have infrequent contact with physicians. Federal regulations require only that patients discharged to SNFs need to be seen within 30 days and then at least once every 30 days thereafter.14 According to the 2014 Office of Inspector General report, one-third of Medicare beneficiaries in SNFs experience adverse events from substandard treatment, inadequate resident monitoring and failure or delay of necessary care, most of which are thought to be preventable.15

To address this issue, the Cleveland Clinic developed a program called “Connected Care SNF,” in which hospital-employed physicians and advanced practice professionals visit patients in selected SNFs 4 to 5 times per week, for the purpose of reducing preventable readmissions. The aim of this study was to assess whether the program reduced 30-day readmissions, and to identify which patients benefited most from the program.

METHODS

Setting and Intervention

The Cleveland Clinic main campus is a tertiary academic medical center with 1400 beds and approximately 50,000 admissions per year. In late 2012, the Cleveland Clinic implemented the Connected Care SNF program, wherein Cleveland Clinic physicians regularly visited patients who were discharged from the Cleveland Clinic main campus to 7 regional SNFs. Beginning in December 2012, these 7 high-volume referral SNFs that were not part of the Cleveland Clinic Health System (CCHS) agreed to participate in the program, which focused on reducing avoidable hospital readmissions and delivering quality care (Table 1). The Connected Care team, comprised of 2 geriatricians (1 of whom was also a palliative medicine specialist), 1 internist, 1 family physician, and 5 advanced practice professionals (nurse practitioners and physician assistants), provided medical services at the participating SNFs. These providers aimed to see patients 4 to 5 times per week, were available on site during working hours, and provided telephone coverage at nights and on weekends. All providers had access to hospital electronic medical records and could communicate with the discharging physician and with specialists familiar with the patient as needed. Prior to the admission, providers were informed about patient arrival and, at the time of admission to the SNF, providers reviewed medications and discussed goals of care with patients and their families. In the SNF, providers worked closely with staff members to deliver medications and timely treatment. They also met monthly with multidisciplinary teams for continuous quality improvement and to review outcomes. Patients at Connected Care SNFs who had their own physicians, including most long-stay and some short-stay residents, did not receive the Connected Care intervention. They constituted less than 10% of the patients discharged from Cleveland Clinic main campus.

Connected Care SNF Program
Table 1

 

 

Study Design and Population

We reviewed administrative and clinical data from a retrospective cohort of patients discharged to SNF from the Cleveland Clinic main campus from January 1, 2011 to December 31, 2014. We included all patients who were discharged to an SNF during the study period. Our main outcome measure was 30-day all-cause readmissions to any hospital in the Cleveland Clinic Health System (CCHS), including the main campus and 8 regional community hospitals. Study patients were followed until January 30, 2015 to capture 30-day readmissions. According to 2012 Medicare data, of CCHS patients who were readmitted within 30 days, 83% of pneumonia, 81% of major joint replacement, 72% of heart failure and 57% of acute myocardial infarction patients were readmitted to a CCHS facility. As the Cleveland Clinic main campus attracts cardiac patients from a 100+-mile radius, they may be more likely to seek care readmission near home and are not reflective of CCHS patients overall. Because we did not have access to readmissions data from non-CCHS hospitals, we excluded patients who were discharged to SNFs beyond a 25-mile radius from the main campus, where they may be more likely to utilize non-CCHS hospitals for acute hospitalization. We also excluded patients discharged to non-CCHS hospital-based SNFs, which may refer readmissions to their own hospital system. Because the Connected Care program began in December 2012, the years 2011-2012 served as the baseline period. The intervention was conducted at 7 SNFs. All other SNFs within the 25-mile radius were included as controls, except for 3 hospital-based SNFs that would be unlikely to admit patients to CCHS. We compared the change in all-cause 30-day readmission rates after implementation of Connected Care, using all patients discharged to SNFs within 25 miles to control for temporal changes in local readmission rates. Discharge to specific SNFs was determined solely by patient choice.

Data Collection

For each patient, we collected the following data that has been shown to be associated with readmissions:16-18 demographics (age, race, sex, ZIP code), lab values on discharge (hemoglobin and sodium); hemodialysis status; medicine or surgical service; elective surgery or nonelective surgery; details of the index admission index (diagnosis-related group [DRG], Medicare severity-diagnosis-related groups [MS-DRG] weight, primary diagnosis code; principal procedure code; admission date; discharge date, length of stay, and post-acute care provider); and common comorbidities, as listed in Table 2. We also calculated each patient’s HOSPITAL19,20 score. The HOSPITAL score was developed to predict risk of preventable 30-day readmissions,19 but it has also been validated to predict 30-day all-cause readmission rates for patients discharged to SNF.21 The model contains 7 elements (hemoglobin, oncology service, sodium, procedure, index type, admissions within the last year, length of stay) (supplemental Table).Patients with a high score (7 or higher) have a 41% chance of readmission, while those with a low score (4 or lower) have only a 15% chance. 21 We assessed all cause 30-day readmission status from CCHS administrative data. Observation patients and outpatient same-day surgeries were not considered to be admissions. For patients with multiple admissions, each admission was counted as a separate index hospitalization. Cleveland Clinic’s Institutional Review Board approved the study.

Characteristics of Patients Discharged in 2011-2012 vs. 2013-2014 to 7 Intervention SNFs and 103 Usual-Care SNFs
Table 2

Statistical Analysis

For the 7 intervention SNFs, patient characteristics were summarized as means and standard deviations or frequencies and percentages for the periods of 2011-2012 and 2013-2014, respectively, and the 2 periods were compared using the Student t test or χ2 test as appropriate.

Mixed-effects logistic regression models were used to model 30-day readmission rates. Since the intervention was implemented in the last quarter of 2012, we examined the difference in readmission rates before and after that time point. The model included the following fixed effects: SNF type (intervention or usual care), time points (quarters of 2011-2014), whether the time is pre- or postintervention (binary), and the 3-way interaction between SNF type, pre- or postintervention and time points, and patient characteristics. The model also contained a Gaussian random effect at the SNF level to account for possible correlations among the outcomes of patients from the same SNF. For each quarter, the mean adjusted readmission rates of 2 types of SNFs were calculated from the fitted mixed models and plotted over time. Furthermore, we compared the mean readmission rates of the 2 groups in the pre- and postintervention periods. Subgroup analyses were performed for medical and surgical patients, and for patients in the low, intermediate and high HOSPITAL score groups.

All analyses were performed using RStudio (Boston, Massachusetts). Statistical significance was established with 2-sided P values less than 0.05.

RESULTS

 

 

We identified 119 SNFs within a 25-mile radius of the hospital. Of these, 6 did not receive any referrals. Three non-CCHS hospital-based SNFs were excluded, leaving a total of 110 SNFs in the study sample: 7 intervention SNFs and 103 usual-care SNFs. Between January 2011 and December 2014, there were 23,408 SNF discharges from Cleveland Clinic main campus, including 13,544 who were discharged to study SNFs (Supplemental Figure). Of these, 3334 were discharged to 7 intervention SNFs and 10,210 were discharged to usual care SNFs. Characteristics of patients in both periods appear in Table 2. At baseline, patients in the intervention and control SNFs varied in a number of ways. Patients at intervention SNFs were older (75.6 vs. 70.2 years; P < 0.001), more likely to be African American (45.5% vs. 35.9%; P < 0.001), female (61% vs. 55.4%; P < 0.001) and to be insured by Medicare (85.2% vs. 71.4%; P < 0.001). Both groups had similar proportions of patients with high, intermediate, and low readmission risk as measured by HOSPITAL score. Compared to the 2011-2012 pre-intervention period, during the 2013-2014 intervention period, there were more surgeries (34.3% vs. 41.9%; P < 0.001), more elective surgeries (21.8% vs. 25.5%; P = 0.01), fewer medical patients (65.7% vs. 58.1%; P < 0.001), and an increase in comorbidities, including myocardial infarction, peripheral vascular disease, and liver disease (Table 2).

Adjusted 30-day Readmission Rates, 2011-2012 vs. 2013-2014 from 7 Intervention SNFs and 103 Usual-Care SNFs
Table 3

Table 3 shows adjusted 30-day readmissions rates, before and during the intervention period at the intervention and usual care SNFs. Compared to the pre-intervention period, 30-day all-cause adjusted readmission rates declined in the intervention SNFs (28.1% to 21.7%, P < 0.001), while it increased slightly at control sites (27.1% to 28.5%, P < 0.001). The Figure shows the adjusted 30-day readmission rates by quarter throughout the study period.

Adjusted 30-day readmission rates on 7 intervention SNF discharged patients
Figure

Declines in 30-day readmission rates were greater for medical patients (31.0% to 24.6%, P < 0.001) than surgical patients (22.4% to 17.7%, P < 0.001). Patients with high HOSPITAL scores had the greatest decline, while those with low HOSPITAL scores had smaller declines.

DISCUSSION

In this retrospective study of 4 years of discharges to 110 SNFs, we report on the impact of a Connected Care program, in which a physician visited patients on admission to the SNF and 4 to 5 times per week during their stay. Introduction of the program was followed by a 6.8% absolute reduction in all-cause 30-day readmission rates compared to usual care. The absolute reductions ranged from 4.6% for patients at low risk for readmission to 9.1% for patients at high risk, and medical patients benefited more than surgical patients.

Most studies of interventions to reduce hospital readmissions have focused on patients discharged to the community setting.7-9 Interventions have centered on discharge planning, medication reconciliation, and close follow-up to assess for medication adherence and early signs of deterioration. Because patients in SNFs have their medications administered by staff and are under frequent surveillance, such interventions are unlikely to be helpful in this population. We found no studies that focus on short-stay or skilled patients discharged to SNF. Two studies have demonstrated that interventions can reduce hospitalization from nursing homes.22,23 Neither study included readmissions. The Evercare model consisted of nurse practitioners providing active primary care services within the nursing home, as well as offering incentive payments to nursing homes for not hospitalizing patients.22 During a 2-year period, long term residents who enrolled in Evercare had an almost 50% reduction in incident hospitalizations compared to those who did not.22 INTERACT II was a quality improvement intervention that provided tools, education, and strategies to help identify and manage acute conditions proactively.23 In 25 nursing homes employing INTERACT II, there was a 17% reduction in self-reported hospital admissions during the 6-month project, with higher rates of reduction among nursing homes rated as more engaged in the process.23 Although nursing homes may serve some short-stay or skilled patients, they generally serve long-term populations, and studies have shown that short-stay patients are at higher risk for 30-day readmissions.24

There are a number of reasons that short-term SNF patients are at higher risk for readmission. Although prior to admission, they were considered hospital level of care and received a physician visit daily, on transfer to the SNF, relatively little medical care is available. Current federal regulations regarding physician services at a SNF require the resident to be seen by a physician at least once every 30 days for the first 90 days after admission, and at least once every 60 days thereafter.25

The Connected Care program physicians provided a smooth transition of care from hospital to SNF as well as frequent reassessment. Physicians were alerted prior to hospital discharge and performed an initial comprehensive visit generally on the day of admission to the SNF and always within 48 hours. The initial evaluation is important because miscommunication during the handoff from hospital to SNF may result in incorrect medication regimens or inaccurate assessments. By performing prompt medication reconciliation and periodic reassessments of a patient’s medical condition, the Connected Care providers recreate some of the essential elements of successful outpatient readmissions prevention programs.

They also worked together with each SNF’s interdisciplinary team to deliver quality care. There were monthly meetings at each participating Connected Care SNF. Physicians reviewed monthly 30-day readmissions and performed root-cause analysis. When they discovered challenges to timely medication and treatment delivery during daily rounds, they provided in-services to SNF nurses.

In addition, Connected Care providers discussed goals of care—something that is often overlooked on admission to a SNF. This is particularly important because patients with chronic illnesses who are discharged to SNF often have poor prognoses. For example, Medicare patients with heart failure who are discharged to SNFs have 1-year mortality in excess of 50%.13 By implementing a plan of care consistent with patient and family goals, inappropriate readmissions for terminal patients may be avoided.

Reducing readmissions is important for hospitals because under the Hospital Readmissions Reduction Program, hospitals now face substantial penalties for higher than expected readmissions rates. Hospitals involved in bundled payments or other total cost-of-care arrangements have additional incentive to avoid readmissions. Beginning in 2019, SNFs will also receive incentive payments based on their 30-day all-cause hospital readmissions as part of the Skilled Nursing Facility Value-Based Purchasing program.25 The Connected Care model offers 1 means of achieving this goal through partnership between hospitals and SNFs.

Our study has several limitations. First, our study was observational in nature, so the observed reduction in readmissions could have been due to temporal trends unrelated to the intervention. However, no significant reduction was noted during the same time period in other area SNFs. There was also little change in the characteristics of patients admitted to the intervention SNFs. Importantly, the HOSPITAL score, which can predict 30-day readmission rates,20 did not change throughout the study period. Second, the results reflect patients discharged from a single hospital and may not be generalizable to other geographic areas. However, because the program included 7 SNFs, we believe it could be reproduced in other settings. Third, our readmissions measure included only those patients who returned to a CCHS facility. Although we may have missed some readmissions to other hospital systems, such leakage is uncommon—more than 80% of CCHS patients are readmitted to CCHS facilities—and would be unlikely to differ across the short duration of the study. Finally, at the intervention SNFs, most long-stay and some short-stay residents did not receive the Connected Care intervention because they were cared for by their own physicians who did not participate in Connected Care. Had these patients’ readmissions been excluded from our results, the intervention might appear even more effective.

 

 

CONCLUSION

A Connected Care intervention reduced 30-day readmission rates among patients discharged to SNFs from a tertiary academic center. While all subgroups had substantial reductions in readmissions following the implementation of the intervention, patients who are at the highest risk of readmission benefited the most. Further study is necessary to know whether Connected Care can be reproduced in other health care systems and whether it reduces overall costs.

Acknowledgments

The authors would like to thank Michael Felver, MD, and teams for their clinical care of patients; Michael Felver, MD, William Zafirau, MD, Dan Blechschmid, MHA, and Kathy Brezine, and Seth Vilensky, MBA, for their administrative support; and Brad Souder, MPT, for assistance with data collection.

Disclosure

Nothing to report.

References

1. Medicare Payment Advisory Commission. Report to the Congress: Medicare Payment Policy. Chapter 8. Skilled Nursing Facility Services. March 2013. http://www.medpac.gov/docs/default-source/reports/mar13_entirereport.pdf?sfvrsn=0. Accessed March 1, 2017.
2. Kim DG, Messinger-Rapport BJ. Clarion call for a dedicated clinical and research approach to post-acute care. J Am Med Dir Assoc. 2014;15(8):607. e1-e3. PubMed
3. Mor V, Intrator O, Feng Z, Grabowski D. The revolving door of rehospitalization from skilled nursing facilities. Health Aff. 2010;29(1):57-64. PubMed
4. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service program. N Engl J Med. 2009;360(14):1418-1428. PubMed
5. Creditor MC. Hazards of hospitalization of the elderly. Ann Intern Med 1993;118(3):219-223. PubMed
6. Van Walraven C, Bennett C, Jennings A, Austin PC, Forester AJ. Proportion of hospital readmissions deemed avoidable: a systematic review. CMAJ. 2011;183(7):E391-E402. PubMed
7. Brenson RA, Paulus RA, Kalman NS. Medicare’s readmissions-reduction program – a positive alternative. N Engl J Med 2012;366(15):1364-1366. PubMed
8. Jack BW, Chetty VK, Anthony D, et al. A reengineered hospital discharge program to decrease rehospitalization: a randomized trial. Ann Intern Med. 2009;150(3):178-187. PubMed
9. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow-up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613-620. PubMed
10. Coleman EA, Parry C, Chalmers S, Min SJ. The care transition intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):1822-1828. PubMed
11. Patel A, Parikh R, Howell EH, Hsich E, Landers SH, Gorodeski EZ. Mini-cog performance: novel marker of post discharge risk among patients hospitalized for heart failure. Circ Heart Fail. 2015;8(1):8-16. PubMed
12. Walter LC, Brand RJ, Counsell SR, et al. Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization. JAMA. 2001;285(23):2987-2994. PubMed
13. Allen LA, Hernandez AF, Peterson ED, et al. Discharge to a skilled nursing facility and subsequent clinical outcomes among older patients hospitalized for heart failure. Circ Heart Fail. 2011;4(3):293-300. PubMed
14. 42 CFR 483.40 – Physician services. US government Publishing Office. https://www.gpo.gov/fdsys/granule/CFR-2011-title42-vol5/CFR-2011-title42-vol5-sec483-40. Published October 1, 2011. Accessed August 31, 2016.
15. Office of Inspector General. Adverse Events in Skilled Nursing Facilities: National Incidence among Medicare Beneficiaries. OEI-06-11-00370. February 2014. http://oig.hhs.gov/oei/reports/oei-06-11-00370.pdf. Accessed March 22, 2016.
16. Hasan O, Meltzer DO, Shaykevich SA, et al. Hospital readmission in general medicine patients: a prediction model. J Gen Intern Med. 2010;25(3):211-219. PubMed
17. Boult C, Dowd B, McCaffrey D, Boult L, Hernandez R, Krulewitch H. Screening elders for risk of hospital admission. J Am Geriatr Soc. 1993;41(8):811-817. PubMed
18. Silverstein MD, Qin H, Mercer SQ, Fong J, Haydar Z. Risk factors for 30-day hospital readmission in patients ≥65 years of age. Proc (Bayl Univ Med Cent). 2008;21(4):363-372. PubMed
19. Donzé J, Aujesky D, Williams D, Schnipper JL. Potentially avoidable 30-day hospital readmissions in medical patients: derivation and validation of a prediction model. JAMA Intern Med. 2013;173(8):632-638. PubMed
20. Donzé JD, Williams MV, Robinson EJ, et al. International validity of the HOSPITAL score to predict 30-day potentially avoidable hospital readmissions. JAMA Intern Med. 2016;176(4):496-502. PubMed
21. Kim LD, Kou L, Messinger-Rapport BJ, Rothberg MB. Validation of the HOSPITAL score for 30-day all-cause readmissions of patients discharged to skilled nursing facilities. J Am Med Dir Assoc. 2016;17(9):e15-e18. PubMed
22. Kane RL, Keckhafer G, Flood S, Bershardsky B, Siadaty MS. The effect of Evercare on hospital use. J Am Geriatr Soc. 2003;51(10):1427-1434. PubMed
23. Ouslander JG, Lamb G, Tappen R, et al. Interventions to reduce hospitalizations from nursing homes: Evaluation of the INTERACT II collaboration quality improvement project. J Am Geriatr Soc. 2011;59(4):745-753. PubMed
24. Cost drivers for dually eligible beneficiaries: Potentially avoidable hospitalizations from nursing facility, skilled nursing facility, and home and community based service waiver programs. http://www.cms.gov/Research-Statistics-Data-and-Systems/Statistics-Trends-and-Reports/Reports/downloads/costdriverstask2.pdf. Accessed August 31, 2016.
25. H.R. 4302 (113th), Section 215, Protecting Access to Medicare Act of 2014 (PAMA). April 2, 2014. https://www.govtrack.us/congress/bills/113/hr4302/text. Accessed August 31, 2016.

References

1. Medicare Payment Advisory Commission. Report to the Congress: Medicare Payment Policy. Chapter 8. Skilled Nursing Facility Services. March 2013. http://www.medpac.gov/docs/default-source/reports/mar13_entirereport.pdf?sfvrsn=0. Accessed March 1, 2017.
2. Kim DG, Messinger-Rapport BJ. Clarion call for a dedicated clinical and research approach to post-acute care. J Am Med Dir Assoc. 2014;15(8):607. e1-e3. PubMed
3. Mor V, Intrator O, Feng Z, Grabowski D. The revolving door of rehospitalization from skilled nursing facilities. Health Aff. 2010;29(1):57-64. PubMed
4. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service program. N Engl J Med. 2009;360(14):1418-1428. PubMed
5. Creditor MC. Hazards of hospitalization of the elderly. Ann Intern Med 1993;118(3):219-223. PubMed
6. Van Walraven C, Bennett C, Jennings A, Austin PC, Forester AJ. Proportion of hospital readmissions deemed avoidable: a systematic review. CMAJ. 2011;183(7):E391-E402. PubMed
7. Brenson RA, Paulus RA, Kalman NS. Medicare’s readmissions-reduction program – a positive alternative. N Engl J Med 2012;366(15):1364-1366. PubMed
8. Jack BW, Chetty VK, Anthony D, et al. A reengineered hospital discharge program to decrease rehospitalization: a randomized trial. Ann Intern Med. 2009;150(3):178-187. PubMed
9. Naylor MD, Brooten D, Campbell R, et al. Comprehensive discharge planning and home follow-up of hospitalized elders: a randomized clinical trial. JAMA. 1999;281(7):613-620. PubMed
10. Coleman EA, Parry C, Chalmers S, Min SJ. The care transition intervention: results of a randomized controlled trial. Arch Intern Med. 2006;166(17):1822-1828. PubMed
11. Patel A, Parikh R, Howell EH, Hsich E, Landers SH, Gorodeski EZ. Mini-cog performance: novel marker of post discharge risk among patients hospitalized for heart failure. Circ Heart Fail. 2015;8(1):8-16. PubMed
12. Walter LC, Brand RJ, Counsell SR, et al. Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization. JAMA. 2001;285(23):2987-2994. PubMed
13. Allen LA, Hernandez AF, Peterson ED, et al. Discharge to a skilled nursing facility and subsequent clinical outcomes among older patients hospitalized for heart failure. Circ Heart Fail. 2011;4(3):293-300. PubMed
14. 42 CFR 483.40 – Physician services. US government Publishing Office. https://www.gpo.gov/fdsys/granule/CFR-2011-title42-vol5/CFR-2011-title42-vol5-sec483-40. Published October 1, 2011. Accessed August 31, 2016.
15. Office of Inspector General. Adverse Events in Skilled Nursing Facilities: National Incidence among Medicare Beneficiaries. OEI-06-11-00370. February 2014. http://oig.hhs.gov/oei/reports/oei-06-11-00370.pdf. Accessed March 22, 2016.
16. Hasan O, Meltzer DO, Shaykevich SA, et al. Hospital readmission in general medicine patients: a prediction model. J Gen Intern Med. 2010;25(3):211-219. PubMed
17. Boult C, Dowd B, McCaffrey D, Boult L, Hernandez R, Krulewitch H. Screening elders for risk of hospital admission. J Am Geriatr Soc. 1993;41(8):811-817. PubMed
18. Silverstein MD, Qin H, Mercer SQ, Fong J, Haydar Z. Risk factors for 30-day hospital readmission in patients ≥65 years of age. Proc (Bayl Univ Med Cent). 2008;21(4):363-372. PubMed
19. Donzé J, Aujesky D, Williams D, Schnipper JL. Potentially avoidable 30-day hospital readmissions in medical patients: derivation and validation of a prediction model. JAMA Intern Med. 2013;173(8):632-638. PubMed
20. Donzé JD, Williams MV, Robinson EJ, et al. International validity of the HOSPITAL score to predict 30-day potentially avoidable hospital readmissions. JAMA Intern Med. 2016;176(4):496-502. PubMed
21. Kim LD, Kou L, Messinger-Rapport BJ, Rothberg MB. Validation of the HOSPITAL score for 30-day all-cause readmissions of patients discharged to skilled nursing facilities. J Am Med Dir Assoc. 2016;17(9):e15-e18. PubMed
22. Kane RL, Keckhafer G, Flood S, Bershardsky B, Siadaty MS. The effect of Evercare on hospital use. J Am Geriatr Soc. 2003;51(10):1427-1434. PubMed
23. Ouslander JG, Lamb G, Tappen R, et al. Interventions to reduce hospitalizations from nursing homes: Evaluation of the INTERACT II collaboration quality improvement project. J Am Geriatr Soc. 2011;59(4):745-753. PubMed
24. Cost drivers for dually eligible beneficiaries: Potentially avoidable hospitalizations from nursing facility, skilled nursing facility, and home and community based service waiver programs. http://www.cms.gov/Research-Statistics-Data-and-Systems/Statistics-Trends-and-Reports/Reports/downloads/costdriverstask2.pdf. Accessed August 31, 2016.
25. H.R. 4302 (113th), Section 215, Protecting Access to Medicare Act of 2014 (PAMA). April 2, 2014. https://www.govtrack.us/congress/bills/113/hr4302/text. Accessed August 31, 2016.

Issue
Journal of Hospital Medicine 12(4)
Issue
Journal of Hospital Medicine 12(4)
Page Number
238-244
Page Number
238-244
Topics
Article Type
Display Headline
Impact of a Connected Care model on 30-day readmission rates from skilled nursing facilities
Display Headline
Impact of a Connected Care model on 30-day readmission rates from skilled nursing facilities
Sections
Article Source

© 2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Luke D. Kim, MD, Center for Geriatric Medicine, Medicine Institute, Cleveland Clinic, 9500 Euclid Ave X10, Cleveland, OH 44195; Telephone: 216-444-6092; Fax: 216-445-8762; E-mail: kiml2@ccf.org
Content Gating
Gated (full article locked unless allowed per User)
Alternative CME
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Prognosticating with the hospitalized-patient one-year mortality risk score using information abstracted from the medical record

Article Type
Changed
Wed, 04/26/2017 - 13:42
Display Headline
Prognosticating with the hospitalized-patient one-year mortality risk score using information abstracted from the medical record

A patient’s prognosis can strongly influence their medical care. Decisions about diagnostic modalities, treatment options, and the use of preventive therapies can all be affected by the likelihood of a patient’s death in the near future. For example, patients with severely limited survival might forego prophylactic therapy, avoid interventions for asymptomatic issues, and cease screening interventions. Knowing survival probability would also be very helpful as a controlling variable in research analyses whenever death risk might be a possible confounder.

Sixteen indices that aim to predict patient death risk have been described by Yourman et al.1 They were all created from secondary analyses of clinical and administrative datasets, were applicable to patients in a variety of settings (including the community, nursing home, or hospital), and predicted survival probabilities in time horizons ranging from 6 months to 5 years. Prognostic factors that were most commonly included in these indices were comorbidity and functional status. In validation populations, the discrimination of these indices for 1-year survival in hospitalized patients was moderate (with C statistics that ranged from 0.64 to 0.79) with good calibration for broad prognostic ranges.

In 2014, we published the Hospitalized-patient One-year Mortality Risk (HOMR) score.2 This study used health administrative data for all adult Ontarians admitted in 2011 to hospital under nonpsychiatric services (n = 640,022) to estimate the probability of dying within 1 year of admission to hospital (which happened in 11.7% of people). The HOMR score included 12 patient and hospitalization factors (Table 1). It was highly discriminative (C statistic, 0.923; [0.922-0.924]) and well calibrated (the mean relative difference between observed and expected death risk was 2.0% [range, 0.0% to 7.0%]). It was externally validated in more than 3 million adults from Ontario, Alberta, and Boston in whom the C statistic ranged from 0.89 to 0.92 and calibration was excellent.3 We concluded from these studies that the HOMR score is excellent for prognosticating a diverse group of patients using health administrative data.

HOMR Scoring System
Table 1


However, we do not know whether the HOMR score can be applied to patients using primary data (ie, those taken directly from the chart). This question is important for 2 reasons. First, if HOMR accurately predicts death risk using data abstracted from the medical record, it could be used in the clinical setting to assist in clinical decision-making. Second, HOMR uses multiple administrative datasets that are difficult to access and use by most clinical researchers; it is, therefore, important to determine if HOMR is accurate for clinical research based on primary medical record review. The primary objective of this study was to determine the accuracy of the HOMR score when calculated using data abstracted from clinical notes that were available when patients were admitted to hospital. Secondary objectives included determining whether functional measures abstracted were significantly associated with death risk beyond the HOMR score and whether HOMR scores calculated from chart review deviated from those calculated from administrative data.

 

 

METHODS

Study Cohort

The study, which was approved by our local research ethics board, took place at the Ottawa Hospital, a 1000-bed teaching hospital that is the primary referral center in our region. We used the hospital admission registry to identify all people 18 years or older who were admitted to a nonpsychiatric service at our hospital between January 1, 2011 and December 31, 2011 (this time frame corresponds with the year used to derive the HOMR score). We excluded overnight patients in the same-day surgery or the bone-marrow transplant units (since they would not have been included in the original study) and those without a valid health card number (which was required to link to provincial data to identify outcomes). From this list, we randomly selected 5000 patients.

Primary Data Collection

For each patient, we retrieved all data required to calculate the HOMR score from the medical record (Table 1). Patient registration information in our electronic medical record was used to identify patient age, sex, admitting service, number of emergency department (ED) visits in the previous year, number of admissions in the previous year (the nursing triage note was reviewed for each admission to determine if it was by ambulance), and whether or not the patient had been discharged from hospital in the previous 30 days. The admitting service consult note was used to determine the admitting diagnosis and whether or not the patient was admitted directly to the intensive care unit. If they were present, the emergency nursing triage note, the ED record of treatment, the admission consult note, the pre-operative consult note, and consult notes were all used to determine the patient’s comorbidities, living status, and home oxygen status. Admission urgency was determined using information from the patient registration information and the ED nursing triage note. All data were abstracted from information that had been registered prior to when the patient was physically transferred to their hospital bed. This ensured that we used only data available at the start of the admission.

Patient functional status has been shown to be strongly associated with survival4 but HOMR only indirectly captures functional information (through the patient’s living status). We, therefore, collected more detailed functional information from the medical record by determining if the patient was dependent for any activities of daily living (ADL) from the emergency nursing triage note, the ED record of treatment, the admission consult note, and the pre-operative consultation. We also collected information that might indicate frailty, which we defined per Clegg et al.5 as “a state of increased vulnerability to poor resolution of homeostasis following a stress.” This information included: delirium or more than 1 fall recorded on the emergency nursing triage note, the ED record of treatment, or the admission consultation note; or whether a geriatric nursing specialist assessment occurred in the ED in the previous 6 months. Finally, we recorded possible indicators of limited social support (no fixed address [from patient registration and nursing triage note], primary contact is not a family member [from the emergency notes, consult, and patient registration], and no religion noted in system [from patient registration]). Patients for whom religion status was missing were classified as having “no religion.”

Analysis

These data were encrypted and linked anonymously to population-based databases to determine whether patients died within 1 year of admission to hospital. We calculated the chart-HOMR score using information from the chart review and determined its association with the outcome using bivariate logistic regression. We compared observed and expected risk of death within 1 year of admission to hospital for each chart-HOMR score value, with expected risks determined from the external validation study.3 We regressed observed death risks on expected death risks for chart-HOMR scores (clustered into 22 groups to ensure adequate numbers in each group); and we gauged overall deviations from expected risk and the relationship between the observed and expected death risk (based on the chart-HOMR score) using the line’s intercept and slope, respectively.6 Next, we replicated methods from our studies2,3 to calculate the administrative-HOMR score in our study cohort using administrative databases. We compared these chart-HOMR and administrative-HOMR scores (and scores for each of its components). Finally, we determined which of the socio-functional factors were associated with 1-year death risk independent of the chart-HOMR score. We used the likelihood ratio test to determine whether these additional socio-functional factors significantly improved the model beyond the chart-HOMR score.7 This test subtracted the -2 logL value of the full model from that containing the chart-HOMR score alone, comparing its value to the χ2 distribution (with degrees of freedom equivalent to the number of additional parameters in the nested model) to determine statistical significance. All analyses were completed using SAS v9.4 (SAS Institute Inc., Cary, North Carolina).

 

 

RESULTS

There were 43,883 overnight hospitalizations at our hospital in 2011, and 38,886 hospitalizations were excluded: 1883 hospitalizations were in the same-day surgery or the bone-marrow transplant unit; 2485 did not have a valid health card number; 34,515 were not randomly selected; the records of 3 randomly selected patients had been blocked by our hospital’s privacy department; and 1 patient could not be linked with the population-based administrative datasets.

The 4996 study patients were middle-aged and predominantly female (Table 2). The extensive majority of patients was admitted from the community, was independent for ADL, had a family member as the principal contact, and had no admissions by ambulance in the previous year. Most people had no significant comorbidities or ED visits in the year prior to their admission. The mean chart-HOMR score was 22 (standard deviation [SD], 12), which is associated with a 1.2% expected risk of death within 1 year of hospital admission (Appendix 1).3

Description of Study Cohort by 1-Year Death Status
Table 2


A total of 563 patients (11.3%) died within 1 year of admission to hospital (Table 2). In the study cohort, each chart-HOMR component was associated with death status. People who died were older, more likely to be male, had a greater number of important comorbidities, had more ED visits and admissions by ambulance in the previous year, and were more likely to have been discharged in the previous 30 days, and were admitted urgently, directly to the intensive care unit, or with complicated diagnoses. The mean chart-HOMR score differed extensively by survival status (37.4 [SD, 7.5] in those who died vs. 19.9 [SD, 12.2] in those who survived). Three of the socio-functional variables (delirium and falls noted on admission documents, and dependent for any ADL) also varied with death status.

The chart-HOMR score was strongly associated with the likelihood of death within 1 year of admission. When included in a logistic regression model having 1-year death as the outcome, a 1-point increase in the chart-HOMR score was associated with a 19% increase in the odds of death (P < 0.0001). This model (with only the chart-HOMR score) was highly discriminative (C statistic, 0.888) and well calibrated (Hosmer-Lemeshow test, 12.9 [8 df, P = 0.11]).

Observed and expected death risks by chart-HOMR score were similar (Figure 1). The observed total number of deaths (n = 563; 11.3%) exceeded the expected number of deaths (n = 437, 8.7%). When we regressed observed death risks on expected death risks for chart-HOMR scores (clustered into 22 groups), the Hosmer-Lemeshow test was significant, indicating that differences between observed and expected risks were beyond that expected by chance (Hosmer-Lemeshow test, 141.9, 21 df, P < 0.0001). The intercept of this model (0.035; 95% CI, 0.01-0.06) was statistically significant (P = 0.01), indicating that the observed number of cases significantly exceeded the expected; however, its calibration slope (1.02; 95% CI, 0.89-1.16) did not deviate significantly from unity, indicating that the relationship between the observed and expected death risk (based on the chart-HOMR score) remained intact (Figure 1).

Observed vs expected 1-year death risk
Figure 1


The deviations between observed and expected death risks reflected deviations between the c chart-HOMR score and the administrative-HOMR score, with the former being significantly lower than the latter (Figure 2). Overall, the chart-HOMR score was 0.96 points lower (95% CI, 0.81-1.12) than the administrative-HOMR score. The HOMR score components that were notably underestimated using chart data included those for the age-Charlson Comorbidity Index interaction, living status, and admit points. Points for only 2 components (admitting service and admission urgency) were higher when calculated using chart data.

HOMR-score values using data from medical record review and health administrative databases
Figure 2


Four additional socio-functional variables collected from medical record review were significantly associated with 1-year death risk independent of the chart-HOMR score (Table 3). Admission documentation noting either delirium or falls were both associated with a significantly increased death risk (adjusted odds ratio [OR], 1.92 [95% CI, 1.24-2.96] and OR 1.96 [95% CI, 1.29-2.99], respectively). An independently increased death risk was also noted in patients who were dependent for any ADL (adjusted OR, 1.99 [95% CI, 1.24-3.19]). The presence of an ED geriatrics consultation within the previous 6 months was associated with a significantly decreased death risk of 60% (adjusted OR, 0.40 [95% CI, 0.20-0.81]). Adding these covariates to the logistic model with the chart-HOMR score significantly improved predictions (likelihood ratio statistic = 33.569, 4df, P < 0.00001).

Association of Additional Socio-functional Variables on 1-Year Death Risk
Table 3

DISCUSSION

In a large random sample of patients from our hospital, we found that the HOMR score using data abstracted from the medical record was significantly associated with 1-year death risk. The expected death risk based on the chart-HOMR score underestimated observed death risk but the relationship between the chart-HOMR score and death risk was similar to that in studies using administrative data. The HOMR score calculated using data from the chart was lower than that calculated using data from population-based administrative datasets; additional variables indicating patient frailty were significantly associated with 1-year death risk independent of the chart-HOMR score. Since the HOMR score was derived and initially validated using health administrative data, this study using data abstracted from the health record shows that the HOMR score has methodological generalizability.8

 

 

We think that our study has several notable findings. First, we found that data abstracted from the medical record can be used to calculate the HOMR score to accurately predict individual death risk. The chart-HOMR score discriminated very well between patients who did and did not die (C statistic, 0.88), which extensively exceeds the discrimination of published death risk indices (whose C statistics range between 0.69 and 0.82). It is also possible that chart abstraction for the HOMR score—without functional status—is simpler than other indices since its components are primarily very objective. (Other indices for hospital-based patients required factors that could be difficult to abstract reliably from the medical record including meeting more than 1 guideline for noncancer hospice care9; ambulation difficulties10; scales such as the Exton-Smith Scale or the Short Portable Mental Status Questionnaire11; weight loss12; functional status4; and pressure sore risk.13) Although expected risks for the chart-HOMR consistently underestimated observed risks (Figure 1), the mean deviation was small (with an absolute difference of 3.5% that can be used as a correction factor when determining expected risks with HOMR scores calculated from chart review), but it was an association between the chart-HOMR score and death risk that remained consistent through the cohort. Second, we found a small but significant decrease in the chart-HOMR score vs. the administrative-HOMR score (Figure 2). Some of these underestimates such as those for the number of ED visits or admissions by ambulance were expected since population-based health administrative databases would best capture such data. However, we were surprised that the comorbidity score was less when calculated using chart vs. database data (Figure 2). This finding is distinct from studies finding that particular comorbidities are documented in the chart are sometimes not coded.14,15 However, we identified comorbidities in the administrative databases using a 1-year ‘look-back’ period so that diagnostic codes from multiple hospitalizations (and from multiple hospitals) could be used to calculate the Charlson Comorbidity Index for a particular patient; this has been shown to increase the capture of comorbidities.16 Third, we found that variables from the chart review indicating frailty were predictive of 1-year death risk independent of the chart-HOMR score (Table 2). This illustrates that mortality risk prediction can be improved for particular patient groups by adding new covariates to the HOMR. Further work is required to determine how to incorporate these (and possibly other) covariates into the HOMR to create a unique chart-HOMR score. Finally, we found that a geriatrics assessment in the ED was associated with a significant (and notable) decrease in death risk. With these data, we are unable to indicate whether this association is causative. However, these findings indicate that the influence of emergency geriatric assessments on patient survival needs to be explored in more detail.

Several issues about our study should be considered when interpreting its results. First, this was a single-center study and the generalizability of our results to other centers is unknown. However, our study had the largest sample size of all primary data prognostic index validation studies1 ensuring that our results are, at the very least, internally reliable. In addition, our simple random sample ensured that we studied a broad assortment of patients to be certain that our results are representative of our institution. Second, we used a single abstractor for the study, which could limit the generalizability of our results. However, almost all the data points that were abstracted for our study were very objective.

In summary, our study shows that the HOMR score can be used to accurately predict 1-year death risk using data abstracted from the patient record. These findings will aid in individual patient prognostication for clinicians and researchers.

Disclosure

The authors report no financial conflicts of interest.

 

Files
References

1.   Yourman LC, Lee SJ, Schonberg MA, Widera EW, Smith AK. Prognostic indices for older adults: a systematic review. JAMA. 2012;307(2):182-192. PubMed
 2.   van Walraven C. The Hospital-patient One-year Mortality Risk score accurately predicts long term death risk in hospitalized patients. J Clin Epidemiol. 2014;67(9):1025-1034. PubMed
3.   van Walraven C, McAlister FA, Bakal JA, Hawken S, Donzé J. External validation of the Hospital-patient One-year Mortality Risk (HOMR) model for predicting death within 1 year after hospital admission. CMAJ. 2015;187(10):725-733. PubMed
4.   Walter LC, Brand RJ, Counsell SR, et al. Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization. JAMA. 2001;285(23):2987-2994. PubMed
5.   Clegg A, Young J, Iliffe S et al. Frailty in elderly people. The Lancet 2002;381:752-762. PubMed
6.   Crowson CS, Atkinson EJ, Therneau TM. Assessing calibration of prognostic risk scores. Stat Methods Med Res. 2016;25(4):1692-1706. PubMed
7.   Harrell FE Jr. Overview of Maximum Likelihood Estimation. Regression Modeling Strategies. New York, NY: Springer-Verlag; 2001: 179-212.
8.   Justice AC, Covinsky KE, Berlin JA. Assessing the generalizability of prognostic information. Ann Intern Med. 1999;130(6):515-524. PubMed
9.   Fischer SM, Gozansky WS, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31(4):285-292. PubMed
10.   Inouye SK, Bogardus ST, Jr, Vitagliano G, et al. Burden of illness score for elderly persons: risk adjustment incorporating the cumulative impact of diseases, physiologic abnormalities, and functional impairments.  Med Care. 2003;41(1):70-83. PubMed
11.   Pilotto A, Ferrucci L, Franceschi M, et al. Development and validation of a multidimensional prognostic index for one-year mortality from comprehensive geriatric assessment in hospitalized older patients. Rejuvenation Res. 2008;11(1):151-161. PubMed
12.   Teno JM, Harrell FE Jr, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. Hospitalized Elderly Longitudinal Project. J Am Geriatr Soc. 2000;48(5 suppl):S16-S24. PubMed
13.   Dramé M, Novella JL, Lang PO, et al. Derivation and validation of a mortality-risk index from a cohort of frail elderly patients hospitalised in medical wards via emergencies: the SAFES study. Eur J Epidemiol. 2008;23(12):783-791. PubMed
14.   Kieszak SM, Flanders WD, Kosinski AS, Shipp CC, Karp H. A comparison of the Charlson comorbidity index derived from medical record data and administrative billing data. J Clin Epidemiol. 1999;52(2):137-142. PubMed
15.   Quan H, Parsons GA, Ghali WA. Validity of procedure codes in International Classification of Diseases, 9th revision, clinical modification administrative data. Med Care. 2004;42(8):801-809. PubMed
16.   Zhang JX, Iwashyna TJ, Christakis NA. The performance of different lookback periods and sources of information for Charlson cComorbidity adjustment in Medicare claims. Med Care. 1999;37(11):1128-1139. PubMed

Article PDF
Issue
Journal of Hospital Medicine 12(4)
Topics
Page Number
224-230
Sections
Files
Files
Article PDF
Article PDF

A patient’s prognosis can strongly influence their medical care. Decisions about diagnostic modalities, treatment options, and the use of preventive therapies can all be affected by the likelihood of a patient’s death in the near future. For example, patients with severely limited survival might forego prophylactic therapy, avoid interventions for asymptomatic issues, and cease screening interventions. Knowing survival probability would also be very helpful as a controlling variable in research analyses whenever death risk might be a possible confounder.

Sixteen indices that aim to predict patient death risk have been described by Yourman et al.1 They were all created from secondary analyses of clinical and administrative datasets, were applicable to patients in a variety of settings (including the community, nursing home, or hospital), and predicted survival probabilities in time horizons ranging from 6 months to 5 years. Prognostic factors that were most commonly included in these indices were comorbidity and functional status. In validation populations, the discrimination of these indices for 1-year survival in hospitalized patients was moderate (with C statistics that ranged from 0.64 to 0.79) with good calibration for broad prognostic ranges.

In 2014, we published the Hospitalized-patient One-year Mortality Risk (HOMR) score.2 This study used health administrative data for all adult Ontarians admitted in 2011 to hospital under nonpsychiatric services (n = 640,022) to estimate the probability of dying within 1 year of admission to hospital (which happened in 11.7% of people). The HOMR score included 12 patient and hospitalization factors (Table 1). It was highly discriminative (C statistic, 0.923; [0.922-0.924]) and well calibrated (the mean relative difference between observed and expected death risk was 2.0% [range, 0.0% to 7.0%]). It was externally validated in more than 3 million adults from Ontario, Alberta, and Boston in whom the C statistic ranged from 0.89 to 0.92 and calibration was excellent.3 We concluded from these studies that the HOMR score is excellent for prognosticating a diverse group of patients using health administrative data.

HOMR Scoring System
Table 1


However, we do not know whether the HOMR score can be applied to patients using primary data (ie, those taken directly from the chart). This question is important for 2 reasons. First, if HOMR accurately predicts death risk using data abstracted from the medical record, it could be used in the clinical setting to assist in clinical decision-making. Second, HOMR uses multiple administrative datasets that are difficult to access and use by most clinical researchers; it is, therefore, important to determine if HOMR is accurate for clinical research based on primary medical record review. The primary objective of this study was to determine the accuracy of the HOMR score when calculated using data abstracted from clinical notes that were available when patients were admitted to hospital. Secondary objectives included determining whether functional measures abstracted were significantly associated with death risk beyond the HOMR score and whether HOMR scores calculated from chart review deviated from those calculated from administrative data.

 

 

METHODS

Study Cohort

The study, which was approved by our local research ethics board, took place at the Ottawa Hospital, a 1000-bed teaching hospital that is the primary referral center in our region. We used the hospital admission registry to identify all people 18 years or older who were admitted to a nonpsychiatric service at our hospital between January 1, 2011 and December 31, 2011 (this time frame corresponds with the year used to derive the HOMR score). We excluded overnight patients in the same-day surgery or the bone-marrow transplant units (since they would not have been included in the original study) and those without a valid health card number (which was required to link to provincial data to identify outcomes). From this list, we randomly selected 5000 patients.

Primary Data Collection

For each patient, we retrieved all data required to calculate the HOMR score from the medical record (Table 1). Patient registration information in our electronic medical record was used to identify patient age, sex, admitting service, number of emergency department (ED) visits in the previous year, number of admissions in the previous year (the nursing triage note was reviewed for each admission to determine if it was by ambulance), and whether or not the patient had been discharged from hospital in the previous 30 days. The admitting service consult note was used to determine the admitting diagnosis and whether or not the patient was admitted directly to the intensive care unit. If they were present, the emergency nursing triage note, the ED record of treatment, the admission consult note, the pre-operative consult note, and consult notes were all used to determine the patient’s comorbidities, living status, and home oxygen status. Admission urgency was determined using information from the patient registration information and the ED nursing triage note. All data were abstracted from information that had been registered prior to when the patient was physically transferred to their hospital bed. This ensured that we used only data available at the start of the admission.

Patient functional status has been shown to be strongly associated with survival4 but HOMR only indirectly captures functional information (through the patient’s living status). We, therefore, collected more detailed functional information from the medical record by determining if the patient was dependent for any activities of daily living (ADL) from the emergency nursing triage note, the ED record of treatment, the admission consult note, and the pre-operative consultation. We also collected information that might indicate frailty, which we defined per Clegg et al.5 as “a state of increased vulnerability to poor resolution of homeostasis following a stress.” This information included: delirium or more than 1 fall recorded on the emergency nursing triage note, the ED record of treatment, or the admission consultation note; or whether a geriatric nursing specialist assessment occurred in the ED in the previous 6 months. Finally, we recorded possible indicators of limited social support (no fixed address [from patient registration and nursing triage note], primary contact is not a family member [from the emergency notes, consult, and patient registration], and no religion noted in system [from patient registration]). Patients for whom religion status was missing were classified as having “no religion.”

Analysis

These data were encrypted and linked anonymously to population-based databases to determine whether patients died within 1 year of admission to hospital. We calculated the chart-HOMR score using information from the chart review and determined its association with the outcome using bivariate logistic regression. We compared observed and expected risk of death within 1 year of admission to hospital for each chart-HOMR score value, with expected risks determined from the external validation study.3 We regressed observed death risks on expected death risks for chart-HOMR scores (clustered into 22 groups to ensure adequate numbers in each group); and we gauged overall deviations from expected risk and the relationship between the observed and expected death risk (based on the chart-HOMR score) using the line’s intercept and slope, respectively.6 Next, we replicated methods from our studies2,3 to calculate the administrative-HOMR score in our study cohort using administrative databases. We compared these chart-HOMR and administrative-HOMR scores (and scores for each of its components). Finally, we determined which of the socio-functional factors were associated with 1-year death risk independent of the chart-HOMR score. We used the likelihood ratio test to determine whether these additional socio-functional factors significantly improved the model beyond the chart-HOMR score.7 This test subtracted the -2 logL value of the full model from that containing the chart-HOMR score alone, comparing its value to the χ2 distribution (with degrees of freedom equivalent to the number of additional parameters in the nested model) to determine statistical significance. All analyses were completed using SAS v9.4 (SAS Institute Inc., Cary, North Carolina).

 

 

RESULTS

There were 43,883 overnight hospitalizations at our hospital in 2011, and 38,886 hospitalizations were excluded: 1883 hospitalizations were in the same-day surgery or the bone-marrow transplant unit; 2485 did not have a valid health card number; 34,515 were not randomly selected; the records of 3 randomly selected patients had been blocked by our hospital’s privacy department; and 1 patient could not be linked with the population-based administrative datasets.

The 4996 study patients were middle-aged and predominantly female (Table 2). The extensive majority of patients was admitted from the community, was independent for ADL, had a family member as the principal contact, and had no admissions by ambulance in the previous year. Most people had no significant comorbidities or ED visits in the year prior to their admission. The mean chart-HOMR score was 22 (standard deviation [SD], 12), which is associated with a 1.2% expected risk of death within 1 year of hospital admission (Appendix 1).3

Description of Study Cohort by 1-Year Death Status
Table 2


A total of 563 patients (11.3%) died within 1 year of admission to hospital (Table 2). In the study cohort, each chart-HOMR component was associated with death status. People who died were older, more likely to be male, had a greater number of important comorbidities, had more ED visits and admissions by ambulance in the previous year, and were more likely to have been discharged in the previous 30 days, and were admitted urgently, directly to the intensive care unit, or with complicated diagnoses. The mean chart-HOMR score differed extensively by survival status (37.4 [SD, 7.5] in those who died vs. 19.9 [SD, 12.2] in those who survived). Three of the socio-functional variables (delirium and falls noted on admission documents, and dependent for any ADL) also varied with death status.

The chart-HOMR score was strongly associated with the likelihood of death within 1 year of admission. When included in a logistic regression model having 1-year death as the outcome, a 1-point increase in the chart-HOMR score was associated with a 19% increase in the odds of death (P < 0.0001). This model (with only the chart-HOMR score) was highly discriminative (C statistic, 0.888) and well calibrated (Hosmer-Lemeshow test, 12.9 [8 df, P = 0.11]).

Observed and expected death risks by chart-HOMR score were similar (Figure 1). The observed total number of deaths (n = 563; 11.3%) exceeded the expected number of deaths (n = 437, 8.7%). When we regressed observed death risks on expected death risks for chart-HOMR scores (clustered into 22 groups), the Hosmer-Lemeshow test was significant, indicating that differences between observed and expected risks were beyond that expected by chance (Hosmer-Lemeshow test, 141.9, 21 df, P < 0.0001). The intercept of this model (0.035; 95% CI, 0.01-0.06) was statistically significant (P = 0.01), indicating that the observed number of cases significantly exceeded the expected; however, its calibration slope (1.02; 95% CI, 0.89-1.16) did not deviate significantly from unity, indicating that the relationship between the observed and expected death risk (based on the chart-HOMR score) remained intact (Figure 1).

Observed vs expected 1-year death risk
Figure 1


The deviations between observed and expected death risks reflected deviations between the c chart-HOMR score and the administrative-HOMR score, with the former being significantly lower than the latter (Figure 2). Overall, the chart-HOMR score was 0.96 points lower (95% CI, 0.81-1.12) than the administrative-HOMR score. The HOMR score components that were notably underestimated using chart data included those for the age-Charlson Comorbidity Index interaction, living status, and admit points. Points for only 2 components (admitting service and admission urgency) were higher when calculated using chart data.

HOMR-score values using data from medical record review and health administrative databases
Figure 2


Four additional socio-functional variables collected from medical record review were significantly associated with 1-year death risk independent of the chart-HOMR score (Table 3). Admission documentation noting either delirium or falls were both associated with a significantly increased death risk (adjusted odds ratio [OR], 1.92 [95% CI, 1.24-2.96] and OR 1.96 [95% CI, 1.29-2.99], respectively). An independently increased death risk was also noted in patients who were dependent for any ADL (adjusted OR, 1.99 [95% CI, 1.24-3.19]). The presence of an ED geriatrics consultation within the previous 6 months was associated with a significantly decreased death risk of 60% (adjusted OR, 0.40 [95% CI, 0.20-0.81]). Adding these covariates to the logistic model with the chart-HOMR score significantly improved predictions (likelihood ratio statistic = 33.569, 4df, P < 0.00001).

Association of Additional Socio-functional Variables on 1-Year Death Risk
Table 3

DISCUSSION

In a large random sample of patients from our hospital, we found that the HOMR score using data abstracted from the medical record was significantly associated with 1-year death risk. The expected death risk based on the chart-HOMR score underestimated observed death risk but the relationship between the chart-HOMR score and death risk was similar to that in studies using administrative data. The HOMR score calculated using data from the chart was lower than that calculated using data from population-based administrative datasets; additional variables indicating patient frailty were significantly associated with 1-year death risk independent of the chart-HOMR score. Since the HOMR score was derived and initially validated using health administrative data, this study using data abstracted from the health record shows that the HOMR score has methodological generalizability.8

 

 

We think that our study has several notable findings. First, we found that data abstracted from the medical record can be used to calculate the HOMR score to accurately predict individual death risk. The chart-HOMR score discriminated very well between patients who did and did not die (C statistic, 0.88), which extensively exceeds the discrimination of published death risk indices (whose C statistics range between 0.69 and 0.82). It is also possible that chart abstraction for the HOMR score—without functional status—is simpler than other indices since its components are primarily very objective. (Other indices for hospital-based patients required factors that could be difficult to abstract reliably from the medical record including meeting more than 1 guideline for noncancer hospice care9; ambulation difficulties10; scales such as the Exton-Smith Scale or the Short Portable Mental Status Questionnaire11; weight loss12; functional status4; and pressure sore risk.13) Although expected risks for the chart-HOMR consistently underestimated observed risks (Figure 1), the mean deviation was small (with an absolute difference of 3.5% that can be used as a correction factor when determining expected risks with HOMR scores calculated from chart review), but it was an association between the chart-HOMR score and death risk that remained consistent through the cohort. Second, we found a small but significant decrease in the chart-HOMR score vs. the administrative-HOMR score (Figure 2). Some of these underestimates such as those for the number of ED visits or admissions by ambulance were expected since population-based health administrative databases would best capture such data. However, we were surprised that the comorbidity score was less when calculated using chart vs. database data (Figure 2). This finding is distinct from studies finding that particular comorbidities are documented in the chart are sometimes not coded.14,15 However, we identified comorbidities in the administrative databases using a 1-year ‘look-back’ period so that diagnostic codes from multiple hospitalizations (and from multiple hospitals) could be used to calculate the Charlson Comorbidity Index for a particular patient; this has been shown to increase the capture of comorbidities.16 Third, we found that variables from the chart review indicating frailty were predictive of 1-year death risk independent of the chart-HOMR score (Table 2). This illustrates that mortality risk prediction can be improved for particular patient groups by adding new covariates to the HOMR. Further work is required to determine how to incorporate these (and possibly other) covariates into the HOMR to create a unique chart-HOMR score. Finally, we found that a geriatrics assessment in the ED was associated with a significant (and notable) decrease in death risk. With these data, we are unable to indicate whether this association is causative. However, these findings indicate that the influence of emergency geriatric assessments on patient survival needs to be explored in more detail.

Several issues about our study should be considered when interpreting its results. First, this was a single-center study and the generalizability of our results to other centers is unknown. However, our study had the largest sample size of all primary data prognostic index validation studies1 ensuring that our results are, at the very least, internally reliable. In addition, our simple random sample ensured that we studied a broad assortment of patients to be certain that our results are representative of our institution. Second, we used a single abstractor for the study, which could limit the generalizability of our results. However, almost all the data points that were abstracted for our study were very objective.

In summary, our study shows that the HOMR score can be used to accurately predict 1-year death risk using data abstracted from the patient record. These findings will aid in individual patient prognostication for clinicians and researchers.

Disclosure

The authors report no financial conflicts of interest.

 

A patient’s prognosis can strongly influence their medical care. Decisions about diagnostic modalities, treatment options, and the use of preventive therapies can all be affected by the likelihood of a patient’s death in the near future. For example, patients with severely limited survival might forego prophylactic therapy, avoid interventions for asymptomatic issues, and cease screening interventions. Knowing survival probability would also be very helpful as a controlling variable in research analyses whenever death risk might be a possible confounder.

Sixteen indices that aim to predict patient death risk have been described by Yourman et al.1 They were all created from secondary analyses of clinical and administrative datasets, were applicable to patients in a variety of settings (including the community, nursing home, or hospital), and predicted survival probabilities in time horizons ranging from 6 months to 5 years. Prognostic factors that were most commonly included in these indices were comorbidity and functional status. In validation populations, the discrimination of these indices for 1-year survival in hospitalized patients was moderate (with C statistics that ranged from 0.64 to 0.79) with good calibration for broad prognostic ranges.

In 2014, we published the Hospitalized-patient One-year Mortality Risk (HOMR) score.2 This study used health administrative data for all adult Ontarians admitted in 2011 to hospital under nonpsychiatric services (n = 640,022) to estimate the probability of dying within 1 year of admission to hospital (which happened in 11.7% of people). The HOMR score included 12 patient and hospitalization factors (Table 1). It was highly discriminative (C statistic, 0.923; [0.922-0.924]) and well calibrated (the mean relative difference between observed and expected death risk was 2.0% [range, 0.0% to 7.0%]). It was externally validated in more than 3 million adults from Ontario, Alberta, and Boston in whom the C statistic ranged from 0.89 to 0.92 and calibration was excellent.3 We concluded from these studies that the HOMR score is excellent for prognosticating a diverse group of patients using health administrative data.

HOMR Scoring System
Table 1


However, we do not know whether the HOMR score can be applied to patients using primary data (ie, those taken directly from the chart). This question is important for 2 reasons. First, if HOMR accurately predicts death risk using data abstracted from the medical record, it could be used in the clinical setting to assist in clinical decision-making. Second, HOMR uses multiple administrative datasets that are difficult to access and use by most clinical researchers; it is, therefore, important to determine if HOMR is accurate for clinical research based on primary medical record review. The primary objective of this study was to determine the accuracy of the HOMR score when calculated using data abstracted from clinical notes that were available when patients were admitted to hospital. Secondary objectives included determining whether functional measures abstracted were significantly associated with death risk beyond the HOMR score and whether HOMR scores calculated from chart review deviated from those calculated from administrative data.

 

 

METHODS

Study Cohort

The study, which was approved by our local research ethics board, took place at the Ottawa Hospital, a 1000-bed teaching hospital that is the primary referral center in our region. We used the hospital admission registry to identify all people 18 years or older who were admitted to a nonpsychiatric service at our hospital between January 1, 2011 and December 31, 2011 (this time frame corresponds with the year used to derive the HOMR score). We excluded overnight patients in the same-day surgery or the bone-marrow transplant units (since they would not have been included in the original study) and those without a valid health card number (which was required to link to provincial data to identify outcomes). From this list, we randomly selected 5000 patients.

Primary Data Collection

For each patient, we retrieved all data required to calculate the HOMR score from the medical record (Table 1). Patient registration information in our electronic medical record was used to identify patient age, sex, admitting service, number of emergency department (ED) visits in the previous year, number of admissions in the previous year (the nursing triage note was reviewed for each admission to determine if it was by ambulance), and whether or not the patient had been discharged from hospital in the previous 30 days. The admitting service consult note was used to determine the admitting diagnosis and whether or not the patient was admitted directly to the intensive care unit. If they were present, the emergency nursing triage note, the ED record of treatment, the admission consult note, the pre-operative consult note, and consult notes were all used to determine the patient’s comorbidities, living status, and home oxygen status. Admission urgency was determined using information from the patient registration information and the ED nursing triage note. All data were abstracted from information that had been registered prior to when the patient was physically transferred to their hospital bed. This ensured that we used only data available at the start of the admission.

Patient functional status has been shown to be strongly associated with survival4 but HOMR only indirectly captures functional information (through the patient’s living status). We, therefore, collected more detailed functional information from the medical record by determining if the patient was dependent for any activities of daily living (ADL) from the emergency nursing triage note, the ED record of treatment, the admission consult note, and the pre-operative consultation. We also collected information that might indicate frailty, which we defined per Clegg et al.5 as “a state of increased vulnerability to poor resolution of homeostasis following a stress.” This information included: delirium or more than 1 fall recorded on the emergency nursing triage note, the ED record of treatment, or the admission consultation note; or whether a geriatric nursing specialist assessment occurred in the ED in the previous 6 months. Finally, we recorded possible indicators of limited social support (no fixed address [from patient registration and nursing triage note], primary contact is not a family member [from the emergency notes, consult, and patient registration], and no religion noted in system [from patient registration]). Patients for whom religion status was missing were classified as having “no religion.”

Analysis

These data were encrypted and linked anonymously to population-based databases to determine whether patients died within 1 year of admission to hospital. We calculated the chart-HOMR score using information from the chart review and determined its association with the outcome using bivariate logistic regression. We compared observed and expected risk of death within 1 year of admission to hospital for each chart-HOMR score value, with expected risks determined from the external validation study.3 We regressed observed death risks on expected death risks for chart-HOMR scores (clustered into 22 groups to ensure adequate numbers in each group); and we gauged overall deviations from expected risk and the relationship between the observed and expected death risk (based on the chart-HOMR score) using the line’s intercept and slope, respectively.6 Next, we replicated methods from our studies2,3 to calculate the administrative-HOMR score in our study cohort using administrative databases. We compared these chart-HOMR and administrative-HOMR scores (and scores for each of its components). Finally, we determined which of the socio-functional factors were associated with 1-year death risk independent of the chart-HOMR score. We used the likelihood ratio test to determine whether these additional socio-functional factors significantly improved the model beyond the chart-HOMR score.7 This test subtracted the -2 logL value of the full model from that containing the chart-HOMR score alone, comparing its value to the χ2 distribution (with degrees of freedom equivalent to the number of additional parameters in the nested model) to determine statistical significance. All analyses were completed using SAS v9.4 (SAS Institute Inc., Cary, North Carolina).

 

 

RESULTS

There were 43,883 overnight hospitalizations at our hospital in 2011, and 38,886 hospitalizations were excluded: 1883 hospitalizations were in the same-day surgery or the bone-marrow transplant unit; 2485 did not have a valid health card number; 34,515 were not randomly selected; the records of 3 randomly selected patients had been blocked by our hospital’s privacy department; and 1 patient could not be linked with the population-based administrative datasets.

The 4996 study patients were middle-aged and predominantly female (Table 2). The extensive majority of patients was admitted from the community, was independent for ADL, had a family member as the principal contact, and had no admissions by ambulance in the previous year. Most people had no significant comorbidities or ED visits in the year prior to their admission. The mean chart-HOMR score was 22 (standard deviation [SD], 12), which is associated with a 1.2% expected risk of death within 1 year of hospital admission (Appendix 1).3

Description of Study Cohort by 1-Year Death Status
Table 2


A total of 563 patients (11.3%) died within 1 year of admission to hospital (Table 2). In the study cohort, each chart-HOMR component was associated with death status. People who died were older, more likely to be male, had a greater number of important comorbidities, had more ED visits and admissions by ambulance in the previous year, and were more likely to have been discharged in the previous 30 days, and were admitted urgently, directly to the intensive care unit, or with complicated diagnoses. The mean chart-HOMR score differed extensively by survival status (37.4 [SD, 7.5] in those who died vs. 19.9 [SD, 12.2] in those who survived). Three of the socio-functional variables (delirium and falls noted on admission documents, and dependent for any ADL) also varied with death status.

The chart-HOMR score was strongly associated with the likelihood of death within 1 year of admission. When included in a logistic regression model having 1-year death as the outcome, a 1-point increase in the chart-HOMR score was associated with a 19% increase in the odds of death (P < 0.0001). This model (with only the chart-HOMR score) was highly discriminative (C statistic, 0.888) and well calibrated (Hosmer-Lemeshow test, 12.9 [8 df, P = 0.11]).

Observed and expected death risks by chart-HOMR score were similar (Figure 1). The observed total number of deaths (n = 563; 11.3%) exceeded the expected number of deaths (n = 437, 8.7%). When we regressed observed death risks on expected death risks for chart-HOMR scores (clustered into 22 groups), the Hosmer-Lemeshow test was significant, indicating that differences between observed and expected risks were beyond that expected by chance (Hosmer-Lemeshow test, 141.9, 21 df, P < 0.0001). The intercept of this model (0.035; 95% CI, 0.01-0.06) was statistically significant (P = 0.01), indicating that the observed number of cases significantly exceeded the expected; however, its calibration slope (1.02; 95% CI, 0.89-1.16) did not deviate significantly from unity, indicating that the relationship between the observed and expected death risk (based on the chart-HOMR score) remained intact (Figure 1).

Observed vs expected 1-year death risk
Figure 1


The deviations between observed and expected death risks reflected deviations between the c chart-HOMR score and the administrative-HOMR score, with the former being significantly lower than the latter (Figure 2). Overall, the chart-HOMR score was 0.96 points lower (95% CI, 0.81-1.12) than the administrative-HOMR score. The HOMR score components that were notably underestimated using chart data included those for the age-Charlson Comorbidity Index interaction, living status, and admit points. Points for only 2 components (admitting service and admission urgency) were higher when calculated using chart data.

HOMR-score values using data from medical record review and health administrative databases
Figure 2


Four additional socio-functional variables collected from medical record review were significantly associated with 1-year death risk independent of the chart-HOMR score (Table 3). Admission documentation noting either delirium or falls were both associated with a significantly increased death risk (adjusted odds ratio [OR], 1.92 [95% CI, 1.24-2.96] and OR 1.96 [95% CI, 1.29-2.99], respectively). An independently increased death risk was also noted in patients who were dependent for any ADL (adjusted OR, 1.99 [95% CI, 1.24-3.19]). The presence of an ED geriatrics consultation within the previous 6 months was associated with a significantly decreased death risk of 60% (adjusted OR, 0.40 [95% CI, 0.20-0.81]). Adding these covariates to the logistic model with the chart-HOMR score significantly improved predictions (likelihood ratio statistic = 33.569, 4df, P < 0.00001).

Association of Additional Socio-functional Variables on 1-Year Death Risk
Table 3

DISCUSSION

In a large random sample of patients from our hospital, we found that the HOMR score using data abstracted from the medical record was significantly associated with 1-year death risk. The expected death risk based on the chart-HOMR score underestimated observed death risk but the relationship between the chart-HOMR score and death risk was similar to that in studies using administrative data. The HOMR score calculated using data from the chart was lower than that calculated using data from population-based administrative datasets; additional variables indicating patient frailty were significantly associated with 1-year death risk independent of the chart-HOMR score. Since the HOMR score was derived and initially validated using health administrative data, this study using data abstracted from the health record shows that the HOMR score has methodological generalizability.8

 

 

We think that our study has several notable findings. First, we found that data abstracted from the medical record can be used to calculate the HOMR score to accurately predict individual death risk. The chart-HOMR score discriminated very well between patients who did and did not die (C statistic, 0.88), which extensively exceeds the discrimination of published death risk indices (whose C statistics range between 0.69 and 0.82). It is also possible that chart abstraction for the HOMR score—without functional status—is simpler than other indices since its components are primarily very objective. (Other indices for hospital-based patients required factors that could be difficult to abstract reliably from the medical record including meeting more than 1 guideline for noncancer hospice care9; ambulation difficulties10; scales such as the Exton-Smith Scale or the Short Portable Mental Status Questionnaire11; weight loss12; functional status4; and pressure sore risk.13) Although expected risks for the chart-HOMR consistently underestimated observed risks (Figure 1), the mean deviation was small (with an absolute difference of 3.5% that can be used as a correction factor when determining expected risks with HOMR scores calculated from chart review), but it was an association between the chart-HOMR score and death risk that remained consistent through the cohort. Second, we found a small but significant decrease in the chart-HOMR score vs. the administrative-HOMR score (Figure 2). Some of these underestimates such as those for the number of ED visits or admissions by ambulance were expected since population-based health administrative databases would best capture such data. However, we were surprised that the comorbidity score was less when calculated using chart vs. database data (Figure 2). This finding is distinct from studies finding that particular comorbidities are documented in the chart are sometimes not coded.14,15 However, we identified comorbidities in the administrative databases using a 1-year ‘look-back’ period so that diagnostic codes from multiple hospitalizations (and from multiple hospitals) could be used to calculate the Charlson Comorbidity Index for a particular patient; this has been shown to increase the capture of comorbidities.16 Third, we found that variables from the chart review indicating frailty were predictive of 1-year death risk independent of the chart-HOMR score (Table 2). This illustrates that mortality risk prediction can be improved for particular patient groups by adding new covariates to the HOMR. Further work is required to determine how to incorporate these (and possibly other) covariates into the HOMR to create a unique chart-HOMR score. Finally, we found that a geriatrics assessment in the ED was associated with a significant (and notable) decrease in death risk. With these data, we are unable to indicate whether this association is causative. However, these findings indicate that the influence of emergency geriatric assessments on patient survival needs to be explored in more detail.

Several issues about our study should be considered when interpreting its results. First, this was a single-center study and the generalizability of our results to other centers is unknown. However, our study had the largest sample size of all primary data prognostic index validation studies1 ensuring that our results are, at the very least, internally reliable. In addition, our simple random sample ensured that we studied a broad assortment of patients to be certain that our results are representative of our institution. Second, we used a single abstractor for the study, which could limit the generalizability of our results. However, almost all the data points that were abstracted for our study were very objective.

In summary, our study shows that the HOMR score can be used to accurately predict 1-year death risk using data abstracted from the patient record. These findings will aid in individual patient prognostication for clinicians and researchers.

Disclosure

The authors report no financial conflicts of interest.

 

References

1.   Yourman LC, Lee SJ, Schonberg MA, Widera EW, Smith AK. Prognostic indices for older adults: a systematic review. JAMA. 2012;307(2):182-192. PubMed
 2.   van Walraven C. The Hospital-patient One-year Mortality Risk score accurately predicts long term death risk in hospitalized patients. J Clin Epidemiol. 2014;67(9):1025-1034. PubMed
3.   van Walraven C, McAlister FA, Bakal JA, Hawken S, Donzé J. External validation of the Hospital-patient One-year Mortality Risk (HOMR) model for predicting death within 1 year after hospital admission. CMAJ. 2015;187(10):725-733. PubMed
4.   Walter LC, Brand RJ, Counsell SR, et al. Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization. JAMA. 2001;285(23):2987-2994. PubMed
5.   Clegg A, Young J, Iliffe S et al. Frailty in elderly people. The Lancet 2002;381:752-762. PubMed
6.   Crowson CS, Atkinson EJ, Therneau TM. Assessing calibration of prognostic risk scores. Stat Methods Med Res. 2016;25(4):1692-1706. PubMed
7.   Harrell FE Jr. Overview of Maximum Likelihood Estimation. Regression Modeling Strategies. New York, NY: Springer-Verlag; 2001: 179-212.
8.   Justice AC, Covinsky KE, Berlin JA. Assessing the generalizability of prognostic information. Ann Intern Med. 1999;130(6):515-524. PubMed
9.   Fischer SM, Gozansky WS, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31(4):285-292. PubMed
10.   Inouye SK, Bogardus ST, Jr, Vitagliano G, et al. Burden of illness score for elderly persons: risk adjustment incorporating the cumulative impact of diseases, physiologic abnormalities, and functional impairments.  Med Care. 2003;41(1):70-83. PubMed
11.   Pilotto A, Ferrucci L, Franceschi M, et al. Development and validation of a multidimensional prognostic index for one-year mortality from comprehensive geriatric assessment in hospitalized older patients. Rejuvenation Res. 2008;11(1):151-161. PubMed
12.   Teno JM, Harrell FE Jr, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. Hospitalized Elderly Longitudinal Project. J Am Geriatr Soc. 2000;48(5 suppl):S16-S24. PubMed
13.   Dramé M, Novella JL, Lang PO, et al. Derivation and validation of a mortality-risk index from a cohort of frail elderly patients hospitalised in medical wards via emergencies: the SAFES study. Eur J Epidemiol. 2008;23(12):783-791. PubMed
14.   Kieszak SM, Flanders WD, Kosinski AS, Shipp CC, Karp H. A comparison of the Charlson comorbidity index derived from medical record data and administrative billing data. J Clin Epidemiol. 1999;52(2):137-142. PubMed
15.   Quan H, Parsons GA, Ghali WA. Validity of procedure codes in International Classification of Diseases, 9th revision, clinical modification administrative data. Med Care. 2004;42(8):801-809. PubMed
16.   Zhang JX, Iwashyna TJ, Christakis NA. The performance of different lookback periods and sources of information for Charlson cComorbidity adjustment in Medicare claims. Med Care. 1999;37(11):1128-1139. PubMed

References

1.   Yourman LC, Lee SJ, Schonberg MA, Widera EW, Smith AK. Prognostic indices for older adults: a systematic review. JAMA. 2012;307(2):182-192. PubMed
 2.   van Walraven C. The Hospital-patient One-year Mortality Risk score accurately predicts long term death risk in hospitalized patients. J Clin Epidemiol. 2014;67(9):1025-1034. PubMed
3.   van Walraven C, McAlister FA, Bakal JA, Hawken S, Donzé J. External validation of the Hospital-patient One-year Mortality Risk (HOMR) model for predicting death within 1 year after hospital admission. CMAJ. 2015;187(10):725-733. PubMed
4.   Walter LC, Brand RJ, Counsell SR, et al. Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization. JAMA. 2001;285(23):2987-2994. PubMed
5.   Clegg A, Young J, Iliffe S et al. Frailty in elderly people. The Lancet 2002;381:752-762. PubMed
6.   Crowson CS, Atkinson EJ, Therneau TM. Assessing calibration of prognostic risk scores. Stat Methods Med Res. 2016;25(4):1692-1706. PubMed
7.   Harrell FE Jr. Overview of Maximum Likelihood Estimation. Regression Modeling Strategies. New York, NY: Springer-Verlag; 2001: 179-212.
8.   Justice AC, Covinsky KE, Berlin JA. Assessing the generalizability of prognostic information. Ann Intern Med. 1999;130(6):515-524. PubMed
9.   Fischer SM, Gozansky WS, Sauaia A, Min SJ, Kutner JS, Kramer A. A practical tool to identify patients who may benefit from a palliative approach: the CARING criteria. J Pain Symptom Manage. 2006;31(4):285-292. PubMed
10.   Inouye SK, Bogardus ST, Jr, Vitagliano G, et al. Burden of illness score for elderly persons: risk adjustment incorporating the cumulative impact of diseases, physiologic abnormalities, and functional impairments.  Med Care. 2003;41(1):70-83. PubMed
11.   Pilotto A, Ferrucci L, Franceschi M, et al. Development and validation of a multidimensional prognostic index for one-year mortality from comprehensive geriatric assessment in hospitalized older patients. Rejuvenation Res. 2008;11(1):151-161. PubMed
12.   Teno JM, Harrell FE Jr, Knaus W, et al. Prediction of survival for older hospitalized patients: the HELP survival model. Hospitalized Elderly Longitudinal Project. J Am Geriatr Soc. 2000;48(5 suppl):S16-S24. PubMed
13.   Dramé M, Novella JL, Lang PO, et al. Derivation and validation of a mortality-risk index from a cohort of frail elderly patients hospitalised in medical wards via emergencies: the SAFES study. Eur J Epidemiol. 2008;23(12):783-791. PubMed
14.   Kieszak SM, Flanders WD, Kosinski AS, Shipp CC, Karp H. A comparison of the Charlson comorbidity index derived from medical record data and administrative billing data. J Clin Epidemiol. 1999;52(2):137-142. PubMed
15.   Quan H, Parsons GA, Ghali WA. Validity of procedure codes in International Classification of Diseases, 9th revision, clinical modification administrative data. Med Care. 2004;42(8):801-809. PubMed
16.   Zhang JX, Iwashyna TJ, Christakis NA. The performance of different lookback periods and sources of information for Charlson cComorbidity adjustment in Medicare claims. Med Care. 1999;37(11):1128-1139. PubMed

Issue
Journal of Hospital Medicine 12(4)
Issue
Journal of Hospital Medicine 12(4)
Page Number
224-230
Page Number
224-230
Topics
Article Type
Display Headline
Prognosticating with the hospitalized-patient one-year mortality risk score using information abstracted from the medical record
Display Headline
Prognosticating with the hospitalized-patient one-year mortality risk score using information abstracted from the medical record
Sections
Article Source

© 2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for Correspondence and Reprint Requests: Carl van Walraven, MD,ASB1-003 1053 Carling Ave., Ottawa ON; K1Y 4E9; Telephone: 613-761-4903 ; Fax: 613-761-5492 ; E-mail: carlv@ohri.ca
Content Gating
Gated (full article locked unless allowed per User)
Alternative CME
Gating Strategy
First Peek Free
Article PDF Media
Media Files

Predicting 30-day pneumonia readmissions using electronic health record data

Article Type
Changed
Fri, 12/14/2018 - 08:24
Display Headline
Predicting 30-day pneumonia readmissions using electronic health record data

Pneumonia is a leading cause of hospitalizations in the U.S., accounting for more than 1.1 million discharges annually.1 Pneumonia is frequently complicated by hospital readmission, which is costly and potentially avoidable.2,3 Due to financial penalties imposed on hospitals for higher than expected 30-day readmission rates, there is increasing attention to implementing interventions to reduce readmissions in this population.4,5 However, because these programs are resource-intensive, interventions are thought to be most cost-effective if they are targeted to high-risk individuals who are most likely to benefit.6-8

Current pneumonia-specific readmission risk-prediction models that could enable identification of high-risk patients suffer from poor predictive ability, greatly limiting their use, and most were validated among older adults or by using data from single academic medical centers, limiting their generalizability.9-14 A potential reason for poor predictive accuracy is the omission of known robust clinical predictors of pneumonia-related outcomes, including pneumonia severity of illness and stability on discharge.15-17 Approaches using electronic health record (EHR) data, which include this clinically granular data, could enable hospitals to more accurately and pragmatically identify high-risk patients during the index hospitalization and enable interventions to be initiated prior to discharge.

An alternative strategy to identifying high-risk patients for readmission is to use a multi-condition risk-prediction model. Developing and implementing models for every condition may be time-consuming and costly. We have derived and validated 2 multi-condition risk-prediction models using EHR data—1 using data from the first day of hospital admission (‘first-day’ model), and the second incorporating data from the entire hospitalization (‘full-stay’ model) to reflect in-hospital complications and clinical stability at discharge.18,19 However, it is unknown if a multi-condition model for pneumonia would perform as well as a disease-specific model.

This study aimed to develop 2 EHR-based pneumonia-specific readmission risk-prediction models using data routinely collected in clinical practice—a ‘first-day’ and a ‘full-stay’ model—and compare the performance of each model to: 1) one another; 2) the corresponding multi-condition EHR model; and 3) to other potentially useful models in predicting pneumonia readmissions (the Centers for Medicare and Medicaid Services [CMS] pneumonia model, and 2 commonly used pneumonia severity of illness scores validated for predicting mortality). We hypothesized that the pneumonia-specific EHR models would outperform other models; and the full-stay pneumonia-specific model would outperform the first-day pneumonia-specific model.

METHODS

Study Design, Population, and Data Sources

 

 

We conducted an observational study using EHR data collected from 6 hospitals (including safety net, community, teaching, and nonteaching hospitals) in north Texas between November 2009 and October 2010, All hospitals used the Epic EHR (Epic Systems Corporation, Verona, WI). Details of this cohort have been published.18,19

We included consecutive hospitalizations among adults 18 years and older discharged from any medicine service with principal discharge diagnoses of pneumonia (ICD-9-CM codes 480-483, 485, 486-487), sepsis (ICD-9-CM codes 038, 995.91, 995.92, 785.52), or respiratory failure (ICD-9-CM codes 518.81, 518.82, 518.84, 799.1) when the latter 2 were also accompanied by a secondary diagnosis of pneumonia.20 For individuals with multiple hospitalizations during the study period, we included only the first hospitalization. We excluded individuals who died during the index hospitalization or within 30 days of discharge, were transferred to another acute care facility, or left against medical advice.

Outcomes

The primary outcome was all-cause 30-day readmission, defined as a nonelective hospitalization within 30 days of discharge to any of 75 acute care hospitals within a 100-mile radius of Dallas, ascertained from an all-payer regional hospitalization database.

Predictor Variables for the Pneumonia-Specific Readmission Models

The selection of candidate predictors was informed by our validated multi-condition risk-prediction models using EHR data available within 24 hours of admission (‘first-day’ multi-condition EHR model) or during the entire hospitalization (‘full-stay’ multi-condition EHR model).18,19 For the pneumonia-specific models, we included all variables in our published multi-condition models as candidate predictors, including sociodemographics, prior utilization, Charlson Comorbidity Index, select laboratory and vital sign abnormalities, length of stay, hospital complications (eg, venous thromboembolism), vital sign instabilities, and disposition status (see Supplemental Table 1 for complete list of variables). We also assessed additional variables specific to pneumonia for inclusion that were: (1) available in the EHR of all participating hospitals; (2) routinely collected or available at the time of admission or discharge; and (3) plausible predictors of adverse outcomes based on literature and clinical expertise. These included select comorbidities (eg, psychiatric conditions, chronic lung disease, history of pneumonia),10,11,21,22 the pneumonia severity index (PSI),16,23,24 intensive care unit stay, and receipt of invasive or noninvasive ventilation. We used a modified PSI score because certain data elements were missing. The modified PSI (henceforth referred to as PSI) did not include nursing home residence and included diagnostic codes as proxies for the presence of pleural effusion (ICD-9-CM codes 510, 511.1, and 511.9) and altered mental status (ICD-9-CM codes 780.0X, 780.97, 293.0, 293.1, and 348.3X).

Statistical Analysis

Model Derivation. Candidate predictor variables were classified as available in the EHR within 24 hours of admission and/or at the time of discharge. For example, socioeconomic factors could be ascertained within the first day of hospitalization, whereas length of stay would not be available until the day of discharge. Predictors with missing values were assumed to be normal (less than 1% missing for each variable). Univariate relationships between readmission and each candidate predictor were assessed in the overall cohort using a pre-specified significance threshold of P ≤ 0.10. Significant variables were entered in the respective first-day and full-stay pneumonia-specific multivariable logistic regression models using stepwise-backward selection with a pre-specified significance threshold of P ≤ 0.05. In sensitivity analyses, we alternately derived our models using stepwise-forward selection, as well as stepwise-backward selection minimizing the Bayesian information criterion and Akaike information criterion separately. These alternate modeling strategies yielded identical predictors to our final models.

Model Validation. Model validation was performed using 5-fold cross-validation, with the overall cohort randomly divided into 5 equal-size subsets.25 For each cycle, 4 subsets were used for training to estimate model coefficients, and the fifth subset was used for validation. This cycle was repeated 5 times with each randomly-divided subset used once as the validation set. We repeated this entire process 50 times and averaged the C statistic estimates to derive an optimism-corrected C statistic. Model calibration was assessed qualitatively by comparing predicted to observed probabilities of readmission by quintiles of predicted risk, and with the Hosmer-Lemeshow goodness-of-fit test.

Comparison to Other Models. The main comparisons of the first-day and full-stay pneumonia-specific EHR model performance were to each other and the corresponding multi-condition EHR model.18,19 The multi-condition EHR models were separately derived and validated within the larger parent cohort from which this study cohort was derived, and outperformed the CMS all-cause model, the HOSPITAL model, and the LACE index.19 To further triangulate our findings, given the lack of other rigorously validated pneumonia-specific risk-prediction models for readmission,14 we compared the pneumonia-specific EHR models to the CMS pneumonia model derived from administrative claims data,10 and 2 commonly used risk-prediction scores for short-term mortality among patients with community-acquired pneumonia, the PSI and CURB-65 scores.16 Although derived and validated using patient-level data, the CMS model was developed to benchmark hospitals according to hospital-level readmission rates.10 The CURB-65 score in this study was also modified to include the same altered mental status diagnostic codes according to the modified PSI as a proxy for “confusion.” Both the PSI and CURB-65 scores were calculated using the most abnormal values within the first 24 hours of admission. The ‘updated’ PSI and the ‘updated’ CURB-65 were calculated using the most abnormal values within 24 hours prior to discharge, or the last known observation prior to discharge if no results were recorded within this time period. A complete list of variables for each of the comparison models are shown in Supplemental Table 1.

We assessed model performance by calculating the C statistic, integrated discrimination index, and net reclassification index (NRI) compared to our pneumonia-specific models. The integrated discrimination index is the difference in the mean predicted probability of readmission between patients who were and were not actually readmitted between 2 models, where more positive values suggest improvement in model performance compared to a reference model.26 The NRI is defined as the sum of the net proportions of correctly reclassified persons with and without the event of interest.27 Here, we calculated a category-based NRI to evaluate the performance of pneumonia-specific models in correctly classifying individuals with and without readmissions into the 2 highest readmission risk quintiles vs the lowest 3 risk quintiles compared to other models.27 This pre-specified cutoff is relevant for hospitals interested in identifying the highest risk individuals for targeted intervention.7 Finally, we assessed calibration of comparator models in our cohort by comparing predicted probability to observed probability of readmission by quintiles of risk for each model. We conducted all analyses using Stata 12.1 (StataCorp, College Station, Texas). This study was approved by the University of Texas Southwestern Medical Center Institutional Review Board.

 

 

RESULTS

Of 1463 index hospitalizations (Supplemental Figure 1), the 30-day all-cause readmission rate was 13.6%. Individuals with a 30-day readmission had markedly different sociodemographic and clinical characteristics compared to those not readmitted (Table 1; see Supplemental Table 2 for additional clinical characteristics).

Baseline Characteristics of Patients Hospitalized with Pneumonia
Table 1

Derivation, Validation, and Performance of the Pneumonia-Specific Readmission Risk-Prediction Models

The final first-day pneumonia-specific EHR model included 7 variables, including sociodemographic characteristics; prior hospitalizations; thrombocytosis, and PSI (Table 2). The first-day pneumonia-specific model had adequate discrimination (C statistic, 0.695; optimism-corrected C statistic 0.675, 95% confidence interval [CI], 0.667-0.685; Table 3). It also effectively stratified individuals across a broad range of risk (average predicted decile of risk ranged from 4% to 33%; Table 3) and was well calibrated (Supplemental Table 3).

Final Pneumonia-Specific EHR Risk-Prediction Models for Readmissions
Table 2

The final full-stay pneumonia-specific EHR readmission model included 8 predictors, including 3 variables from the first-day model (median income, thrombocytosis, and prior hospitalizations; Table 2). The full-stay pneumonia-specific EHR model also included vital sign instabilities on discharge, updated PSI, and disposition status (ie, being discharged with home health or to a post-acute care facility was associated with greater odds of readmission, and hospice with lower odds). The full-stay pneumonia-specific EHR model had good discrimination (C statistic, 0.731; optimism-corrected C statistic, 0.714; 95% CI, 0.706-0.720), and stratified individuals across a broad range of risk (average predicted decile of risk ranged from 3% to 37%; Table 3), and was also well calibrated (Supplemental Table 3).

Model Performance and Comparison of Pneumonia-Specific EHR Readmissions Models vs Other Models
Table 3

First-Day Pneumonia-Specific EHR Model vs First-Day Multi-Condition EHR Model

The first-day pneumonia-specific EHR model outperformed the first-day multi-condition EHR model with better discrimination (P = 0.029) and more correctly classified individuals in the top 2 highest risk quintiles vs the bottom 3 risk quintiles (Table 3, Supplemental Table 4, and Supplemental Figure 2A). With respect to calibration, the first-day multi-condition EHR model overestimated risk among the highest quintile risk group compared to the first-day pneumonia-specific EHR model (Figure 1A, 1B).

Comparison of the calibration of different readmission models
Figure 1

Full-Stay Pneumonia-Specific EHR Model vs Other Models

The full-stay pneumonia-specific EHR model comparatively outperformed the corresponding full-stay multi-condition EHR model, as well as the first-day pneumonia-specific EHR model, the CMS pneumonia model, the updated PSI, and the updated CURB-65 (Table 3, Supplemental Table 5, Supplemental Table 6, and Supplemental Figures 2B and 2C). Compared to the full-stay multi-condition and first-day pneumonia-specific EHR models, the full-stay pneumonia-specific EHR model had better discrimination, better reclassification (NRI, 0.09 and 0.08, respectively), and was able to stratify individuals across a broader range of readmission risk (Table 3). It also had better calibration in the highest quintile risk group compared to the full-stay multi-condition EHR model (Figure 1C and 1D).

Updated vs First-Day Modified PSI and CURB-65 Scores

The updated PSI was more strongly predictive of readmission than the PSI calculated on the day of admission (Wald test, 9.83; P = 0.002). Each 10-point increase in the updated PSI was associated with a 22% increased odds of readmission vs an 11% increase for the PSI calculated upon admission (Table 2). The improved predictive ability of the updated PSI and CURB-65 scores was also reflected in the superior discrimination and calibration vs the respective first-day pneumonia severity of illness scores (Table 3).

DISCUSSION

Using routinely available EHR data from 6 diverse hospitals, we developed 2 pneumonia-specific readmission risk-prediction models that aimed to allow hospitals to identify patients hospitalized with pneumonia at high risk for readmission. Overall, we found that a pneumonia-specific model using EHR data from the entire hospitalization outperformed all other models—including the first-day pneumonia-specific model using data present only on admission, our own multi-condition EHR models, and the CMS pneumonia model based on administrative claims data—in all aspects of model performance (discrimination, calibration, and reclassification). We found that socioeconomic status, prior hospitalizations, thrombocytosis, and measures of clinical severity and stability were important predictors of 30-day all-cause readmissions among patients hospitalized with pneumonia. Additionally, an updated discharge PSI score was a stronger independent predictor of readmissions compared to the PSI score calculated upon admission; and inclusion of the updated PSI in our full-stay pneumonia model led to improved prediction of 30-day readmissions.

The marked improvement in performance of the full-stay pneumonia-specific EHR model compared to the first-day pneumonia-specific model suggests that clinical stability and trajectory during hospitalization (as modeled through disposition status, updated PSI, and vital sign instabilities at discharge) are important predictors of 30-day readmission among patients hospitalized for pneumonia, which was not the case for our EHR-based multi-condition models.19 With the inclusion of these measures, the full-stay pneumonia-specific model correctly reclassified an additional 8% of patients according to their true risk compared to the first-day pneumonia-specific model. One implication of these findings is that hospitals interested in targeting their highest risk individuals with pneumonia for transitional care interventions could do so using the first-day pneumonia-specific EHR model and could refine their targeted strategy at the time of discharge by using the full-stay pneumonia model. This staged risk-prediction strategy would enable hospitals to initiate transitional care interventions for high-risk individuals in the inpatient setting (ie, patient education).7 Then, hospitals could enroll both persistent and newly identified high-risk individuals for outpatient interventions (ie, follow-up telephone call) in the immediate post-discharge period, an interval characterized by heightened vulnerability for adverse events,28 based on patients’ illness severity and stability at discharge. This approach can be implemented by hospitals by building these risk-prediction models directly into the EHR, or by extracting EHR data in near real time as our group has done successfully for heart failure.7

Another key implication of our study is that, for pneumonia, a disease-specific modeling approach has better predictive ability than using a multi-condition model. Compared to multi-condition models, the first-day and full-stay pneumonia-specific EHR models correctly reclassified an additional 6% and 9% of patients, respectively. Thus, hospitals interested in identifying the highest risk patients with pneumonia for targeted interventions should do so using the disease-specific models, if the costs and resources of doing so are within reach of the healthcare system.

An additional novel finding of our study is the added value of an updated PSI for predicting adverse events. Studies of pneumonia severity of illness scores have calculated the PSI and CURB-65 scores using data present only on admission.16,24 While our study also confirms that the PSI calculated upon admission is a significant predictor of readmission,23,29 this study extends this work by showing that an updated PSI score calculated at the time of discharge is an even stronger predictor for readmission, and its inclusion in the model significantly improves risk stratification and prognostication.

Our study was noteworthy for several strengths. First, we used data from a common EHR system, thus potentially allowing for the implementation of the pneumonia-specific models in real time across a number of hospitals. The use of routinely collected data for risk-prediction modeling makes this approach scalable and sustainable, because it obviates the need for burdensome data collection and entry. Second, to our knowledge, this is the first study to measure the additive influence of illness severity and stability at discharge on the readmission risk among patients hospitalized with pneumonia. Third, our study population was derived from 6 hospitals diverse in payer status, age, race/ethnicity, and socioeconomic status. Fourth, our models are less likely to be overfit to the idiosyncrasies of our data given that several predictors included in our final pneumonia-specific models have been associated with readmission in this population, including marital status,13,30 income,11,31 prior hospitalizations,11,13 thrombocytosis,32-34 and vital sign instabilities on discharge.17 Lastly, the discrimination of the CMS pneumonia model in our cohort (C statistic, 0.64) closely matched the discrimination observed in 4 independent cohorts (C statistic, 0.63), suggesting adequate generalizability of our study setting and population.10,12

Our results should be interpreted in the context of several limitations. First, generalizability to other regions beyond north Texas is unknown. Second, although we included a diverse cohort of safety net, community, teaching, and nonteaching hospitals, the pneumonia-specific models were not externally validated in a separate cohort, which may lead to more optimistic estimates of model performance. Third, PSI and CURB-65 scores were modified to use diagnostic codes for altered mental status and pleural effusion, and omitted nursing home residence. Thus, the independent associations for the PSI and CURB-65 scores and their predictive ability are likely attenuated. Fourth, we were unable to include data on medications (antibiotics and steroid use) and outpatient visits, which may influence readmission risk.2,9,13,35-40 Fifth, we included only the first pneumonia hospitalization per patient in this study. Had we included multiple hospitalizations per patient, we anticipate better model performance for the 2 pneumonia-specific EHR models since prior hospitalization was a robust predictor of readmission.

In conclusion, the full-stay pneumonia-specific EHR readmission risk-prediction model outperformed the first-day pneumonia-specific model, multi-condition EHR models, and the CMS pneumonia model. This suggests that: measures of clinical severity and stability at the time of discharge are important predictors for identifying patients at highest risk for readmission; and that EHR data routinely collected for clinical practice can be used to accurately predict risk of readmission among patients hospitalized for pneumonia.

 

 

Acknowledgments

The authors would like to acknowledge Ruben Amarasingham, MD, MBA, president and chief executive officer of Parkland Center for Clinical Innovation, and Ferdinand Velasco, MD, chief health information officer at Texas Health Resources for their assistance in assembling the 6-hospital cohort used in this study.

Disclosures

This work was supported by the Agency for Healthcare Research and Quality-funded UT Southwestern Center for Patient-Centered Outcomes Research (R24 HS022418-01); the Commonwealth Foundation (#20100323); the UT Southwestern KL2 Scholars Program supported by the National Institutes of Health (KL2 TR001103 to ANM and OKN); and the National Center for Advancing Translational Sciences at the National Institute of Health (U54 RFA-TR-12-006 to E.A.H.). The study sponsors had no role in design and conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript. The authors have no financial conflicts of interest to disclose

Files
References

1. Centers for Disease Control and Prevention. Pneumonia. http://www.cdc.gov/nchs/fastats/pneumonia.htm. Accessed January 26, 2016.
2. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service program. N Engl J Med. 2009;364(16):1582. PubMed
3. van Walraven C, Bennett C, Jennings A, Austin PC, Forster AJ. Proportion of hospital readmissions deemed avoidable: a systematic review. CMAJ. 2011;183(7):E391-E402. PubMed
4. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital-initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433-440. PubMed
5. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30-day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520-528. PubMed
6. Rennke S, Shoeb MH, Nguyen OK, Magan Y, Wachter RM, Ranji SR. Interventions to Improve Care Transitions at Hospital Discharge. Rockville, MD: Agency for Healthcare Research and Quality, US Department of Health and Human Services;March 2013. PubMed
7. Amarasingham R, Patel PC, Toto K, et al. Allocating scarce resources in real-time to reduce heart failure readmissions: a prospective, controlled study. BMJ Qual Saf. 2013;22(12):998-1005. PubMed
8. Amarasingham R, Patzer RE, Huesch M, Nguyen NQ, Xie B. Implementing electronic health care predictive analytics: considerations and challenges. Health Aff (Millwood). 2014;33(7):1148-1154. PubMed
9. Hebert C, Shivade C, Foraker R, et al. Diagnosis-specific readmission risk prediction using electronic health data: a retrospective cohort study. BMC Med Inform Decis Mak. 2014;14:65. PubMed
10. Lindenauer PK, Normand SL, Drye EE, et al. Development, validation, and results of a measure of 30-day readmission following hospitalization for pneumonia. J Hosp Med. 2011;6(3):142-150. PubMed
11. Mather JF, Fortunato GJ, Ash JL, Davis MJ, Kumar A. Prediction of pneumonia 30-day readmissions: a single-center attempt to increase model performance. Respir Care. 2014;59(2):199-208. PubMed
12. O’Brien WJ, Chen Q, Mull HJ, et al. What is the value of adding Medicare data in estimating VA hospital readmission rates? Health Serv Res. 2015;50(1):40-57. PubMed
13. Tang VL, Halm EA, Fine MJ, Johnson CS, Anzueto A, Mortensen EM. Predictors of rehospitalization after admission for pneumonia in the veterans affairs healthcare system. J Hosp Med. 2014;9(6):379-383. PubMed
14. Weinreich M, Nguyen OK, Wang D, et al. Predicting the risk of readmission in pneumonia: a systematic review of model performance. Ann Am Thorac Soc. 2016;13(9):1607-1614. PubMed
15. Kwok CS, Loke YK, Woo K, Myint PK. Risk prediction models for mortality in community-acquired pneumonia: a systematic review. Biomed Res Int. 2013;2013:504136. PubMed
16. Loke YK, Kwok CS, Niruban A, Myint PK. Value of severity scales in predicting mortality from community-acquired pneumonia: systematic review and meta-analysis. Thorax. 2010;65(10):884-890. PubMed
17. Halm EA, Fine MJ, Kapoor WN, Singer DE, Marrie TJ, Siu AL. Instability on hospital discharge and the risk of adverse outcomes in patients with pneumonia. Arch Intern Med. 2002;162(11):1278-1284. PubMed
18. Amarasingham R, Velasco F, Xie B, et al. Electronic medical record-based multicondition models to predict the risk of 30 day readmission or death among adult medicine patients: validation and comparison to existing models. BMC Med Inform Decis Mak. 2015;15:39. PubMed
19. Nguyen OK, Makam AN, Clark C, et al. Predicting all-cause readmissions using electronic health record data from the entire hospitalization: Model development and comparison. J Hosp Med. 2016;11(7):473-480. PubMed
20. Lindenauer PK, Lagu T, Shieh MS, Pekow PS, Rothberg MB. Association of diagnostic coding with trends in hospitalizations and mortality of patients with pneumonia, 2003-2009. JAMA. 2012;307(13):1405-1413. PubMed
21. Ahmedani BK, Solberg LI, Copeland LA, et al. Psychiatric comorbidity and 30-day readmissions after hospitalization for heart failure, AMI, and pneumonia. Psychiatr Serv. 2015;66(2):134-140. PubMed
22. Jasti H, Mortensen EM, Obrosky DS, Kapoor WN, Fine MJ. Causes and risk factors for rehospitalization of patients hospitalized with community-acquired pneumonia. Clin Infect Dis. 2008;46(4):550-556. PubMed
23. Capelastegui A, España Yandiola PP, Quintana JM, et al. Predictors of short-term rehospitalization following discharge of patients hospitalized with community-acquired pneumonia. Chest. 2009;136(4):1079-1085. PubMed
24. Fine MJ, Auble TE, Yealy DM, et al. A prediction rule to identify low-risk patients with community-acquired pneumonia. N Engl J Med. 1997;336(4):243-250. PubMed
25. Vittinghoff E, Glidden D, Shiboski S, McCulloch C. Regression Methods in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models (Statistics for Biology and Health). New York City, NY: Springer; 2012.
26. Pencina MJ, D’Agostino RB Sr, D’Agostino RB Jr, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27(2):157-172; discussion 207-112. PubMed
27. Leening MJ, Vedder MM, Witteman JC, Pencina MJ, Steyerberg EW. Net reclassification improvement: computation, interpretation, and controversies: a literature review and clinician’s guide. Ann Intern Med. 2014;160(2):122-131. PubMed
28. Krumholz HM. Post-hospital syndrome--an acquired, transient condition of generalized risk. N Engl J Med. 2013;368(2):100-102. PubMed
29. Micek ST, Lang A, Fuller BM, Hampton NB, Kollef MH. Clinical implications for patients treated inappropriately for community-acquired pneumonia in the emergency department. BMC Infect Dis. 2014;14:61. PubMed
30. Metersky ML, Fine MJ, Mortensen EM. The effect of marital status on the presentation and outcomes of elderly male veterans hospitalized for pneumonia. Chest. 2012;142(4):982-987. PubMed
31. Calvillo-King L, Arnold D, Eubank KJ, et al. Impact of social factors on risk of readmission or mortality in pneumonia and heart failure: systematic review. J Gen Intern Med. 2013;28(2):269-282. PubMed
32. Mirsaeidi M, Peyrani P, Aliberti S, et al. Thrombocytopenia and thrombocytosis at time of hospitalization predict mortality in patients with community-acquired pneumonia. Chest. 2010;137(2):416-420. PubMed
33. Prina E, Ferrer M, Ranzani OT, et al. Thrombocytosis is a marker of poor outcome in community-acquired pneumonia. Chest. 2013;143(3):767-775. PubMed

34. Violi F, Cangemi R, Calvieri C. Pneumonia, thrombosis and vascular disease. J Thromb Haemost. 2014;12(9):1391-1400. PubMed
35. Weinberger M, Oddone EZ, Henderson WG. Does increased access to primary care reduce hospital readmissions? Veterans Affairs Cooperative Study Group on Primary Care and Hospital Readmission. N Engl J Med. 1996;334(22):1441-1447. PubMed
36. Field TS, Ogarek J, Garber L, Reed G, Gurwitz JH. Association of early post-discharge follow-up by a primary care physician and 30-day rehospitalization among older adults. J Gen Intern Med. 2015;30(5):565-571. PubMed
37. Spatz ES, Sheth SD, Gosch KL, et al. Usual source of care and outcomes following acute myocardial infarction. J Gen Intern Med. 2014;29(6):862-869. PubMed
38. Brooke BS, Stone DH, Cronenwett JL, et al. Early primary care provider follow-up and readmission after high-risk surgery. JAMA Surg. 2014;149(8):821-828. PubMed
39. Adamuz J, Viasus D, Campreciós-Rodriguez P, et al. A prospective cohort study of healthcare visits and rehospitalizations after discharge of patients with community-acquired pneumonia. Respirology. 2011;16(7):1119-1126. PubMed
40. Shorr AF, Zilberberg MD, Reichley R, et al. Readmission following hospitalization for pneumonia: the impact of pneumonia type and its implication for hospitals. Clin Infect Dis. 2013;57(3):362-367. PubMed

Article PDF
Issue
Journal of Hospital Medicine 12(4)
Topics
Page Number
209-216
Sections
Files
Files
Article PDF
Article PDF

Pneumonia is a leading cause of hospitalizations in the U.S., accounting for more than 1.1 million discharges annually.1 Pneumonia is frequently complicated by hospital readmission, which is costly and potentially avoidable.2,3 Due to financial penalties imposed on hospitals for higher than expected 30-day readmission rates, there is increasing attention to implementing interventions to reduce readmissions in this population.4,5 However, because these programs are resource-intensive, interventions are thought to be most cost-effective if they are targeted to high-risk individuals who are most likely to benefit.6-8

Current pneumonia-specific readmission risk-prediction models that could enable identification of high-risk patients suffer from poor predictive ability, greatly limiting their use, and most were validated among older adults or by using data from single academic medical centers, limiting their generalizability.9-14 A potential reason for poor predictive accuracy is the omission of known robust clinical predictors of pneumonia-related outcomes, including pneumonia severity of illness and stability on discharge.15-17 Approaches using electronic health record (EHR) data, which include this clinically granular data, could enable hospitals to more accurately and pragmatically identify high-risk patients during the index hospitalization and enable interventions to be initiated prior to discharge.

An alternative strategy to identifying high-risk patients for readmission is to use a multi-condition risk-prediction model. Developing and implementing models for every condition may be time-consuming and costly. We have derived and validated 2 multi-condition risk-prediction models using EHR data—1 using data from the first day of hospital admission (‘first-day’ model), and the second incorporating data from the entire hospitalization (‘full-stay’ model) to reflect in-hospital complications and clinical stability at discharge.18,19 However, it is unknown if a multi-condition model for pneumonia would perform as well as a disease-specific model.

This study aimed to develop 2 EHR-based pneumonia-specific readmission risk-prediction models using data routinely collected in clinical practice—a ‘first-day’ and a ‘full-stay’ model—and compare the performance of each model to: 1) one another; 2) the corresponding multi-condition EHR model; and 3) to other potentially useful models in predicting pneumonia readmissions (the Centers for Medicare and Medicaid Services [CMS] pneumonia model, and 2 commonly used pneumonia severity of illness scores validated for predicting mortality). We hypothesized that the pneumonia-specific EHR models would outperform other models; and the full-stay pneumonia-specific model would outperform the first-day pneumonia-specific model.

METHODS

Study Design, Population, and Data Sources

 

 

We conducted an observational study using EHR data collected from 6 hospitals (including safety net, community, teaching, and nonteaching hospitals) in north Texas between November 2009 and October 2010, All hospitals used the Epic EHR (Epic Systems Corporation, Verona, WI). Details of this cohort have been published.18,19

We included consecutive hospitalizations among adults 18 years and older discharged from any medicine service with principal discharge diagnoses of pneumonia (ICD-9-CM codes 480-483, 485, 486-487), sepsis (ICD-9-CM codes 038, 995.91, 995.92, 785.52), or respiratory failure (ICD-9-CM codes 518.81, 518.82, 518.84, 799.1) when the latter 2 were also accompanied by a secondary diagnosis of pneumonia.20 For individuals with multiple hospitalizations during the study period, we included only the first hospitalization. We excluded individuals who died during the index hospitalization or within 30 days of discharge, were transferred to another acute care facility, or left against medical advice.

Outcomes

The primary outcome was all-cause 30-day readmission, defined as a nonelective hospitalization within 30 days of discharge to any of 75 acute care hospitals within a 100-mile radius of Dallas, ascertained from an all-payer regional hospitalization database.

Predictor Variables for the Pneumonia-Specific Readmission Models

The selection of candidate predictors was informed by our validated multi-condition risk-prediction models using EHR data available within 24 hours of admission (‘first-day’ multi-condition EHR model) or during the entire hospitalization (‘full-stay’ multi-condition EHR model).18,19 For the pneumonia-specific models, we included all variables in our published multi-condition models as candidate predictors, including sociodemographics, prior utilization, Charlson Comorbidity Index, select laboratory and vital sign abnormalities, length of stay, hospital complications (eg, venous thromboembolism), vital sign instabilities, and disposition status (see Supplemental Table 1 for complete list of variables). We also assessed additional variables specific to pneumonia for inclusion that were: (1) available in the EHR of all participating hospitals; (2) routinely collected or available at the time of admission or discharge; and (3) plausible predictors of adverse outcomes based on literature and clinical expertise. These included select comorbidities (eg, psychiatric conditions, chronic lung disease, history of pneumonia),10,11,21,22 the pneumonia severity index (PSI),16,23,24 intensive care unit stay, and receipt of invasive or noninvasive ventilation. We used a modified PSI score because certain data elements were missing. The modified PSI (henceforth referred to as PSI) did not include nursing home residence and included diagnostic codes as proxies for the presence of pleural effusion (ICD-9-CM codes 510, 511.1, and 511.9) and altered mental status (ICD-9-CM codes 780.0X, 780.97, 293.0, 293.1, and 348.3X).

Statistical Analysis

Model Derivation. Candidate predictor variables were classified as available in the EHR within 24 hours of admission and/or at the time of discharge. For example, socioeconomic factors could be ascertained within the first day of hospitalization, whereas length of stay would not be available until the day of discharge. Predictors with missing values were assumed to be normal (less than 1% missing for each variable). Univariate relationships between readmission and each candidate predictor were assessed in the overall cohort using a pre-specified significance threshold of P ≤ 0.10. Significant variables were entered in the respective first-day and full-stay pneumonia-specific multivariable logistic regression models using stepwise-backward selection with a pre-specified significance threshold of P ≤ 0.05. In sensitivity analyses, we alternately derived our models using stepwise-forward selection, as well as stepwise-backward selection minimizing the Bayesian information criterion and Akaike information criterion separately. These alternate modeling strategies yielded identical predictors to our final models.

Model Validation. Model validation was performed using 5-fold cross-validation, with the overall cohort randomly divided into 5 equal-size subsets.25 For each cycle, 4 subsets were used for training to estimate model coefficients, and the fifth subset was used for validation. This cycle was repeated 5 times with each randomly-divided subset used once as the validation set. We repeated this entire process 50 times and averaged the C statistic estimates to derive an optimism-corrected C statistic. Model calibration was assessed qualitatively by comparing predicted to observed probabilities of readmission by quintiles of predicted risk, and with the Hosmer-Lemeshow goodness-of-fit test.

Comparison to Other Models. The main comparisons of the first-day and full-stay pneumonia-specific EHR model performance were to each other and the corresponding multi-condition EHR model.18,19 The multi-condition EHR models were separately derived and validated within the larger parent cohort from which this study cohort was derived, and outperformed the CMS all-cause model, the HOSPITAL model, and the LACE index.19 To further triangulate our findings, given the lack of other rigorously validated pneumonia-specific risk-prediction models for readmission,14 we compared the pneumonia-specific EHR models to the CMS pneumonia model derived from administrative claims data,10 and 2 commonly used risk-prediction scores for short-term mortality among patients with community-acquired pneumonia, the PSI and CURB-65 scores.16 Although derived and validated using patient-level data, the CMS model was developed to benchmark hospitals according to hospital-level readmission rates.10 The CURB-65 score in this study was also modified to include the same altered mental status diagnostic codes according to the modified PSI as a proxy for “confusion.” Both the PSI and CURB-65 scores were calculated using the most abnormal values within the first 24 hours of admission. The ‘updated’ PSI and the ‘updated’ CURB-65 were calculated using the most abnormal values within 24 hours prior to discharge, or the last known observation prior to discharge if no results were recorded within this time period. A complete list of variables for each of the comparison models are shown in Supplemental Table 1.

We assessed model performance by calculating the C statistic, integrated discrimination index, and net reclassification index (NRI) compared to our pneumonia-specific models. The integrated discrimination index is the difference in the mean predicted probability of readmission between patients who were and were not actually readmitted between 2 models, where more positive values suggest improvement in model performance compared to a reference model.26 The NRI is defined as the sum of the net proportions of correctly reclassified persons with and without the event of interest.27 Here, we calculated a category-based NRI to evaluate the performance of pneumonia-specific models in correctly classifying individuals with and without readmissions into the 2 highest readmission risk quintiles vs the lowest 3 risk quintiles compared to other models.27 This pre-specified cutoff is relevant for hospitals interested in identifying the highest risk individuals for targeted intervention.7 Finally, we assessed calibration of comparator models in our cohort by comparing predicted probability to observed probability of readmission by quintiles of risk for each model. We conducted all analyses using Stata 12.1 (StataCorp, College Station, Texas). This study was approved by the University of Texas Southwestern Medical Center Institutional Review Board.

 

 

RESULTS

Of 1463 index hospitalizations (Supplemental Figure 1), the 30-day all-cause readmission rate was 13.6%. Individuals with a 30-day readmission had markedly different sociodemographic and clinical characteristics compared to those not readmitted (Table 1; see Supplemental Table 2 for additional clinical characteristics).

Baseline Characteristics of Patients Hospitalized with Pneumonia
Table 1

Derivation, Validation, and Performance of the Pneumonia-Specific Readmission Risk-Prediction Models

The final first-day pneumonia-specific EHR model included 7 variables, including sociodemographic characteristics; prior hospitalizations; thrombocytosis, and PSI (Table 2). The first-day pneumonia-specific model had adequate discrimination (C statistic, 0.695; optimism-corrected C statistic 0.675, 95% confidence interval [CI], 0.667-0.685; Table 3). It also effectively stratified individuals across a broad range of risk (average predicted decile of risk ranged from 4% to 33%; Table 3) and was well calibrated (Supplemental Table 3).

Final Pneumonia-Specific EHR Risk-Prediction Models for Readmissions
Table 2

The final full-stay pneumonia-specific EHR readmission model included 8 predictors, including 3 variables from the first-day model (median income, thrombocytosis, and prior hospitalizations; Table 2). The full-stay pneumonia-specific EHR model also included vital sign instabilities on discharge, updated PSI, and disposition status (ie, being discharged with home health or to a post-acute care facility was associated with greater odds of readmission, and hospice with lower odds). The full-stay pneumonia-specific EHR model had good discrimination (C statistic, 0.731; optimism-corrected C statistic, 0.714; 95% CI, 0.706-0.720), and stratified individuals across a broad range of risk (average predicted decile of risk ranged from 3% to 37%; Table 3), and was also well calibrated (Supplemental Table 3).

Model Performance and Comparison of Pneumonia-Specific EHR Readmissions Models vs Other Models
Table 3

First-Day Pneumonia-Specific EHR Model vs First-Day Multi-Condition EHR Model

The first-day pneumonia-specific EHR model outperformed the first-day multi-condition EHR model with better discrimination (P = 0.029) and more correctly classified individuals in the top 2 highest risk quintiles vs the bottom 3 risk quintiles (Table 3, Supplemental Table 4, and Supplemental Figure 2A). With respect to calibration, the first-day multi-condition EHR model overestimated risk among the highest quintile risk group compared to the first-day pneumonia-specific EHR model (Figure 1A, 1B).

Comparison of the calibration of different readmission models
Figure 1

Full-Stay Pneumonia-Specific EHR Model vs Other Models

The full-stay pneumonia-specific EHR model comparatively outperformed the corresponding full-stay multi-condition EHR model, as well as the first-day pneumonia-specific EHR model, the CMS pneumonia model, the updated PSI, and the updated CURB-65 (Table 3, Supplemental Table 5, Supplemental Table 6, and Supplemental Figures 2B and 2C). Compared to the full-stay multi-condition and first-day pneumonia-specific EHR models, the full-stay pneumonia-specific EHR model had better discrimination, better reclassification (NRI, 0.09 and 0.08, respectively), and was able to stratify individuals across a broader range of readmission risk (Table 3). It also had better calibration in the highest quintile risk group compared to the full-stay multi-condition EHR model (Figure 1C and 1D).

Updated vs First-Day Modified PSI and CURB-65 Scores

The updated PSI was more strongly predictive of readmission than the PSI calculated on the day of admission (Wald test, 9.83; P = 0.002). Each 10-point increase in the updated PSI was associated with a 22% increased odds of readmission vs an 11% increase for the PSI calculated upon admission (Table 2). The improved predictive ability of the updated PSI and CURB-65 scores was also reflected in the superior discrimination and calibration vs the respective first-day pneumonia severity of illness scores (Table 3).

DISCUSSION

Using routinely available EHR data from 6 diverse hospitals, we developed 2 pneumonia-specific readmission risk-prediction models that aimed to allow hospitals to identify patients hospitalized with pneumonia at high risk for readmission. Overall, we found that a pneumonia-specific model using EHR data from the entire hospitalization outperformed all other models—including the first-day pneumonia-specific model using data present only on admission, our own multi-condition EHR models, and the CMS pneumonia model based on administrative claims data—in all aspects of model performance (discrimination, calibration, and reclassification). We found that socioeconomic status, prior hospitalizations, thrombocytosis, and measures of clinical severity and stability were important predictors of 30-day all-cause readmissions among patients hospitalized with pneumonia. Additionally, an updated discharge PSI score was a stronger independent predictor of readmissions compared to the PSI score calculated upon admission; and inclusion of the updated PSI in our full-stay pneumonia model led to improved prediction of 30-day readmissions.

The marked improvement in performance of the full-stay pneumonia-specific EHR model compared to the first-day pneumonia-specific model suggests that clinical stability and trajectory during hospitalization (as modeled through disposition status, updated PSI, and vital sign instabilities at discharge) are important predictors of 30-day readmission among patients hospitalized for pneumonia, which was not the case for our EHR-based multi-condition models.19 With the inclusion of these measures, the full-stay pneumonia-specific model correctly reclassified an additional 8% of patients according to their true risk compared to the first-day pneumonia-specific model. One implication of these findings is that hospitals interested in targeting their highest risk individuals with pneumonia for transitional care interventions could do so using the first-day pneumonia-specific EHR model and could refine their targeted strategy at the time of discharge by using the full-stay pneumonia model. This staged risk-prediction strategy would enable hospitals to initiate transitional care interventions for high-risk individuals in the inpatient setting (ie, patient education).7 Then, hospitals could enroll both persistent and newly identified high-risk individuals for outpatient interventions (ie, follow-up telephone call) in the immediate post-discharge period, an interval characterized by heightened vulnerability for adverse events,28 based on patients’ illness severity and stability at discharge. This approach can be implemented by hospitals by building these risk-prediction models directly into the EHR, or by extracting EHR data in near real time as our group has done successfully for heart failure.7

Another key implication of our study is that, for pneumonia, a disease-specific modeling approach has better predictive ability than using a multi-condition model. Compared to multi-condition models, the first-day and full-stay pneumonia-specific EHR models correctly reclassified an additional 6% and 9% of patients, respectively. Thus, hospitals interested in identifying the highest risk patients with pneumonia for targeted interventions should do so using the disease-specific models, if the costs and resources of doing so are within reach of the healthcare system.

An additional novel finding of our study is the added value of an updated PSI for predicting adverse events. Studies of pneumonia severity of illness scores have calculated the PSI and CURB-65 scores using data present only on admission.16,24 While our study also confirms that the PSI calculated upon admission is a significant predictor of readmission,23,29 this study extends this work by showing that an updated PSI score calculated at the time of discharge is an even stronger predictor for readmission, and its inclusion in the model significantly improves risk stratification and prognostication.

Our study was noteworthy for several strengths. First, we used data from a common EHR system, thus potentially allowing for the implementation of the pneumonia-specific models in real time across a number of hospitals. The use of routinely collected data for risk-prediction modeling makes this approach scalable and sustainable, because it obviates the need for burdensome data collection and entry. Second, to our knowledge, this is the first study to measure the additive influence of illness severity and stability at discharge on the readmission risk among patients hospitalized with pneumonia. Third, our study population was derived from 6 hospitals diverse in payer status, age, race/ethnicity, and socioeconomic status. Fourth, our models are less likely to be overfit to the idiosyncrasies of our data given that several predictors included in our final pneumonia-specific models have been associated with readmission in this population, including marital status,13,30 income,11,31 prior hospitalizations,11,13 thrombocytosis,32-34 and vital sign instabilities on discharge.17 Lastly, the discrimination of the CMS pneumonia model in our cohort (C statistic, 0.64) closely matched the discrimination observed in 4 independent cohorts (C statistic, 0.63), suggesting adequate generalizability of our study setting and population.10,12

Our results should be interpreted in the context of several limitations. First, generalizability to other regions beyond north Texas is unknown. Second, although we included a diverse cohort of safety net, community, teaching, and nonteaching hospitals, the pneumonia-specific models were not externally validated in a separate cohort, which may lead to more optimistic estimates of model performance. Third, PSI and CURB-65 scores were modified to use diagnostic codes for altered mental status and pleural effusion, and omitted nursing home residence. Thus, the independent associations for the PSI and CURB-65 scores and their predictive ability are likely attenuated. Fourth, we were unable to include data on medications (antibiotics and steroid use) and outpatient visits, which may influence readmission risk.2,9,13,35-40 Fifth, we included only the first pneumonia hospitalization per patient in this study. Had we included multiple hospitalizations per patient, we anticipate better model performance for the 2 pneumonia-specific EHR models since prior hospitalization was a robust predictor of readmission.

In conclusion, the full-stay pneumonia-specific EHR readmission risk-prediction model outperformed the first-day pneumonia-specific model, multi-condition EHR models, and the CMS pneumonia model. This suggests that: measures of clinical severity and stability at the time of discharge are important predictors for identifying patients at highest risk for readmission; and that EHR data routinely collected for clinical practice can be used to accurately predict risk of readmission among patients hospitalized for pneumonia.

 

 

Acknowledgments

The authors would like to acknowledge Ruben Amarasingham, MD, MBA, president and chief executive officer of Parkland Center for Clinical Innovation, and Ferdinand Velasco, MD, chief health information officer at Texas Health Resources for their assistance in assembling the 6-hospital cohort used in this study.

Disclosures

This work was supported by the Agency for Healthcare Research and Quality-funded UT Southwestern Center for Patient-Centered Outcomes Research (R24 HS022418-01); the Commonwealth Foundation (#20100323); the UT Southwestern KL2 Scholars Program supported by the National Institutes of Health (KL2 TR001103 to ANM and OKN); and the National Center for Advancing Translational Sciences at the National Institute of Health (U54 RFA-TR-12-006 to E.A.H.). The study sponsors had no role in design and conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript. The authors have no financial conflicts of interest to disclose

Pneumonia is a leading cause of hospitalizations in the U.S., accounting for more than 1.1 million discharges annually.1 Pneumonia is frequently complicated by hospital readmission, which is costly and potentially avoidable.2,3 Due to financial penalties imposed on hospitals for higher than expected 30-day readmission rates, there is increasing attention to implementing interventions to reduce readmissions in this population.4,5 However, because these programs are resource-intensive, interventions are thought to be most cost-effective if they are targeted to high-risk individuals who are most likely to benefit.6-8

Current pneumonia-specific readmission risk-prediction models that could enable identification of high-risk patients suffer from poor predictive ability, greatly limiting their use, and most were validated among older adults or by using data from single academic medical centers, limiting their generalizability.9-14 A potential reason for poor predictive accuracy is the omission of known robust clinical predictors of pneumonia-related outcomes, including pneumonia severity of illness and stability on discharge.15-17 Approaches using electronic health record (EHR) data, which include this clinically granular data, could enable hospitals to more accurately and pragmatically identify high-risk patients during the index hospitalization and enable interventions to be initiated prior to discharge.

An alternative strategy to identifying high-risk patients for readmission is to use a multi-condition risk-prediction model. Developing and implementing models for every condition may be time-consuming and costly. We have derived and validated 2 multi-condition risk-prediction models using EHR data—1 using data from the first day of hospital admission (‘first-day’ model), and the second incorporating data from the entire hospitalization (‘full-stay’ model) to reflect in-hospital complications and clinical stability at discharge.18,19 However, it is unknown if a multi-condition model for pneumonia would perform as well as a disease-specific model.

This study aimed to develop 2 EHR-based pneumonia-specific readmission risk-prediction models using data routinely collected in clinical practice—a ‘first-day’ and a ‘full-stay’ model—and compare the performance of each model to: 1) one another; 2) the corresponding multi-condition EHR model; and 3) to other potentially useful models in predicting pneumonia readmissions (the Centers for Medicare and Medicaid Services [CMS] pneumonia model, and 2 commonly used pneumonia severity of illness scores validated for predicting mortality). We hypothesized that the pneumonia-specific EHR models would outperform other models; and the full-stay pneumonia-specific model would outperform the first-day pneumonia-specific model.

METHODS

Study Design, Population, and Data Sources

 

 

We conducted an observational study using EHR data collected from 6 hospitals (including safety net, community, teaching, and nonteaching hospitals) in north Texas between November 2009 and October 2010, All hospitals used the Epic EHR (Epic Systems Corporation, Verona, WI). Details of this cohort have been published.18,19

We included consecutive hospitalizations among adults 18 years and older discharged from any medicine service with principal discharge diagnoses of pneumonia (ICD-9-CM codes 480-483, 485, 486-487), sepsis (ICD-9-CM codes 038, 995.91, 995.92, 785.52), or respiratory failure (ICD-9-CM codes 518.81, 518.82, 518.84, 799.1) when the latter 2 were also accompanied by a secondary diagnosis of pneumonia.20 For individuals with multiple hospitalizations during the study period, we included only the first hospitalization. We excluded individuals who died during the index hospitalization or within 30 days of discharge, were transferred to another acute care facility, or left against medical advice.

Outcomes

The primary outcome was all-cause 30-day readmission, defined as a nonelective hospitalization within 30 days of discharge to any of 75 acute care hospitals within a 100-mile radius of Dallas, ascertained from an all-payer regional hospitalization database.

Predictor Variables for the Pneumonia-Specific Readmission Models

The selection of candidate predictors was informed by our validated multi-condition risk-prediction models using EHR data available within 24 hours of admission (‘first-day’ multi-condition EHR model) or during the entire hospitalization (‘full-stay’ multi-condition EHR model).18,19 For the pneumonia-specific models, we included all variables in our published multi-condition models as candidate predictors, including sociodemographics, prior utilization, Charlson Comorbidity Index, select laboratory and vital sign abnormalities, length of stay, hospital complications (eg, venous thromboembolism), vital sign instabilities, and disposition status (see Supplemental Table 1 for complete list of variables). We also assessed additional variables specific to pneumonia for inclusion that were: (1) available in the EHR of all participating hospitals; (2) routinely collected or available at the time of admission or discharge; and (3) plausible predictors of adverse outcomes based on literature and clinical expertise. These included select comorbidities (eg, psychiatric conditions, chronic lung disease, history of pneumonia),10,11,21,22 the pneumonia severity index (PSI),16,23,24 intensive care unit stay, and receipt of invasive or noninvasive ventilation. We used a modified PSI score because certain data elements were missing. The modified PSI (henceforth referred to as PSI) did not include nursing home residence and included diagnostic codes as proxies for the presence of pleural effusion (ICD-9-CM codes 510, 511.1, and 511.9) and altered mental status (ICD-9-CM codes 780.0X, 780.97, 293.0, 293.1, and 348.3X).

Statistical Analysis

Model Derivation. Candidate predictor variables were classified as available in the EHR within 24 hours of admission and/or at the time of discharge. For example, socioeconomic factors could be ascertained within the first day of hospitalization, whereas length of stay would not be available until the day of discharge. Predictors with missing values were assumed to be normal (less than 1% missing for each variable). Univariate relationships between readmission and each candidate predictor were assessed in the overall cohort using a pre-specified significance threshold of P ≤ 0.10. Significant variables were entered in the respective first-day and full-stay pneumonia-specific multivariable logistic regression models using stepwise-backward selection with a pre-specified significance threshold of P ≤ 0.05. In sensitivity analyses, we alternately derived our models using stepwise-forward selection, as well as stepwise-backward selection minimizing the Bayesian information criterion and Akaike information criterion separately. These alternate modeling strategies yielded identical predictors to our final models.

Model Validation. Model validation was performed using 5-fold cross-validation, with the overall cohort randomly divided into 5 equal-size subsets.25 For each cycle, 4 subsets were used for training to estimate model coefficients, and the fifth subset was used for validation. This cycle was repeated 5 times with each randomly-divided subset used once as the validation set. We repeated this entire process 50 times and averaged the C statistic estimates to derive an optimism-corrected C statistic. Model calibration was assessed qualitatively by comparing predicted to observed probabilities of readmission by quintiles of predicted risk, and with the Hosmer-Lemeshow goodness-of-fit test.

Comparison to Other Models. The main comparisons of the first-day and full-stay pneumonia-specific EHR model performance were to each other and the corresponding multi-condition EHR model.18,19 The multi-condition EHR models were separately derived and validated within the larger parent cohort from which this study cohort was derived, and outperformed the CMS all-cause model, the HOSPITAL model, and the LACE index.19 To further triangulate our findings, given the lack of other rigorously validated pneumonia-specific risk-prediction models for readmission,14 we compared the pneumonia-specific EHR models to the CMS pneumonia model derived from administrative claims data,10 and 2 commonly used risk-prediction scores for short-term mortality among patients with community-acquired pneumonia, the PSI and CURB-65 scores.16 Although derived and validated using patient-level data, the CMS model was developed to benchmark hospitals according to hospital-level readmission rates.10 The CURB-65 score in this study was also modified to include the same altered mental status diagnostic codes according to the modified PSI as a proxy for “confusion.” Both the PSI and CURB-65 scores were calculated using the most abnormal values within the first 24 hours of admission. The ‘updated’ PSI and the ‘updated’ CURB-65 were calculated using the most abnormal values within 24 hours prior to discharge, or the last known observation prior to discharge if no results were recorded within this time period. A complete list of variables for each of the comparison models are shown in Supplemental Table 1.

We assessed model performance by calculating the C statistic, integrated discrimination index, and net reclassification index (NRI) compared to our pneumonia-specific models. The integrated discrimination index is the difference in the mean predicted probability of readmission between patients who were and were not actually readmitted between 2 models, where more positive values suggest improvement in model performance compared to a reference model.26 The NRI is defined as the sum of the net proportions of correctly reclassified persons with and without the event of interest.27 Here, we calculated a category-based NRI to evaluate the performance of pneumonia-specific models in correctly classifying individuals with and without readmissions into the 2 highest readmission risk quintiles vs the lowest 3 risk quintiles compared to other models.27 This pre-specified cutoff is relevant for hospitals interested in identifying the highest risk individuals for targeted intervention.7 Finally, we assessed calibration of comparator models in our cohort by comparing predicted probability to observed probability of readmission by quintiles of risk for each model. We conducted all analyses using Stata 12.1 (StataCorp, College Station, Texas). This study was approved by the University of Texas Southwestern Medical Center Institutional Review Board.

 

 

RESULTS

Of 1463 index hospitalizations (Supplemental Figure 1), the 30-day all-cause readmission rate was 13.6%. Individuals with a 30-day readmission had markedly different sociodemographic and clinical characteristics compared to those not readmitted (Table 1; see Supplemental Table 2 for additional clinical characteristics).

Baseline Characteristics of Patients Hospitalized with Pneumonia
Table 1

Derivation, Validation, and Performance of the Pneumonia-Specific Readmission Risk-Prediction Models

The final first-day pneumonia-specific EHR model included 7 variables, including sociodemographic characteristics; prior hospitalizations; thrombocytosis, and PSI (Table 2). The first-day pneumonia-specific model had adequate discrimination (C statistic, 0.695; optimism-corrected C statistic 0.675, 95% confidence interval [CI], 0.667-0.685; Table 3). It also effectively stratified individuals across a broad range of risk (average predicted decile of risk ranged from 4% to 33%; Table 3) and was well calibrated (Supplemental Table 3).

Final Pneumonia-Specific EHR Risk-Prediction Models for Readmissions
Table 2

The final full-stay pneumonia-specific EHR readmission model included 8 predictors, including 3 variables from the first-day model (median income, thrombocytosis, and prior hospitalizations; Table 2). The full-stay pneumonia-specific EHR model also included vital sign instabilities on discharge, updated PSI, and disposition status (ie, being discharged with home health or to a post-acute care facility was associated with greater odds of readmission, and hospice with lower odds). The full-stay pneumonia-specific EHR model had good discrimination (C statistic, 0.731; optimism-corrected C statistic, 0.714; 95% CI, 0.706-0.720), and stratified individuals across a broad range of risk (average predicted decile of risk ranged from 3% to 37%; Table 3), and was also well calibrated (Supplemental Table 3).

Model Performance and Comparison of Pneumonia-Specific EHR Readmissions Models vs Other Models
Table 3

First-Day Pneumonia-Specific EHR Model vs First-Day Multi-Condition EHR Model

The first-day pneumonia-specific EHR model outperformed the first-day multi-condition EHR model with better discrimination (P = 0.029) and more correctly classified individuals in the top 2 highest risk quintiles vs the bottom 3 risk quintiles (Table 3, Supplemental Table 4, and Supplemental Figure 2A). With respect to calibration, the first-day multi-condition EHR model overestimated risk among the highest quintile risk group compared to the first-day pneumonia-specific EHR model (Figure 1A, 1B).

Comparison of the calibration of different readmission models
Figure 1

Full-Stay Pneumonia-Specific EHR Model vs Other Models

The full-stay pneumonia-specific EHR model comparatively outperformed the corresponding full-stay multi-condition EHR model, as well as the first-day pneumonia-specific EHR model, the CMS pneumonia model, the updated PSI, and the updated CURB-65 (Table 3, Supplemental Table 5, Supplemental Table 6, and Supplemental Figures 2B and 2C). Compared to the full-stay multi-condition and first-day pneumonia-specific EHR models, the full-stay pneumonia-specific EHR model had better discrimination, better reclassification (NRI, 0.09 and 0.08, respectively), and was able to stratify individuals across a broader range of readmission risk (Table 3). It also had better calibration in the highest quintile risk group compared to the full-stay multi-condition EHR model (Figure 1C and 1D).

Updated vs First-Day Modified PSI and CURB-65 Scores

The updated PSI was more strongly predictive of readmission than the PSI calculated on the day of admission (Wald test, 9.83; P = 0.002). Each 10-point increase in the updated PSI was associated with a 22% increased odds of readmission vs an 11% increase for the PSI calculated upon admission (Table 2). The improved predictive ability of the updated PSI and CURB-65 scores was also reflected in the superior discrimination and calibration vs the respective first-day pneumonia severity of illness scores (Table 3).

DISCUSSION

Using routinely available EHR data from 6 diverse hospitals, we developed 2 pneumonia-specific readmission risk-prediction models that aimed to allow hospitals to identify patients hospitalized with pneumonia at high risk for readmission. Overall, we found that a pneumonia-specific model using EHR data from the entire hospitalization outperformed all other models—including the first-day pneumonia-specific model using data present only on admission, our own multi-condition EHR models, and the CMS pneumonia model based on administrative claims data—in all aspects of model performance (discrimination, calibration, and reclassification). We found that socioeconomic status, prior hospitalizations, thrombocytosis, and measures of clinical severity and stability were important predictors of 30-day all-cause readmissions among patients hospitalized with pneumonia. Additionally, an updated discharge PSI score was a stronger independent predictor of readmissions compared to the PSI score calculated upon admission; and inclusion of the updated PSI in our full-stay pneumonia model led to improved prediction of 30-day readmissions.

The marked improvement in performance of the full-stay pneumonia-specific EHR model compared to the first-day pneumonia-specific model suggests that clinical stability and trajectory during hospitalization (as modeled through disposition status, updated PSI, and vital sign instabilities at discharge) are important predictors of 30-day readmission among patients hospitalized for pneumonia, which was not the case for our EHR-based multi-condition models.19 With the inclusion of these measures, the full-stay pneumonia-specific model correctly reclassified an additional 8% of patients according to their true risk compared to the first-day pneumonia-specific model. One implication of these findings is that hospitals interested in targeting their highest risk individuals with pneumonia for transitional care interventions could do so using the first-day pneumonia-specific EHR model and could refine their targeted strategy at the time of discharge by using the full-stay pneumonia model. This staged risk-prediction strategy would enable hospitals to initiate transitional care interventions for high-risk individuals in the inpatient setting (ie, patient education).7 Then, hospitals could enroll both persistent and newly identified high-risk individuals for outpatient interventions (ie, follow-up telephone call) in the immediate post-discharge period, an interval characterized by heightened vulnerability for adverse events,28 based on patients’ illness severity and stability at discharge. This approach can be implemented by hospitals by building these risk-prediction models directly into the EHR, or by extracting EHR data in near real time as our group has done successfully for heart failure.7

Another key implication of our study is that, for pneumonia, a disease-specific modeling approach has better predictive ability than using a multi-condition model. Compared to multi-condition models, the first-day and full-stay pneumonia-specific EHR models correctly reclassified an additional 6% and 9% of patients, respectively. Thus, hospitals interested in identifying the highest risk patients with pneumonia for targeted interventions should do so using the disease-specific models, if the costs and resources of doing so are within reach of the healthcare system.

An additional novel finding of our study is the added value of an updated PSI for predicting adverse events. Studies of pneumonia severity of illness scores have calculated the PSI and CURB-65 scores using data present only on admission.16,24 While our study also confirms that the PSI calculated upon admission is a significant predictor of readmission,23,29 this study extends this work by showing that an updated PSI score calculated at the time of discharge is an even stronger predictor for readmission, and its inclusion in the model significantly improves risk stratification and prognostication.

Our study was noteworthy for several strengths. First, we used data from a common EHR system, thus potentially allowing for the implementation of the pneumonia-specific models in real time across a number of hospitals. The use of routinely collected data for risk-prediction modeling makes this approach scalable and sustainable, because it obviates the need for burdensome data collection and entry. Second, to our knowledge, this is the first study to measure the additive influence of illness severity and stability at discharge on the readmission risk among patients hospitalized with pneumonia. Third, our study population was derived from 6 hospitals diverse in payer status, age, race/ethnicity, and socioeconomic status. Fourth, our models are less likely to be overfit to the idiosyncrasies of our data given that several predictors included in our final pneumonia-specific models have been associated with readmission in this population, including marital status,13,30 income,11,31 prior hospitalizations,11,13 thrombocytosis,32-34 and vital sign instabilities on discharge.17 Lastly, the discrimination of the CMS pneumonia model in our cohort (C statistic, 0.64) closely matched the discrimination observed in 4 independent cohorts (C statistic, 0.63), suggesting adequate generalizability of our study setting and population.10,12

Our results should be interpreted in the context of several limitations. First, generalizability to other regions beyond north Texas is unknown. Second, although we included a diverse cohort of safety net, community, teaching, and nonteaching hospitals, the pneumonia-specific models were not externally validated in a separate cohort, which may lead to more optimistic estimates of model performance. Third, PSI and CURB-65 scores were modified to use diagnostic codes for altered mental status and pleural effusion, and omitted nursing home residence. Thus, the independent associations for the PSI and CURB-65 scores and their predictive ability are likely attenuated. Fourth, we were unable to include data on medications (antibiotics and steroid use) and outpatient visits, which may influence readmission risk.2,9,13,35-40 Fifth, we included only the first pneumonia hospitalization per patient in this study. Had we included multiple hospitalizations per patient, we anticipate better model performance for the 2 pneumonia-specific EHR models since prior hospitalization was a robust predictor of readmission.

In conclusion, the full-stay pneumonia-specific EHR readmission risk-prediction model outperformed the first-day pneumonia-specific model, multi-condition EHR models, and the CMS pneumonia model. This suggests that: measures of clinical severity and stability at the time of discharge are important predictors for identifying patients at highest risk for readmission; and that EHR data routinely collected for clinical practice can be used to accurately predict risk of readmission among patients hospitalized for pneumonia.

 

 

Acknowledgments

The authors would like to acknowledge Ruben Amarasingham, MD, MBA, president and chief executive officer of Parkland Center for Clinical Innovation, and Ferdinand Velasco, MD, chief health information officer at Texas Health Resources for their assistance in assembling the 6-hospital cohort used in this study.

Disclosures

This work was supported by the Agency for Healthcare Research and Quality-funded UT Southwestern Center for Patient-Centered Outcomes Research (R24 HS022418-01); the Commonwealth Foundation (#20100323); the UT Southwestern KL2 Scholars Program supported by the National Institutes of Health (KL2 TR001103 to ANM and OKN); and the National Center for Advancing Translational Sciences at the National Institute of Health (U54 RFA-TR-12-006 to E.A.H.). The study sponsors had no role in design and conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript. The authors have no financial conflicts of interest to disclose

References

1. Centers for Disease Control and Prevention. Pneumonia. http://www.cdc.gov/nchs/fastats/pneumonia.htm. Accessed January 26, 2016.
2. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service program. N Engl J Med. 2009;364(16):1582. PubMed
3. van Walraven C, Bennett C, Jennings A, Austin PC, Forster AJ. Proportion of hospital readmissions deemed avoidable: a systematic review. CMAJ. 2011;183(7):E391-E402. PubMed
4. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital-initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433-440. PubMed
5. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30-day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520-528. PubMed
6. Rennke S, Shoeb MH, Nguyen OK, Magan Y, Wachter RM, Ranji SR. Interventions to Improve Care Transitions at Hospital Discharge. Rockville, MD: Agency for Healthcare Research and Quality, US Department of Health and Human Services;March 2013. PubMed
7. Amarasingham R, Patel PC, Toto K, et al. Allocating scarce resources in real-time to reduce heart failure readmissions: a prospective, controlled study. BMJ Qual Saf. 2013;22(12):998-1005. PubMed
8. Amarasingham R, Patzer RE, Huesch M, Nguyen NQ, Xie B. Implementing electronic health care predictive analytics: considerations and challenges. Health Aff (Millwood). 2014;33(7):1148-1154. PubMed
9. Hebert C, Shivade C, Foraker R, et al. Diagnosis-specific readmission risk prediction using electronic health data: a retrospective cohort study. BMC Med Inform Decis Mak. 2014;14:65. PubMed
10. Lindenauer PK, Normand SL, Drye EE, et al. Development, validation, and results of a measure of 30-day readmission following hospitalization for pneumonia. J Hosp Med. 2011;6(3):142-150. PubMed
11. Mather JF, Fortunato GJ, Ash JL, Davis MJ, Kumar A. Prediction of pneumonia 30-day readmissions: a single-center attempt to increase model performance. Respir Care. 2014;59(2):199-208. PubMed
12. O’Brien WJ, Chen Q, Mull HJ, et al. What is the value of adding Medicare data in estimating VA hospital readmission rates? Health Serv Res. 2015;50(1):40-57. PubMed
13. Tang VL, Halm EA, Fine MJ, Johnson CS, Anzueto A, Mortensen EM. Predictors of rehospitalization after admission for pneumonia in the veterans affairs healthcare system. J Hosp Med. 2014;9(6):379-383. PubMed
14. Weinreich M, Nguyen OK, Wang D, et al. Predicting the risk of readmission in pneumonia: a systematic review of model performance. Ann Am Thorac Soc. 2016;13(9):1607-1614. PubMed
15. Kwok CS, Loke YK, Woo K, Myint PK. Risk prediction models for mortality in community-acquired pneumonia: a systematic review. Biomed Res Int. 2013;2013:504136. PubMed
16. Loke YK, Kwok CS, Niruban A, Myint PK. Value of severity scales in predicting mortality from community-acquired pneumonia: systematic review and meta-analysis. Thorax. 2010;65(10):884-890. PubMed
17. Halm EA, Fine MJ, Kapoor WN, Singer DE, Marrie TJ, Siu AL. Instability on hospital discharge and the risk of adverse outcomes in patients with pneumonia. Arch Intern Med. 2002;162(11):1278-1284. PubMed
18. Amarasingham R, Velasco F, Xie B, et al. Electronic medical record-based multicondition models to predict the risk of 30 day readmission or death among adult medicine patients: validation and comparison to existing models. BMC Med Inform Decis Mak. 2015;15:39. PubMed
19. Nguyen OK, Makam AN, Clark C, et al. Predicting all-cause readmissions using electronic health record data from the entire hospitalization: Model development and comparison. J Hosp Med. 2016;11(7):473-480. PubMed
20. Lindenauer PK, Lagu T, Shieh MS, Pekow PS, Rothberg MB. Association of diagnostic coding with trends in hospitalizations and mortality of patients with pneumonia, 2003-2009. JAMA. 2012;307(13):1405-1413. PubMed
21. Ahmedani BK, Solberg LI, Copeland LA, et al. Psychiatric comorbidity and 30-day readmissions after hospitalization for heart failure, AMI, and pneumonia. Psychiatr Serv. 2015;66(2):134-140. PubMed
22. Jasti H, Mortensen EM, Obrosky DS, Kapoor WN, Fine MJ. Causes and risk factors for rehospitalization of patients hospitalized with community-acquired pneumonia. Clin Infect Dis. 2008;46(4):550-556. PubMed
23. Capelastegui A, España Yandiola PP, Quintana JM, et al. Predictors of short-term rehospitalization following discharge of patients hospitalized with community-acquired pneumonia. Chest. 2009;136(4):1079-1085. PubMed
24. Fine MJ, Auble TE, Yealy DM, et al. A prediction rule to identify low-risk patients with community-acquired pneumonia. N Engl J Med. 1997;336(4):243-250. PubMed
25. Vittinghoff E, Glidden D, Shiboski S, McCulloch C. Regression Methods in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models (Statistics for Biology and Health). New York City, NY: Springer; 2012.
26. Pencina MJ, D’Agostino RB Sr, D’Agostino RB Jr, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27(2):157-172; discussion 207-112. PubMed
27. Leening MJ, Vedder MM, Witteman JC, Pencina MJ, Steyerberg EW. Net reclassification improvement: computation, interpretation, and controversies: a literature review and clinician’s guide. Ann Intern Med. 2014;160(2):122-131. PubMed
28. Krumholz HM. Post-hospital syndrome--an acquired, transient condition of generalized risk. N Engl J Med. 2013;368(2):100-102. PubMed
29. Micek ST, Lang A, Fuller BM, Hampton NB, Kollef MH. Clinical implications for patients treated inappropriately for community-acquired pneumonia in the emergency department. BMC Infect Dis. 2014;14:61. PubMed
30. Metersky ML, Fine MJ, Mortensen EM. The effect of marital status on the presentation and outcomes of elderly male veterans hospitalized for pneumonia. Chest. 2012;142(4):982-987. PubMed
31. Calvillo-King L, Arnold D, Eubank KJ, et al. Impact of social factors on risk of readmission or mortality in pneumonia and heart failure: systematic review. J Gen Intern Med. 2013;28(2):269-282. PubMed
32. Mirsaeidi M, Peyrani P, Aliberti S, et al. Thrombocytopenia and thrombocytosis at time of hospitalization predict mortality in patients with community-acquired pneumonia. Chest. 2010;137(2):416-420. PubMed
33. Prina E, Ferrer M, Ranzani OT, et al. Thrombocytosis is a marker of poor outcome in community-acquired pneumonia. Chest. 2013;143(3):767-775. PubMed

34. Violi F, Cangemi R, Calvieri C. Pneumonia, thrombosis and vascular disease. J Thromb Haemost. 2014;12(9):1391-1400. PubMed
35. Weinberger M, Oddone EZ, Henderson WG. Does increased access to primary care reduce hospital readmissions? Veterans Affairs Cooperative Study Group on Primary Care and Hospital Readmission. N Engl J Med. 1996;334(22):1441-1447. PubMed
36. Field TS, Ogarek J, Garber L, Reed G, Gurwitz JH. Association of early post-discharge follow-up by a primary care physician and 30-day rehospitalization among older adults. J Gen Intern Med. 2015;30(5):565-571. PubMed
37. Spatz ES, Sheth SD, Gosch KL, et al. Usual source of care and outcomes following acute myocardial infarction. J Gen Intern Med. 2014;29(6):862-869. PubMed
38. Brooke BS, Stone DH, Cronenwett JL, et al. Early primary care provider follow-up and readmission after high-risk surgery. JAMA Surg. 2014;149(8):821-828. PubMed
39. Adamuz J, Viasus D, Campreciós-Rodriguez P, et al. A prospective cohort study of healthcare visits and rehospitalizations after discharge of patients with community-acquired pneumonia. Respirology. 2011;16(7):1119-1126. PubMed
40. Shorr AF, Zilberberg MD, Reichley R, et al. Readmission following hospitalization for pneumonia: the impact of pneumonia type and its implication for hospitals. Clin Infect Dis. 2013;57(3):362-367. PubMed

References

1. Centers for Disease Control and Prevention. Pneumonia. http://www.cdc.gov/nchs/fastats/pneumonia.htm. Accessed January 26, 2016.
2. Jencks SF, Williams MV, Coleman EA. Rehospitalizations among patients in the Medicare fee-for-service program. N Engl J Med. 2009;364(16):1582. PubMed
3. van Walraven C, Bennett C, Jennings A, Austin PC, Forster AJ. Proportion of hospital readmissions deemed avoidable: a systematic review. CMAJ. 2011;183(7):E391-E402. PubMed
4. Rennke S, Nguyen OK, Shoeb MH, Magan Y, Wachter RM, Ranji SR. Hospital-initiated transitional care interventions as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):433-440. PubMed
5. Hansen LO, Young RS, Hinami K, Leung A, Williams MV. Interventions to reduce 30-day rehospitalization: a systematic review. Ann Intern Med. 2011;155(8):520-528. PubMed
6. Rennke S, Shoeb MH, Nguyen OK, Magan Y, Wachter RM, Ranji SR. Interventions to Improve Care Transitions at Hospital Discharge. Rockville, MD: Agency for Healthcare Research and Quality, US Department of Health and Human Services;March 2013. PubMed
7. Amarasingham R, Patel PC, Toto K, et al. Allocating scarce resources in real-time to reduce heart failure readmissions: a prospective, controlled study. BMJ Qual Saf. 2013;22(12):998-1005. PubMed
8. Amarasingham R, Patzer RE, Huesch M, Nguyen NQ, Xie B. Implementing electronic health care predictive analytics: considerations and challenges. Health Aff (Millwood). 2014;33(7):1148-1154. PubMed
9. Hebert C, Shivade C, Foraker R, et al. Diagnosis-specific readmission risk prediction using electronic health data: a retrospective cohort study. BMC Med Inform Decis Mak. 2014;14:65. PubMed
10. Lindenauer PK, Normand SL, Drye EE, et al. Development, validation, and results of a measure of 30-day readmission following hospitalization for pneumonia. J Hosp Med. 2011;6(3):142-150. PubMed
11. Mather JF, Fortunato GJ, Ash JL, Davis MJ, Kumar A. Prediction of pneumonia 30-day readmissions: a single-center attempt to increase model performance. Respir Care. 2014;59(2):199-208. PubMed
12. O’Brien WJ, Chen Q, Mull HJ, et al. What is the value of adding Medicare data in estimating VA hospital readmission rates? Health Serv Res. 2015;50(1):40-57. PubMed
13. Tang VL, Halm EA, Fine MJ, Johnson CS, Anzueto A, Mortensen EM. Predictors of rehospitalization after admission for pneumonia in the veterans affairs healthcare system. J Hosp Med. 2014;9(6):379-383. PubMed
14. Weinreich M, Nguyen OK, Wang D, et al. Predicting the risk of readmission in pneumonia: a systematic review of model performance. Ann Am Thorac Soc. 2016;13(9):1607-1614. PubMed
15. Kwok CS, Loke YK, Woo K, Myint PK. Risk prediction models for mortality in community-acquired pneumonia: a systematic review. Biomed Res Int. 2013;2013:504136. PubMed
16. Loke YK, Kwok CS, Niruban A, Myint PK. Value of severity scales in predicting mortality from community-acquired pneumonia: systematic review and meta-analysis. Thorax. 2010;65(10):884-890. PubMed
17. Halm EA, Fine MJ, Kapoor WN, Singer DE, Marrie TJ, Siu AL. Instability on hospital discharge and the risk of adverse outcomes in patients with pneumonia. Arch Intern Med. 2002;162(11):1278-1284. PubMed
18. Amarasingham R, Velasco F, Xie B, et al. Electronic medical record-based multicondition models to predict the risk of 30 day readmission or death among adult medicine patients: validation and comparison to existing models. BMC Med Inform Decis Mak. 2015;15:39. PubMed
19. Nguyen OK, Makam AN, Clark C, et al. Predicting all-cause readmissions using electronic health record data from the entire hospitalization: Model development and comparison. J Hosp Med. 2016;11(7):473-480. PubMed
20. Lindenauer PK, Lagu T, Shieh MS, Pekow PS, Rothberg MB. Association of diagnostic coding with trends in hospitalizations and mortality of patients with pneumonia, 2003-2009. JAMA. 2012;307(13):1405-1413. PubMed
21. Ahmedani BK, Solberg LI, Copeland LA, et al. Psychiatric comorbidity and 30-day readmissions after hospitalization for heart failure, AMI, and pneumonia. Psychiatr Serv. 2015;66(2):134-140. PubMed
22. Jasti H, Mortensen EM, Obrosky DS, Kapoor WN, Fine MJ. Causes and risk factors for rehospitalization of patients hospitalized with community-acquired pneumonia. Clin Infect Dis. 2008;46(4):550-556. PubMed
23. Capelastegui A, España Yandiola PP, Quintana JM, et al. Predictors of short-term rehospitalization following discharge of patients hospitalized with community-acquired pneumonia. Chest. 2009;136(4):1079-1085. PubMed
24. Fine MJ, Auble TE, Yealy DM, et al. A prediction rule to identify low-risk patients with community-acquired pneumonia. N Engl J Med. 1997;336(4):243-250. PubMed
25. Vittinghoff E, Glidden D, Shiboski S, McCulloch C. Regression Methods in Biostatistics: Linear, Logistic, Survival, and Repeated Measures Models (Statistics for Biology and Health). New York City, NY: Springer; 2012.
26. Pencina MJ, D’Agostino RB Sr, D’Agostino RB Jr, Vasan RS. Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond. Stat Med. 2008;27(2):157-172; discussion 207-112. PubMed
27. Leening MJ, Vedder MM, Witteman JC, Pencina MJ, Steyerberg EW. Net reclassification improvement: computation, interpretation, and controversies: a literature review and clinician’s guide. Ann Intern Med. 2014;160(2):122-131. PubMed
28. Krumholz HM. Post-hospital syndrome--an acquired, transient condition of generalized risk. N Engl J Med. 2013;368(2):100-102. PubMed
29. Micek ST, Lang A, Fuller BM, Hampton NB, Kollef MH. Clinical implications for patients treated inappropriately for community-acquired pneumonia in the emergency department. BMC Infect Dis. 2014;14:61. PubMed
30. Metersky ML, Fine MJ, Mortensen EM. The effect of marital status on the presentation and outcomes of elderly male veterans hospitalized for pneumonia. Chest. 2012;142(4):982-987. PubMed
31. Calvillo-King L, Arnold D, Eubank KJ, et al. Impact of social factors on risk of readmission or mortality in pneumonia and heart failure: systematic review. J Gen Intern Med. 2013;28(2):269-282. PubMed
32. Mirsaeidi M, Peyrani P, Aliberti S, et al. Thrombocytopenia and thrombocytosis at time of hospitalization predict mortality in patients with community-acquired pneumonia. Chest. 2010;137(2):416-420. PubMed
33. Prina E, Ferrer M, Ranzani OT, et al. Thrombocytosis is a marker of poor outcome in community-acquired pneumonia. Chest. 2013;143(3):767-775. PubMed

34. Violi F, Cangemi R, Calvieri C. Pneumonia, thrombosis and vascular disease. J Thromb Haemost. 2014;12(9):1391-1400. PubMed
35. Weinberger M, Oddone EZ, Henderson WG. Does increased access to primary care reduce hospital readmissions? Veterans Affairs Cooperative Study Group on Primary Care and Hospital Readmission. N Engl J Med. 1996;334(22):1441-1447. PubMed
36. Field TS, Ogarek J, Garber L, Reed G, Gurwitz JH. Association of early post-discharge follow-up by a primary care physician and 30-day rehospitalization among older adults. J Gen Intern Med. 2015;30(5):565-571. PubMed
37. Spatz ES, Sheth SD, Gosch KL, et al. Usual source of care and outcomes following acute myocardial infarction. J Gen Intern Med. 2014;29(6):862-869. PubMed
38. Brooke BS, Stone DH, Cronenwett JL, et al. Early primary care provider follow-up and readmission after high-risk surgery. JAMA Surg. 2014;149(8):821-828. PubMed
39. Adamuz J, Viasus D, Campreciós-Rodriguez P, et al. A prospective cohort study of healthcare visits and rehospitalizations after discharge of patients with community-acquired pneumonia. Respirology. 2011;16(7):1119-1126. PubMed
40. Shorr AF, Zilberberg MD, Reichley R, et al. Readmission following hospitalization for pneumonia: the impact of pneumonia type and its implication for hospitals. Clin Infect Dis. 2013;57(3):362-367. PubMed

Issue
Journal of Hospital Medicine 12(4)
Issue
Journal of Hospital Medicine 12(4)
Page Number
209-216
Page Number
209-216
Topics
Article Type
Display Headline
Predicting 30-day pneumonia readmissions using electronic health record data
Display Headline
Predicting 30-day pneumonia readmissions using electronic health record data
Sections
Article Source

© 2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Anil N. Makam, MD, MAS; 5323 Harry Hines Blvd., Dallas, TX, 75390-9169; Telephone: 214-648-3272; Fax: 214-648-3232; E-mail: anil.makam@utsouthwestern.edu
Content Gating
Open Access (article Unlocked/Open Access)
Alternative CME
Use ProPublica
Article PDF Media
Media Files

Evaluating automated rules for rapid response system alarm triggers in medical and surgical patients

Article Type
Changed
Wed, 04/26/2017 - 13:40
Display Headline
Evaluating automated rules for rapid response system alarm triggers in medical and surgical patients

Patients typically show signs and symptoms of deterioration hours to days prior to cardiorespiratory arrest.1,2 The rate of inhospital cardiorespiratory arrest (CRA) requiring cardiopulmonary resuscitation is estimated to be 0.174 per bed per year in the United States.3 After CRA, survival to discharge is estimated to be as low as 18%.3,4 Efforts to predict and prevent arrest could prove beneficial.1,2

Rapid response systems (RRS) have been proposed as a means of identifying clinical deterioration and facilitating a timely response. These systems were designed to bring clinicians with critical care expertise to the bedside to prevent unnecessary deaths. They typically include an afferent limb (detects deteriorating patients), an efferent limb (responds to calls and acts to avoid further deterioration), and administrative and data analysis limbs.5,6 Automatic provision of recommendations and computer-based systems are desirable components of the afferent limb of the detection system.6 Both are independent predictors of improved clinical practices for clinical decision support systems.7 However, the existing early warning scores (EWS) may not be ready for automation due to low positive predictive values (PPV) and sensitivities.8

It is possible that the low discriminatory accuracy of the published EWS may be secondary to the use of aggregate patient populations for derivation of scores. We hypothesized that these EWS perform differently in medical and in surgical subpopulations. Also, the EWS need to be tested in a time-dependent manner to serve as a realistic clinical support tool for hospitalized patients.

STUDY AIM

The aim of this study was to evaluate the differential performance of widely used EWS in medical vs surgical patients.

METHODS

Site

The study was conducted in an academic center with 2 hospitals in Southeastern Minnesota totaling approximately 1500 general care nonintensive care unit (ICU) beds. The Mayo Clinic Institutional Review Board approved the research proposal.

Subjects

Our retrospective cohort was comprised of all adult inpatients discharged from 2 academic hospitals between January 1, 2011 and December 31, 2011 who spent any time in a general care (non-ICU) unit. We excluded patients younger than 18 years, psychiatric or rehabilitation inpatients, those without research authorization, and patients admitted for research purposes.

Study patients were divided into medical and surgical cohorts. Hospitalizations were considered surgical if patients had surgery at any time during their hospital stay according to billing data. A trigger was an instance in which a patient met the conditions of a specific rule (score/vital sign exceeded the published/defined threshold).

A resuscitation call was defined as a call for cardiopulmonary resuscitation when a patient has a CRA.

An event was an occurrence of 1 of the following in a general care setting: unplanned transfer to the ICU, resuscitation call, or RRS activation.

The RRS activation criteria consisted of an “acute and persistent change” in any 1 or more of the following: oxygen saturations less than 90%, heart rate less than 40 or greater than 130 beats/minute, systolic blood pressure less than 90 mm Hg, or respiratory rate less than 10 or greater than 28 breaths/minute. The RRS activation requires health provider action; they are not electronically generated. Nurses and physicians may also activate the RRS if they are concerned about a patient, even if calling criteria are not met. This is in contrast to the EWS analyzed, which are aggregate composites of multiple parameters. However, whether or not a derangement in vital signs is considered an “acute and persistent change” still involves clinical judgment. Any movement from a general care bed to an ICU bed, or from a general care bed to a procedure area, and from there to an ICU, was considered unplanned. Transfers to the ICU directly from the emergency department or operating room (OR) were not considered as an unplanned transfer and were not included in the analyses.

Coverage time was the period observed for events after a rule was triggered. In this analysis, a coverage time of 24 hours was considered, with a 1-hour look-back. A trigger was counted as a true positive if an event occurred during the following 24 hours. The 1-hour look-back was included to take into account the nursing clinical process of prioritizing a call to the RRS followed by documentation of the altered vital signs that prompted the call.

An episode was the continuous time on the general care floor within a hospitalization, excluding times when a patient was in the OR or ICU. For example, if a patient was admitted to a general bed on a surgery floor, subsequently went to the OR, and then returned to the surgery floor, the 2 episodes were considered separate: the time on the floor before surgery, and the time on the floor after surgery.

Assessment of implementation of RRS in our hospitals showed a significant drop in the failure-to-rescue rate (issues considered related to delay or failure to identify or intervene appropriately when a patient was deteriorating, as identified through mortality review) and a decrease in non-ICU mortality.9,10 This suggests that our current process captures many of the relevant episodes of acute deterioration when a rapid response team is needed and supports using RRS activation as outcomes.

 

 

Data Sources

We developed a time-stamped longitudinal database of patient data from the electronic health record, including vital signs, laboratory test results, demographics (age, sex), administrative data (including length of stay), comorbidities, resuscitation code status, location in hospital, and at the minute level throughout each patient’s hospital stay. Physiologically impossible values (eg, blood pressures of 1200 mm Hg) were considered entered in error and eliminated from the database. Time spent in the OR or ICU was excluded because RRS activation would not be applied in these already highly monitored areas. SAS Statistical software (SAS Institute Inc. Cary, North Carolina) was used for database creation.

We applied the current RRS calling criteria in our institution and calculated the Kirkland score,11 along with some of the most widely used early warning scores:12 Modified Early Warning System (MEWS),13 Standardized Early Warning Scoring System (SEWS),14 Global Modified Early Warning Score (GMEWS),15 Worthing physiologic scoring system,16 National Early Warning Score (NEWS),17 and VitaPAC Early Warning Score (ViEWS).18 Published thresholds for these scores were used to create rule triggers in the data. Once a trigger was created to calculate the number of false positives and true positives, all subsequent triggers were ignored until the end of the episode or until 24 hours elapsed. We calculated triggers in a rolling fashion throughout the episodes of care. The EWS score was updated every time a new parameter was entered into the analytical electronic health record, and the most recent value for each was used to calculate the score. SAS statistical software was used for calculation of scores and identification of outcomes.

For our analysis, events were treated as dependent variables, and triggers were independent variables. We calculated the score for each EWS to the minute level throughout our retrospective database. If the score for a specific EWS was higher than the published/recommended threshold for that EWS, an alert was considered to have been issued, and the patient was followed for 24 hours. If the patient had an event in the subsequent 24 hours, or 1 hour before (1-hour look-back), the alert was considered a true positive; if not, a false positive. Events that were not preceded by an alert were false negatives, and 24-hour intervals without either an alert or an event were considered true negatives. This simulation exercise was performed for each EWS in both subcohorts (medical and surgical). Clusters of RRS calls followed by transfers to the ICU within 3 hours were considered as a single adverse event (RRS calls, as it was the first event to occur) to avoid double counting. We have described how well this simulation methodology,8 correlates with results from prospective studies.19

Statistical Analysis

To calculate whether results were statistically significant for subgroups, a jackknife method of calculating variance20 was used. The jackknife method calculates variance by repeating the calculations of the statistic leaving out 1 sample at a time. In our case, we repeated the calculation of sensitivity and PPV leaving out 1 patient at a time. Once the simulation method had been run and the false/true positives/negatives had been assigned, calculation of each metric (PPV and sensitivity) was repeated for n subsamples, each leaving out 1 patient. The variance was calculated and 2 Student t tests were performed for each EWS: 1 for PPV and another for sensitivity. SAS statistical software v 9.3 was used for the simulation analysis; R statistical software v 3.0.2 (The R Foundation, Vienna, Austria) was used for the calculation of the statistical significance of results. A univariable analysis was also performed to assess the sensitivity and PPVs for the published thresholds of the most common variables in each EWS: respiratory rate, systolic blood pressure, heart rate, temperature, and mental status as measured by the modified Richmond Agitation Sedation Score.21

RESULTS

The initial cohort included 60,020 hospitalizations, of which the following were excluded: 2751 because of a lack of appropriate research authorization; 6433 because the patients were younger than 18 years; 2129 as psychiatric admissions; 284 as rehabilitation admissions; 872 as research purposes-only admissions; and 1185 because the patient was never in a general care bed (eg, they were either admitted directly to the ICU, or they were admitted for an outpatient surgical procedure and spent time in the postanesthesia care unit).

Table 1 summarizes patient and trigger characteristics, overall and by subgroup. The final cohort included 75,240 total episodes in 46,366 hospitalizations, from 34,898 unique patients, of which 48.7% were male. There were 23,831 medical and 22,535 surgical hospitalizations. Median length of episode was 2 days both for medical and surgical patients. Median length of stay was 3 days, both for medical and for surgical patients.

Patient Characteristics, Events, and Triggers
Table 1


There were 3332 events in total, of which 1709 were RRS calls, 185 were resuscitation calls, and 1438 were unscheduled transfers to the ICU. The rate of events was 4.67 events per 100 episodes in the aggregate adult population. There were 3.93 events per 100 episodes for surgical hospitalizations, and 5.86 events per 100 episodes for medical hospitalizations (P < .001). The number of CRAs in our cohort was 0.27 per 100 episodes, 0.128 per hospital bed per year, or 4.37 per 1000 hospital admissions, similar to other reported numbers in the literature.3, 22,23

The total number of EWS triggers varied greatly between EWS rules, with the volume ranging during the study year from 1363 triggers with the GMEWS rule to 77,711 triggers with the ViEWS score.

Performance of scores in medical and surgical patients
Figure


All scores had PPVs less than 25%. As seen in Table 2 and shown graphically in the Figure, all scores performed better on medical patients (blue) than on surgical patients (yellow). The P value was < .0001 for both PPV and sensitivity. The Worthing score had the highest sensitivity (0.78 for medical and 0.68 for surgical) but a very low PPV (0.04 for medical and 0.03 for surgical), while GMEWS was the opposite: low sensitivity (0.10 and 0.07) but the highest PPV (0.22 and 0.18).

Comparison of the Predictive Performance of Widely Used EWS in a Surgical and a Medical Population
Table 2


The results of the univariable analysis can be seen in Table 3. Most of the criteria performed better (higher sensitivity and PPV) as predictors in the medical hospitalizations than in the surgical hospitalizations.

Univariable Analysis in the Medical and Surgical Subpopulations
Table 3

 

 

DISCUSSION

We hypothesized that EWS may perform differently when applied to medical rather than surgical patients. Studies had not analyzed this in a time-dependent manner,24-26 which limited the applicability of the results.8

All analyzed scores performed better in medical patients than in surgical patients (Figure). This could reflect a behavioral difference by the teams on surgical and medical floors in the decision to activate the RRS, or a bias of the clinicians who designed the scores (mostly nonsurgeons). The difference could also mean that physiological deteriorations are intrinsically different in patients who have undergone anesthesia and surgery. For example, in surgical patients, a bleeding episode is more likely to be the cause of their physiological deterioration, or the lingering effects of anesthesia could mask underlying deterioration. Such patients would benefit from scores where variables such as heart rate, blood pressure, or hemoglobin had more influence.

When comparing the different scores, it was much easier for a patient to meet the alerting score with the Worthing score than with GMEWS. In the Worthing score, a respiratory rate greater than 22 breaths per minute, or a systolic blood pressure less than 100 mm Hg, already meet alerting criteria. Similar vital signs result in 0 and 1 points (respectively) in GMEWS, far from its alerting score of 5. This reflects the intrinsic tradeoff of EWS: as the threshold for considering a patient “at risk” drops, not only does the number of true positives (and the sensitivity) increase, but also the number of false positives, thus lowering the PPV.

However, none of the scores analyzed were considered to perform well based on their PPV and sensitivity, particularly in the surgical subpopulation. Focusing on another metric, the area under the receiver operator curve can give misleadingly optimistic results.24,27 However, the extremely low prevalence of acute physiological deterioration can produce low PPVs even when specificity seems acceptable, which is why it is important to evaluate PPV directly.28

To use EWS effectively to activate RRS, they need to be combined with clinical judgment to avoid high levels of false alerts, particularly in surgical patients. It has been reported that RRS is activated only 30% of the time a patient meets RRS calling criteria.29 While there may be cultural characteristics inhibiting the decision to call,30 our study hints at another explanation: if RRS was activated every time a patient met calling criteria based on the scores analyzed, the number of RRS calls would be very high and difficult to manage. So health providers may be doing the right thing when “filtering” RRS calls and not applying the criteria strictly, but in conjunction with clinical judgment.

A limitation of any study like this is how to define “acute physiological deterioration.” We defined an event as recognized episodes of acute physiological deterioration that are signaled by escalations of care (eg, RRS, resuscitation calls, or transfers to an ICU) or unexpected death. By definition, our calculated PPV is affected by clinicians’ recognition of clinical deteriorations. This definition, common in the literature, has the limitation of potentially underestimating EWS’ performance by missing some events that are resolved by the primary care team without an escalation of care. However, we believe our interpretation is not unreasonable since the purpose of EWS is to trigger escalations of care in a timely fashion. Prospective studies could define an event in a way that is less affected by the clinicians’ judgment.

Regarding patient demographics, age was similar between the 2 groups (average, 58.2 years for medical vs 58.9 years for surgical), and there was only a small difference in gender ratios (45.1% male in the medical vs 51.4% in the surgical group). These differences are unlikely to have affected the results significantly, but unknown differences in demographics or other patient characteristics between groups may account for differences in score performance between surgical and medical patients.

Several of the EWS analyzed had overlapping trigger criteria with our own RRS activation criteria (although as single-parameter triggers and not as aggregate). To test how these potential biases could affect our results, we performed a post hoc sensitivity analysis eliminating calls to the RRS as an outcome (so using the alternative outcome of unexpected transfers to the ICU and resuscitation calls). The results are similar to those of our main analysis, with all analyzed scores having lower sensitivity and PPV in surgical hospitalizations when compared to medical hospitalizations.

Our study suggests that, to optimize detection of physiological deterioration events, EWS should try to take into account different patient types, with the most basic distinction being surgical vs medical. This tailoring will make EWS more complex, and less suited for paper-based calculation, but new electronic health records are increasingly able to incorporate decision support, and some EWS have been developed for electronic calculation only. Of particular interest in this regard is the score developed by Escobar et al,31 which groups patients into categories according to the reason for admission, and calculates a different subscore based on that category. While the score by Escobar et al. does not split patients based on medical or surgical status, a more general interpretation of our results suggests that a score may be more accurate if it classifies patients into subgroups with different subscores. This seems to be confirmed by the fact that the score by Escobar et al performs better than MEWS.28 Unfortunately, the paper describing it does not provide enough detail to use it in our database.

A recent systematic review showed increasing evidence that RRS may be effective in reducing CRAs occurring in a non-ICU setting and, more important, overall inhospital mortality.32 While differing implementation strategies (eg, different length of the educational effort, changes in the frequency of vital signs monitoring) can impact the success of such an initiative, it has been speculated that the afferent limb (which often includes an EWS) might be the most critical part of the system.33 Our results show that the most widely used EWS perform significantly worse on surgical patients, and suggest that a way to improve the accuracy of EWS would be to tailor the risk calculation to different patient subgroups (eg, medical and surgical patients). Plausible next steps would be to demonstrate that tailoring risk calculation to medical and surgical patients separately can improve risk predictions and accuracy of EWS.

 

 

Disclosure

The authors report no financial conflicts of interest.

References

1. Buist MD, Jarmolowski E, Burton PR, Bernard SA, Waxman BP, Anderson J. Recognising clinical instability in hospital patients before cardiac arrest or unplanned admission to intensive care. A pilot study in a tertiary-care hospital. Med J Aust. 1999; 171(1):22-25. PubMed
2. Schein RM, Hazday N, Pena M, Ruben BH, Sprung CL. Clinical antecedents to in-hospital cardiopulmonary arrest. Chest. 1990;98(6):1388-1392. PubMed
3. Peberdy MA, Kaye W, Ornato JP, Larkin GL, Nadkarni V, Mancini ME, et al. Cardiopulmonary resuscitation of adults in the hospital: a report of 14720 cardiac arrests from the National Registry of Cardiopulmonary Resuscitation. Resuscitation. 2003; 58(3):297-308. PubMed
4. Nadkarni VM, Larkin GL, Peberdy MA, Carey SM, Kaye W, Mancini ME, et al. First documented rhythm and clinical outcome from in-hospital cardiac arrest among children and adults. JAMA. 2006;295(1):50-57. PubMed
5. Devita MA, Bellomo R, Hillman K, Kellum J, Rotondi A, Teres D, et al. Findings of the first consensus conference on medical emergency teams. Crit Care Med. 2006;34(9):2463-2478. PubMed
6. DeVita MA, Smith GB, Adam SK, Adams-Pizarro I, Buist M, Bellomo R, et al. “Identifying the hospitalised patient in crisis”--a consensus conference on the afferent limb of rapid response systems. Resuscitation. 2010;81(4):375-382. PubMed
7. Kawamoto K, Houlihan CA, Balas EA, Lobach DF. Improving clinical practice using clinical decision support systems: a systematic review of trials to identify features critical to success. BMJ. 2005;330(7494):765. PubMed
8. Romero-Brufau S, Huddleston JM, Naessens JM, Johnson MG, Hickman J, Morlan BW, et al. Widely used track and trigger scores: are they ready for automation in practice? Resuscitation. 2014;85(4):549-552. PubMed
9. Huddleston JM, Diedrich DA, Kinsey GC, Enzler MJ, Manning DM. Learning from every death. J Patient Saf. 2014;10(1):6-12. PubMed
10. Moriarty JP, Schiebel NE, Johnson MG, Jensen JB, Caples SM, Morlan BW, et al. Evaluating implementation of a rapid response team: considering alternative outcome measures. Int J Qual Health Care. 2014;26(1):49-57. PubMed
11. Kirkland LL, Malinchoc M, O’Byrne M, Benson JT, Kashiwagi DT, Burton MC, et al. A clinical deterioration prediction tool for internal medicine patients. Am J Med Qual. 2013;28(2):135-142. PubMed
12. Griffiths JR, Kidney EM. Current use of early warning scores in UK emergency departments. Emerg Med J. 2012;29(1):65-66. PubMed
13. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521-526. PubMed
14. Paterson R, MacLeod DC, Thetford D, Beattie A, Graham C, Lam S, et al.. Prediction of in-hospital mortality and length of stay using an early warning scoring system: clinical audit. Clin Med (Lond). 2006;6(3):281-284. PubMed
15. Harrison GA, Jacques T, McLaws ML, Kilborn G. Combinations of early signs of critical illness predict in-hospital death–the SOCCER study (signs of critical conditions and emergency responses). Resuscitation. 2006;71(3):327-334. PubMed
16. Duckitt RW, Buxton-Thomas R, Walker J, Cheek E, Bewick V, Venn R, et al. Worthing physiological scoring system: derivation and validation of a physiological early-warning system for medical admissions. An observational, population-based single-centre study. Br J Anaesth. 2007; 98(6):769-774. PubMed
17. Smith GB, Prytherch DR, Meredith P, Schmidt PE, Featherstone PI. The ability of the National Early Warning Score (NEWS) to discriminate patients at risk of early cardiac arrest, unanticipated intensive care unit admission, and death. Resuscitation. 2013;84(4):465-470. PubMed
18. Prytherch DR, Smith GB, Schmidt PE, Featherstone PI. ViEWS--Towards a national early warning score for detecting adult inpatient deterioration. Resuscitation. 2010;81(8):932-937. PubMed
19. Romero-Brufau S, Huddleston JM. Reply to letter: widely used track and trigger scores: are they ready for automation in practice? Resuscitation. 2014;85(10):e159. PubMed
20. Efron B, Stein C. The jackknife estimate of variance. Annals of Statistics. 1981;586-596. 
21. Sessler CN, Gosnell MS, Grap MJ, Brophy GM, O’Neal PV, Keane KA, et al. The Richmond Agitation-Sedation Scale: validity and reliability in adult intensive care unit patients. Am J Respir Crit Care Med. 2002;166(10):1338-1344. PubMed
22. DeVita MA, Braithwaite RS, Mahidhara R, Stuart S, Foraida M, Simmons RL. Medical Emergency Response Improvement Team (MERIT). Use of medical emergency team responses to reduce hospital cardiopulmonary arrests. Qual Saf Health Care. 2004;13(4):251-254. PubMed
23. Goncales PD, Polessi JA, Bass LM, Santos Gde P, Yokota PK, Laselva CR, et al. Reduced frequency of cardiopulmonary arrests by rapid response teams. Einstein (Sao Paulo). 2012;10(4):442-448. PubMed
24. Cuthbertson BH, Boroujerdi M, McKie L, Aucott L, Prescott G. Can physiological variables and early warning scoring systems allow early recognition of the deteriorating surgical patient? Crit Care Med. 2007;35(2):402-409. PubMed
25. Gardner-Thorpe J, Love N, Wrightson J, Walsh S, Keeling N. The value of Modified Early Warning Score (MEWS) in surgical in-patients: a prospective observational study. Ann R Coll Surg Engl. 2006;88(6):571-575. PubMed
26. Stenhouse C, Coates S, Tivey M, Allsop P, Parker T. Prospective evaluation of a modified Early Warning Score to aid earlier detection of patients developing critical illness on a general surgical ward. British Journal of Anaesthesia. 2000;84(5):663-663. 
27. Smith GB, Prytherch DR, Schmidt PE, Featherstone PI. Review and performance evaluation of aggregate weighted ‘track and trigger’ systems. Resuscitation. 2008;77(2):170-179. PubMed
28. Romero-Brufau S, Huddleston JM, Escobar GJ, Liebow M. Why the C-statistic is not informative to evaluate early warning scores and what metrics to use. Crit Care. 2015; 19:285. PubMed
29. Hillman K, Chen J, Cretikos M, Bellomo R, Brown D, Doig G, et al. Introduction of the medical emergency team (MET) system: a cluster-randomised controlled trial. Lancet. 2005;365(9477):2091-2097. PubMed
30. Shearer B, Marshall S, Buist MD, Finnigan M, Kitto S, Hore T, et al. What stops hospital clinical staff from following protocols? An analysis of the incidence and factors behind the failure of bedside clinical staff to activate the rapid response system in a multi-campus Australian metropolitan healthcare service. BMJ Qual Saf. 2012;21(7):569-575. PubMed
31. Escobar GJ, LaGuardia JC, Turk BJ, Ragins A, Kipnis P, Draper D. Early detection of impending physiologic deterioration among patients who are not in intensive care: development of predictive models using data from an automated electronic medical record. J Hosp Med. 2012;7(5):388-395. PubMed
32. Winters BD, Weaver SJ, Pfoh ER, Yang T, Pham JC, Dy SM. Rapid-response systems as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):417-425. PubMed
33. Jones DA, DeVita MA, Bellomo R. Rapid-response teams. N Engl J Med. 2011;365(2):139-146. PubMed

Article PDF
Issue
Journal of Hospital Medicine 12(4)
Topics
Page Number
217-223
Sections
Article PDF
Article PDF

Patients typically show signs and symptoms of deterioration hours to days prior to cardiorespiratory arrest.1,2 The rate of inhospital cardiorespiratory arrest (CRA) requiring cardiopulmonary resuscitation is estimated to be 0.174 per bed per year in the United States.3 After CRA, survival to discharge is estimated to be as low as 18%.3,4 Efforts to predict and prevent arrest could prove beneficial.1,2

Rapid response systems (RRS) have been proposed as a means of identifying clinical deterioration and facilitating a timely response. These systems were designed to bring clinicians with critical care expertise to the bedside to prevent unnecessary deaths. They typically include an afferent limb (detects deteriorating patients), an efferent limb (responds to calls and acts to avoid further deterioration), and administrative and data analysis limbs.5,6 Automatic provision of recommendations and computer-based systems are desirable components of the afferent limb of the detection system.6 Both are independent predictors of improved clinical practices for clinical decision support systems.7 However, the existing early warning scores (EWS) may not be ready for automation due to low positive predictive values (PPV) and sensitivities.8

It is possible that the low discriminatory accuracy of the published EWS may be secondary to the use of aggregate patient populations for derivation of scores. We hypothesized that these EWS perform differently in medical and in surgical subpopulations. Also, the EWS need to be tested in a time-dependent manner to serve as a realistic clinical support tool for hospitalized patients.

STUDY AIM

The aim of this study was to evaluate the differential performance of widely used EWS in medical vs surgical patients.

METHODS

Site

The study was conducted in an academic center with 2 hospitals in Southeastern Minnesota totaling approximately 1500 general care nonintensive care unit (ICU) beds. The Mayo Clinic Institutional Review Board approved the research proposal.

Subjects

Our retrospective cohort was comprised of all adult inpatients discharged from 2 academic hospitals between January 1, 2011 and December 31, 2011 who spent any time in a general care (non-ICU) unit. We excluded patients younger than 18 years, psychiatric or rehabilitation inpatients, those without research authorization, and patients admitted for research purposes.

Study patients were divided into medical and surgical cohorts. Hospitalizations were considered surgical if patients had surgery at any time during their hospital stay according to billing data. A trigger was an instance in which a patient met the conditions of a specific rule (score/vital sign exceeded the published/defined threshold).

A resuscitation call was defined as a call for cardiopulmonary resuscitation when a patient has a CRA.

An event was an occurrence of 1 of the following in a general care setting: unplanned transfer to the ICU, resuscitation call, or RRS activation.

The RRS activation criteria consisted of an “acute and persistent change” in any 1 or more of the following: oxygen saturations less than 90%, heart rate less than 40 or greater than 130 beats/minute, systolic blood pressure less than 90 mm Hg, or respiratory rate less than 10 or greater than 28 breaths/minute. The RRS activation requires health provider action; they are not electronically generated. Nurses and physicians may also activate the RRS if they are concerned about a patient, even if calling criteria are not met. This is in contrast to the EWS analyzed, which are aggregate composites of multiple parameters. However, whether or not a derangement in vital signs is considered an “acute and persistent change” still involves clinical judgment. Any movement from a general care bed to an ICU bed, or from a general care bed to a procedure area, and from there to an ICU, was considered unplanned. Transfers to the ICU directly from the emergency department or operating room (OR) were not considered as an unplanned transfer and were not included in the analyses.

Coverage time was the period observed for events after a rule was triggered. In this analysis, a coverage time of 24 hours was considered, with a 1-hour look-back. A trigger was counted as a true positive if an event occurred during the following 24 hours. The 1-hour look-back was included to take into account the nursing clinical process of prioritizing a call to the RRS followed by documentation of the altered vital signs that prompted the call.

An episode was the continuous time on the general care floor within a hospitalization, excluding times when a patient was in the OR or ICU. For example, if a patient was admitted to a general bed on a surgery floor, subsequently went to the OR, and then returned to the surgery floor, the 2 episodes were considered separate: the time on the floor before surgery, and the time on the floor after surgery.

Assessment of implementation of RRS in our hospitals showed a significant drop in the failure-to-rescue rate (issues considered related to delay or failure to identify or intervene appropriately when a patient was deteriorating, as identified through mortality review) and a decrease in non-ICU mortality.9,10 This suggests that our current process captures many of the relevant episodes of acute deterioration when a rapid response team is needed and supports using RRS activation as outcomes.

 

 

Data Sources

We developed a time-stamped longitudinal database of patient data from the electronic health record, including vital signs, laboratory test results, demographics (age, sex), administrative data (including length of stay), comorbidities, resuscitation code status, location in hospital, and at the minute level throughout each patient’s hospital stay. Physiologically impossible values (eg, blood pressures of 1200 mm Hg) were considered entered in error and eliminated from the database. Time spent in the OR or ICU was excluded because RRS activation would not be applied in these already highly monitored areas. SAS Statistical software (SAS Institute Inc. Cary, North Carolina) was used for database creation.

We applied the current RRS calling criteria in our institution and calculated the Kirkland score,11 along with some of the most widely used early warning scores:12 Modified Early Warning System (MEWS),13 Standardized Early Warning Scoring System (SEWS),14 Global Modified Early Warning Score (GMEWS),15 Worthing physiologic scoring system,16 National Early Warning Score (NEWS),17 and VitaPAC Early Warning Score (ViEWS).18 Published thresholds for these scores were used to create rule triggers in the data. Once a trigger was created to calculate the number of false positives and true positives, all subsequent triggers were ignored until the end of the episode or until 24 hours elapsed. We calculated triggers in a rolling fashion throughout the episodes of care. The EWS score was updated every time a new parameter was entered into the analytical electronic health record, and the most recent value for each was used to calculate the score. SAS statistical software was used for calculation of scores and identification of outcomes.

For our analysis, events were treated as dependent variables, and triggers were independent variables. We calculated the score for each EWS to the minute level throughout our retrospective database. If the score for a specific EWS was higher than the published/recommended threshold for that EWS, an alert was considered to have been issued, and the patient was followed for 24 hours. If the patient had an event in the subsequent 24 hours, or 1 hour before (1-hour look-back), the alert was considered a true positive; if not, a false positive. Events that were not preceded by an alert were false negatives, and 24-hour intervals without either an alert or an event were considered true negatives. This simulation exercise was performed for each EWS in both subcohorts (medical and surgical). Clusters of RRS calls followed by transfers to the ICU within 3 hours were considered as a single adverse event (RRS calls, as it was the first event to occur) to avoid double counting. We have described how well this simulation methodology,8 correlates with results from prospective studies.19

Statistical Analysis

To calculate whether results were statistically significant for subgroups, a jackknife method of calculating variance20 was used. The jackknife method calculates variance by repeating the calculations of the statistic leaving out 1 sample at a time. In our case, we repeated the calculation of sensitivity and PPV leaving out 1 patient at a time. Once the simulation method had been run and the false/true positives/negatives had been assigned, calculation of each metric (PPV and sensitivity) was repeated for n subsamples, each leaving out 1 patient. The variance was calculated and 2 Student t tests were performed for each EWS: 1 for PPV and another for sensitivity. SAS statistical software v 9.3 was used for the simulation analysis; R statistical software v 3.0.2 (The R Foundation, Vienna, Austria) was used for the calculation of the statistical significance of results. A univariable analysis was also performed to assess the sensitivity and PPVs for the published thresholds of the most common variables in each EWS: respiratory rate, systolic blood pressure, heart rate, temperature, and mental status as measured by the modified Richmond Agitation Sedation Score.21

RESULTS

The initial cohort included 60,020 hospitalizations, of which the following were excluded: 2751 because of a lack of appropriate research authorization; 6433 because the patients were younger than 18 years; 2129 as psychiatric admissions; 284 as rehabilitation admissions; 872 as research purposes-only admissions; and 1185 because the patient was never in a general care bed (eg, they were either admitted directly to the ICU, or they were admitted for an outpatient surgical procedure and spent time in the postanesthesia care unit).

Table 1 summarizes patient and trigger characteristics, overall and by subgroup. The final cohort included 75,240 total episodes in 46,366 hospitalizations, from 34,898 unique patients, of which 48.7% were male. There were 23,831 medical and 22,535 surgical hospitalizations. Median length of episode was 2 days both for medical and surgical patients. Median length of stay was 3 days, both for medical and for surgical patients.

Patient Characteristics, Events, and Triggers
Table 1


There were 3332 events in total, of which 1709 were RRS calls, 185 were resuscitation calls, and 1438 were unscheduled transfers to the ICU. The rate of events was 4.67 events per 100 episodes in the aggregate adult population. There were 3.93 events per 100 episodes for surgical hospitalizations, and 5.86 events per 100 episodes for medical hospitalizations (P < .001). The number of CRAs in our cohort was 0.27 per 100 episodes, 0.128 per hospital bed per year, or 4.37 per 1000 hospital admissions, similar to other reported numbers in the literature.3, 22,23

The total number of EWS triggers varied greatly between EWS rules, with the volume ranging during the study year from 1363 triggers with the GMEWS rule to 77,711 triggers with the ViEWS score.

Performance of scores in medical and surgical patients
Figure


All scores had PPVs less than 25%. As seen in Table 2 and shown graphically in the Figure, all scores performed better on medical patients (blue) than on surgical patients (yellow). The P value was < .0001 for both PPV and sensitivity. The Worthing score had the highest sensitivity (0.78 for medical and 0.68 for surgical) but a very low PPV (0.04 for medical and 0.03 for surgical), while GMEWS was the opposite: low sensitivity (0.10 and 0.07) but the highest PPV (0.22 and 0.18).

Comparison of the Predictive Performance of Widely Used EWS in a Surgical and a Medical Population
Table 2


The results of the univariable analysis can be seen in Table 3. Most of the criteria performed better (higher sensitivity and PPV) as predictors in the medical hospitalizations than in the surgical hospitalizations.

Univariable Analysis in the Medical and Surgical Subpopulations
Table 3

 

 

DISCUSSION

We hypothesized that EWS may perform differently when applied to medical rather than surgical patients. Studies had not analyzed this in a time-dependent manner,24-26 which limited the applicability of the results.8

All analyzed scores performed better in medical patients than in surgical patients (Figure). This could reflect a behavioral difference by the teams on surgical and medical floors in the decision to activate the RRS, or a bias of the clinicians who designed the scores (mostly nonsurgeons). The difference could also mean that physiological deteriorations are intrinsically different in patients who have undergone anesthesia and surgery. For example, in surgical patients, a bleeding episode is more likely to be the cause of their physiological deterioration, or the lingering effects of anesthesia could mask underlying deterioration. Such patients would benefit from scores where variables such as heart rate, blood pressure, or hemoglobin had more influence.

When comparing the different scores, it was much easier for a patient to meet the alerting score with the Worthing score than with GMEWS. In the Worthing score, a respiratory rate greater than 22 breaths per minute, or a systolic blood pressure less than 100 mm Hg, already meet alerting criteria. Similar vital signs result in 0 and 1 points (respectively) in GMEWS, far from its alerting score of 5. This reflects the intrinsic tradeoff of EWS: as the threshold for considering a patient “at risk” drops, not only does the number of true positives (and the sensitivity) increase, but also the number of false positives, thus lowering the PPV.

However, none of the scores analyzed were considered to perform well based on their PPV and sensitivity, particularly in the surgical subpopulation. Focusing on another metric, the area under the receiver operator curve can give misleadingly optimistic results.24,27 However, the extremely low prevalence of acute physiological deterioration can produce low PPVs even when specificity seems acceptable, which is why it is important to evaluate PPV directly.28

To use EWS effectively to activate RRS, they need to be combined with clinical judgment to avoid high levels of false alerts, particularly in surgical patients. It has been reported that RRS is activated only 30% of the time a patient meets RRS calling criteria.29 While there may be cultural characteristics inhibiting the decision to call,30 our study hints at another explanation: if RRS was activated every time a patient met calling criteria based on the scores analyzed, the number of RRS calls would be very high and difficult to manage. So health providers may be doing the right thing when “filtering” RRS calls and not applying the criteria strictly, but in conjunction with clinical judgment.

A limitation of any study like this is how to define “acute physiological deterioration.” We defined an event as recognized episodes of acute physiological deterioration that are signaled by escalations of care (eg, RRS, resuscitation calls, or transfers to an ICU) or unexpected death. By definition, our calculated PPV is affected by clinicians’ recognition of clinical deteriorations. This definition, common in the literature, has the limitation of potentially underestimating EWS’ performance by missing some events that are resolved by the primary care team without an escalation of care. However, we believe our interpretation is not unreasonable since the purpose of EWS is to trigger escalations of care in a timely fashion. Prospective studies could define an event in a way that is less affected by the clinicians’ judgment.

Regarding patient demographics, age was similar between the 2 groups (average, 58.2 years for medical vs 58.9 years for surgical), and there was only a small difference in gender ratios (45.1% male in the medical vs 51.4% in the surgical group). These differences are unlikely to have affected the results significantly, but unknown differences in demographics or other patient characteristics between groups may account for differences in score performance between surgical and medical patients.

Several of the EWS analyzed had overlapping trigger criteria with our own RRS activation criteria (although as single-parameter triggers and not as aggregate). To test how these potential biases could affect our results, we performed a post hoc sensitivity analysis eliminating calls to the RRS as an outcome (so using the alternative outcome of unexpected transfers to the ICU and resuscitation calls). The results are similar to those of our main analysis, with all analyzed scores having lower sensitivity and PPV in surgical hospitalizations when compared to medical hospitalizations.

Our study suggests that, to optimize detection of physiological deterioration events, EWS should try to take into account different patient types, with the most basic distinction being surgical vs medical. This tailoring will make EWS more complex, and less suited for paper-based calculation, but new electronic health records are increasingly able to incorporate decision support, and some EWS have been developed for electronic calculation only. Of particular interest in this regard is the score developed by Escobar et al,31 which groups patients into categories according to the reason for admission, and calculates a different subscore based on that category. While the score by Escobar et al. does not split patients based on medical or surgical status, a more general interpretation of our results suggests that a score may be more accurate if it classifies patients into subgroups with different subscores. This seems to be confirmed by the fact that the score by Escobar et al performs better than MEWS.28 Unfortunately, the paper describing it does not provide enough detail to use it in our database.

A recent systematic review showed increasing evidence that RRS may be effective in reducing CRAs occurring in a non-ICU setting and, more important, overall inhospital mortality.32 While differing implementation strategies (eg, different length of the educational effort, changes in the frequency of vital signs monitoring) can impact the success of such an initiative, it has been speculated that the afferent limb (which often includes an EWS) might be the most critical part of the system.33 Our results show that the most widely used EWS perform significantly worse on surgical patients, and suggest that a way to improve the accuracy of EWS would be to tailor the risk calculation to different patient subgroups (eg, medical and surgical patients). Plausible next steps would be to demonstrate that tailoring risk calculation to medical and surgical patients separately can improve risk predictions and accuracy of EWS.

 

 

Disclosure

The authors report no financial conflicts of interest.

Patients typically show signs and symptoms of deterioration hours to days prior to cardiorespiratory arrest.1,2 The rate of inhospital cardiorespiratory arrest (CRA) requiring cardiopulmonary resuscitation is estimated to be 0.174 per bed per year in the United States.3 After CRA, survival to discharge is estimated to be as low as 18%.3,4 Efforts to predict and prevent arrest could prove beneficial.1,2

Rapid response systems (RRS) have been proposed as a means of identifying clinical deterioration and facilitating a timely response. These systems were designed to bring clinicians with critical care expertise to the bedside to prevent unnecessary deaths. They typically include an afferent limb (detects deteriorating patients), an efferent limb (responds to calls and acts to avoid further deterioration), and administrative and data analysis limbs.5,6 Automatic provision of recommendations and computer-based systems are desirable components of the afferent limb of the detection system.6 Both are independent predictors of improved clinical practices for clinical decision support systems.7 However, the existing early warning scores (EWS) may not be ready for automation due to low positive predictive values (PPV) and sensitivities.8

It is possible that the low discriminatory accuracy of the published EWS may be secondary to the use of aggregate patient populations for derivation of scores. We hypothesized that these EWS perform differently in medical and in surgical subpopulations. Also, the EWS need to be tested in a time-dependent manner to serve as a realistic clinical support tool for hospitalized patients.

STUDY AIM

The aim of this study was to evaluate the differential performance of widely used EWS in medical vs surgical patients.

METHODS

Site

The study was conducted in an academic center with 2 hospitals in Southeastern Minnesota totaling approximately 1500 general care nonintensive care unit (ICU) beds. The Mayo Clinic Institutional Review Board approved the research proposal.

Subjects

Our retrospective cohort was comprised of all adult inpatients discharged from 2 academic hospitals between January 1, 2011 and December 31, 2011 who spent any time in a general care (non-ICU) unit. We excluded patients younger than 18 years, psychiatric or rehabilitation inpatients, those without research authorization, and patients admitted for research purposes.

Study patients were divided into medical and surgical cohorts. Hospitalizations were considered surgical if patients had surgery at any time during their hospital stay according to billing data. A trigger was an instance in which a patient met the conditions of a specific rule (score/vital sign exceeded the published/defined threshold).

A resuscitation call was defined as a call for cardiopulmonary resuscitation when a patient has a CRA.

An event was an occurrence of 1 of the following in a general care setting: unplanned transfer to the ICU, resuscitation call, or RRS activation.

The RRS activation criteria consisted of an “acute and persistent change” in any 1 or more of the following: oxygen saturations less than 90%, heart rate less than 40 or greater than 130 beats/minute, systolic blood pressure less than 90 mm Hg, or respiratory rate less than 10 or greater than 28 breaths/minute. The RRS activation requires health provider action; they are not electronically generated. Nurses and physicians may also activate the RRS if they are concerned about a patient, even if calling criteria are not met. This is in contrast to the EWS analyzed, which are aggregate composites of multiple parameters. However, whether or not a derangement in vital signs is considered an “acute and persistent change” still involves clinical judgment. Any movement from a general care bed to an ICU bed, or from a general care bed to a procedure area, and from there to an ICU, was considered unplanned. Transfers to the ICU directly from the emergency department or operating room (OR) were not considered as an unplanned transfer and were not included in the analyses.

Coverage time was the period observed for events after a rule was triggered. In this analysis, a coverage time of 24 hours was considered, with a 1-hour look-back. A trigger was counted as a true positive if an event occurred during the following 24 hours. The 1-hour look-back was included to take into account the nursing clinical process of prioritizing a call to the RRS followed by documentation of the altered vital signs that prompted the call.

An episode was the continuous time on the general care floor within a hospitalization, excluding times when a patient was in the OR or ICU. For example, if a patient was admitted to a general bed on a surgery floor, subsequently went to the OR, and then returned to the surgery floor, the 2 episodes were considered separate: the time on the floor before surgery, and the time on the floor after surgery.

Assessment of implementation of RRS in our hospitals showed a significant drop in the failure-to-rescue rate (issues considered related to delay or failure to identify or intervene appropriately when a patient was deteriorating, as identified through mortality review) and a decrease in non-ICU mortality.9,10 This suggests that our current process captures many of the relevant episodes of acute deterioration when a rapid response team is needed and supports using RRS activation as outcomes.

 

 

Data Sources

We developed a time-stamped longitudinal database of patient data from the electronic health record, including vital signs, laboratory test results, demographics (age, sex), administrative data (including length of stay), comorbidities, resuscitation code status, location in hospital, and at the minute level throughout each patient’s hospital stay. Physiologically impossible values (eg, blood pressures of 1200 mm Hg) were considered entered in error and eliminated from the database. Time spent in the OR or ICU was excluded because RRS activation would not be applied in these already highly monitored areas. SAS Statistical software (SAS Institute Inc. Cary, North Carolina) was used for database creation.

We applied the current RRS calling criteria in our institution and calculated the Kirkland score,11 along with some of the most widely used early warning scores:12 Modified Early Warning System (MEWS),13 Standardized Early Warning Scoring System (SEWS),14 Global Modified Early Warning Score (GMEWS),15 Worthing physiologic scoring system,16 National Early Warning Score (NEWS),17 and VitaPAC Early Warning Score (ViEWS).18 Published thresholds for these scores were used to create rule triggers in the data. Once a trigger was created to calculate the number of false positives and true positives, all subsequent triggers were ignored until the end of the episode or until 24 hours elapsed. We calculated triggers in a rolling fashion throughout the episodes of care. The EWS score was updated every time a new parameter was entered into the analytical electronic health record, and the most recent value for each was used to calculate the score. SAS statistical software was used for calculation of scores and identification of outcomes.

For our analysis, events were treated as dependent variables, and triggers were independent variables. We calculated the score for each EWS to the minute level throughout our retrospective database. If the score for a specific EWS was higher than the published/recommended threshold for that EWS, an alert was considered to have been issued, and the patient was followed for 24 hours. If the patient had an event in the subsequent 24 hours, or 1 hour before (1-hour look-back), the alert was considered a true positive; if not, a false positive. Events that were not preceded by an alert were false negatives, and 24-hour intervals without either an alert or an event were considered true negatives. This simulation exercise was performed for each EWS in both subcohorts (medical and surgical). Clusters of RRS calls followed by transfers to the ICU within 3 hours were considered as a single adverse event (RRS calls, as it was the first event to occur) to avoid double counting. We have described how well this simulation methodology,8 correlates with results from prospective studies.19

Statistical Analysis

To calculate whether results were statistically significant for subgroups, a jackknife method of calculating variance20 was used. The jackknife method calculates variance by repeating the calculations of the statistic leaving out 1 sample at a time. In our case, we repeated the calculation of sensitivity and PPV leaving out 1 patient at a time. Once the simulation method had been run and the false/true positives/negatives had been assigned, calculation of each metric (PPV and sensitivity) was repeated for n subsamples, each leaving out 1 patient. The variance was calculated and 2 Student t tests were performed for each EWS: 1 for PPV and another for sensitivity. SAS statistical software v 9.3 was used for the simulation analysis; R statistical software v 3.0.2 (The R Foundation, Vienna, Austria) was used for the calculation of the statistical significance of results. A univariable analysis was also performed to assess the sensitivity and PPVs for the published thresholds of the most common variables in each EWS: respiratory rate, systolic blood pressure, heart rate, temperature, and mental status as measured by the modified Richmond Agitation Sedation Score.21

RESULTS

The initial cohort included 60,020 hospitalizations, of which the following were excluded: 2751 because of a lack of appropriate research authorization; 6433 because the patients were younger than 18 years; 2129 as psychiatric admissions; 284 as rehabilitation admissions; 872 as research purposes-only admissions; and 1185 because the patient was never in a general care bed (eg, they were either admitted directly to the ICU, or they were admitted for an outpatient surgical procedure and spent time in the postanesthesia care unit).

Table 1 summarizes patient and trigger characteristics, overall and by subgroup. The final cohort included 75,240 total episodes in 46,366 hospitalizations, from 34,898 unique patients, of which 48.7% were male. There were 23,831 medical and 22,535 surgical hospitalizations. Median length of episode was 2 days both for medical and surgical patients. Median length of stay was 3 days, both for medical and for surgical patients.

Patient Characteristics, Events, and Triggers
Table 1


There were 3332 events in total, of which 1709 were RRS calls, 185 were resuscitation calls, and 1438 were unscheduled transfers to the ICU. The rate of events was 4.67 events per 100 episodes in the aggregate adult population. There were 3.93 events per 100 episodes for surgical hospitalizations, and 5.86 events per 100 episodes for medical hospitalizations (P < .001). The number of CRAs in our cohort was 0.27 per 100 episodes, 0.128 per hospital bed per year, or 4.37 per 1000 hospital admissions, similar to other reported numbers in the literature.3, 22,23

The total number of EWS triggers varied greatly between EWS rules, with the volume ranging during the study year from 1363 triggers with the GMEWS rule to 77,711 triggers with the ViEWS score.

Performance of scores in medical and surgical patients
Figure


All scores had PPVs less than 25%. As seen in Table 2 and shown graphically in the Figure, all scores performed better on medical patients (blue) than on surgical patients (yellow). The P value was < .0001 for both PPV and sensitivity. The Worthing score had the highest sensitivity (0.78 for medical and 0.68 for surgical) but a very low PPV (0.04 for medical and 0.03 for surgical), while GMEWS was the opposite: low sensitivity (0.10 and 0.07) but the highest PPV (0.22 and 0.18).

Comparison of the Predictive Performance of Widely Used EWS in a Surgical and a Medical Population
Table 2


The results of the univariable analysis can be seen in Table 3. Most of the criteria performed better (higher sensitivity and PPV) as predictors in the medical hospitalizations than in the surgical hospitalizations.

Univariable Analysis in the Medical and Surgical Subpopulations
Table 3

 

 

DISCUSSION

We hypothesized that EWS may perform differently when applied to medical rather than surgical patients. Studies had not analyzed this in a time-dependent manner,24-26 which limited the applicability of the results.8

All analyzed scores performed better in medical patients than in surgical patients (Figure). This could reflect a behavioral difference by the teams on surgical and medical floors in the decision to activate the RRS, or a bias of the clinicians who designed the scores (mostly nonsurgeons). The difference could also mean that physiological deteriorations are intrinsically different in patients who have undergone anesthesia and surgery. For example, in surgical patients, a bleeding episode is more likely to be the cause of their physiological deterioration, or the lingering effects of anesthesia could mask underlying deterioration. Such patients would benefit from scores where variables such as heart rate, blood pressure, or hemoglobin had more influence.

When comparing the different scores, it was much easier for a patient to meet the alerting score with the Worthing score than with GMEWS. In the Worthing score, a respiratory rate greater than 22 breaths per minute, or a systolic blood pressure less than 100 mm Hg, already meet alerting criteria. Similar vital signs result in 0 and 1 points (respectively) in GMEWS, far from its alerting score of 5. This reflects the intrinsic tradeoff of EWS: as the threshold for considering a patient “at risk” drops, not only does the number of true positives (and the sensitivity) increase, but also the number of false positives, thus lowering the PPV.

However, none of the scores analyzed were considered to perform well based on their PPV and sensitivity, particularly in the surgical subpopulation. Focusing on another metric, the area under the receiver operator curve can give misleadingly optimistic results.24,27 However, the extremely low prevalence of acute physiological deterioration can produce low PPVs even when specificity seems acceptable, which is why it is important to evaluate PPV directly.28

To use EWS effectively to activate RRS, they need to be combined with clinical judgment to avoid high levels of false alerts, particularly in surgical patients. It has been reported that RRS is activated only 30% of the time a patient meets RRS calling criteria.29 While there may be cultural characteristics inhibiting the decision to call,30 our study hints at another explanation: if RRS was activated every time a patient met calling criteria based on the scores analyzed, the number of RRS calls would be very high and difficult to manage. So health providers may be doing the right thing when “filtering” RRS calls and not applying the criteria strictly, but in conjunction with clinical judgment.

A limitation of any study like this is how to define “acute physiological deterioration.” We defined an event as recognized episodes of acute physiological deterioration that are signaled by escalations of care (eg, RRS, resuscitation calls, or transfers to an ICU) or unexpected death. By definition, our calculated PPV is affected by clinicians’ recognition of clinical deteriorations. This definition, common in the literature, has the limitation of potentially underestimating EWS’ performance by missing some events that are resolved by the primary care team without an escalation of care. However, we believe our interpretation is not unreasonable since the purpose of EWS is to trigger escalations of care in a timely fashion. Prospective studies could define an event in a way that is less affected by the clinicians’ judgment.

Regarding patient demographics, age was similar between the 2 groups (average, 58.2 years for medical vs 58.9 years for surgical), and there was only a small difference in gender ratios (45.1% male in the medical vs 51.4% in the surgical group). These differences are unlikely to have affected the results significantly, but unknown differences in demographics or other patient characteristics between groups may account for differences in score performance between surgical and medical patients.

Several of the EWS analyzed had overlapping trigger criteria with our own RRS activation criteria (although as single-parameter triggers and not as aggregate). To test how these potential biases could affect our results, we performed a post hoc sensitivity analysis eliminating calls to the RRS as an outcome (so using the alternative outcome of unexpected transfers to the ICU and resuscitation calls). The results are similar to those of our main analysis, with all analyzed scores having lower sensitivity and PPV in surgical hospitalizations when compared to medical hospitalizations.

Our study suggests that, to optimize detection of physiological deterioration events, EWS should try to take into account different patient types, with the most basic distinction being surgical vs medical. This tailoring will make EWS more complex, and less suited for paper-based calculation, but new electronic health records are increasingly able to incorporate decision support, and some EWS have been developed for electronic calculation only. Of particular interest in this regard is the score developed by Escobar et al,31 which groups patients into categories according to the reason for admission, and calculates a different subscore based on that category. While the score by Escobar et al. does not split patients based on medical or surgical status, a more general interpretation of our results suggests that a score may be more accurate if it classifies patients into subgroups with different subscores. This seems to be confirmed by the fact that the score by Escobar et al performs better than MEWS.28 Unfortunately, the paper describing it does not provide enough detail to use it in our database.

A recent systematic review showed increasing evidence that RRS may be effective in reducing CRAs occurring in a non-ICU setting and, more important, overall inhospital mortality.32 While differing implementation strategies (eg, different length of the educational effort, changes in the frequency of vital signs monitoring) can impact the success of such an initiative, it has been speculated that the afferent limb (which often includes an EWS) might be the most critical part of the system.33 Our results show that the most widely used EWS perform significantly worse on surgical patients, and suggest that a way to improve the accuracy of EWS would be to tailor the risk calculation to different patient subgroups (eg, medical and surgical patients). Plausible next steps would be to demonstrate that tailoring risk calculation to medical and surgical patients separately can improve risk predictions and accuracy of EWS.

 

 

Disclosure

The authors report no financial conflicts of interest.

References

1. Buist MD, Jarmolowski E, Burton PR, Bernard SA, Waxman BP, Anderson J. Recognising clinical instability in hospital patients before cardiac arrest or unplanned admission to intensive care. A pilot study in a tertiary-care hospital. Med J Aust. 1999; 171(1):22-25. PubMed
2. Schein RM, Hazday N, Pena M, Ruben BH, Sprung CL. Clinical antecedents to in-hospital cardiopulmonary arrest. Chest. 1990;98(6):1388-1392. PubMed
3. Peberdy MA, Kaye W, Ornato JP, Larkin GL, Nadkarni V, Mancini ME, et al. Cardiopulmonary resuscitation of adults in the hospital: a report of 14720 cardiac arrests from the National Registry of Cardiopulmonary Resuscitation. Resuscitation. 2003; 58(3):297-308. PubMed
4. Nadkarni VM, Larkin GL, Peberdy MA, Carey SM, Kaye W, Mancini ME, et al. First documented rhythm and clinical outcome from in-hospital cardiac arrest among children and adults. JAMA. 2006;295(1):50-57. PubMed
5. Devita MA, Bellomo R, Hillman K, Kellum J, Rotondi A, Teres D, et al. Findings of the first consensus conference on medical emergency teams. Crit Care Med. 2006;34(9):2463-2478. PubMed
6. DeVita MA, Smith GB, Adam SK, Adams-Pizarro I, Buist M, Bellomo R, et al. “Identifying the hospitalised patient in crisis”--a consensus conference on the afferent limb of rapid response systems. Resuscitation. 2010;81(4):375-382. PubMed
7. Kawamoto K, Houlihan CA, Balas EA, Lobach DF. Improving clinical practice using clinical decision support systems: a systematic review of trials to identify features critical to success. BMJ. 2005;330(7494):765. PubMed
8. Romero-Brufau S, Huddleston JM, Naessens JM, Johnson MG, Hickman J, Morlan BW, et al. Widely used track and trigger scores: are they ready for automation in practice? Resuscitation. 2014;85(4):549-552. PubMed
9. Huddleston JM, Diedrich DA, Kinsey GC, Enzler MJ, Manning DM. Learning from every death. J Patient Saf. 2014;10(1):6-12. PubMed
10. Moriarty JP, Schiebel NE, Johnson MG, Jensen JB, Caples SM, Morlan BW, et al. Evaluating implementation of a rapid response team: considering alternative outcome measures. Int J Qual Health Care. 2014;26(1):49-57. PubMed
11. Kirkland LL, Malinchoc M, O’Byrne M, Benson JT, Kashiwagi DT, Burton MC, et al. A clinical deterioration prediction tool for internal medicine patients. Am J Med Qual. 2013;28(2):135-142. PubMed
12. Griffiths JR, Kidney EM. Current use of early warning scores in UK emergency departments. Emerg Med J. 2012;29(1):65-66. PubMed
13. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521-526. PubMed
14. Paterson R, MacLeod DC, Thetford D, Beattie A, Graham C, Lam S, et al.. Prediction of in-hospital mortality and length of stay using an early warning scoring system: clinical audit. Clin Med (Lond). 2006;6(3):281-284. PubMed
15. Harrison GA, Jacques T, McLaws ML, Kilborn G. Combinations of early signs of critical illness predict in-hospital death–the SOCCER study (signs of critical conditions and emergency responses). Resuscitation. 2006;71(3):327-334. PubMed
16. Duckitt RW, Buxton-Thomas R, Walker J, Cheek E, Bewick V, Venn R, et al. Worthing physiological scoring system: derivation and validation of a physiological early-warning system for medical admissions. An observational, population-based single-centre study. Br J Anaesth. 2007; 98(6):769-774. PubMed
17. Smith GB, Prytherch DR, Meredith P, Schmidt PE, Featherstone PI. The ability of the National Early Warning Score (NEWS) to discriminate patients at risk of early cardiac arrest, unanticipated intensive care unit admission, and death. Resuscitation. 2013;84(4):465-470. PubMed
18. Prytherch DR, Smith GB, Schmidt PE, Featherstone PI. ViEWS--Towards a national early warning score for detecting adult inpatient deterioration. Resuscitation. 2010;81(8):932-937. PubMed
19. Romero-Brufau S, Huddleston JM. Reply to letter: widely used track and trigger scores: are they ready for automation in practice? Resuscitation. 2014;85(10):e159. PubMed
20. Efron B, Stein C. The jackknife estimate of variance. Annals of Statistics. 1981;586-596. 
21. Sessler CN, Gosnell MS, Grap MJ, Brophy GM, O’Neal PV, Keane KA, et al. The Richmond Agitation-Sedation Scale: validity and reliability in adult intensive care unit patients. Am J Respir Crit Care Med. 2002;166(10):1338-1344. PubMed
22. DeVita MA, Braithwaite RS, Mahidhara R, Stuart S, Foraida M, Simmons RL. Medical Emergency Response Improvement Team (MERIT). Use of medical emergency team responses to reduce hospital cardiopulmonary arrests. Qual Saf Health Care. 2004;13(4):251-254. PubMed
23. Goncales PD, Polessi JA, Bass LM, Santos Gde P, Yokota PK, Laselva CR, et al. Reduced frequency of cardiopulmonary arrests by rapid response teams. Einstein (Sao Paulo). 2012;10(4):442-448. PubMed
24. Cuthbertson BH, Boroujerdi M, McKie L, Aucott L, Prescott G. Can physiological variables and early warning scoring systems allow early recognition of the deteriorating surgical patient? Crit Care Med. 2007;35(2):402-409. PubMed
25. Gardner-Thorpe J, Love N, Wrightson J, Walsh S, Keeling N. The value of Modified Early Warning Score (MEWS) in surgical in-patients: a prospective observational study. Ann R Coll Surg Engl. 2006;88(6):571-575. PubMed
26. Stenhouse C, Coates S, Tivey M, Allsop P, Parker T. Prospective evaluation of a modified Early Warning Score to aid earlier detection of patients developing critical illness on a general surgical ward. British Journal of Anaesthesia. 2000;84(5):663-663. 
27. Smith GB, Prytherch DR, Schmidt PE, Featherstone PI. Review and performance evaluation of aggregate weighted ‘track and trigger’ systems. Resuscitation. 2008;77(2):170-179. PubMed
28. Romero-Brufau S, Huddleston JM, Escobar GJ, Liebow M. Why the C-statistic is not informative to evaluate early warning scores and what metrics to use. Crit Care. 2015; 19:285. PubMed
29. Hillman K, Chen J, Cretikos M, Bellomo R, Brown D, Doig G, et al. Introduction of the medical emergency team (MET) system: a cluster-randomised controlled trial. Lancet. 2005;365(9477):2091-2097. PubMed
30. Shearer B, Marshall S, Buist MD, Finnigan M, Kitto S, Hore T, et al. What stops hospital clinical staff from following protocols? An analysis of the incidence and factors behind the failure of bedside clinical staff to activate the rapid response system in a multi-campus Australian metropolitan healthcare service. BMJ Qual Saf. 2012;21(7):569-575. PubMed
31. Escobar GJ, LaGuardia JC, Turk BJ, Ragins A, Kipnis P, Draper D. Early detection of impending physiologic deterioration among patients who are not in intensive care: development of predictive models using data from an automated electronic medical record. J Hosp Med. 2012;7(5):388-395. PubMed
32. Winters BD, Weaver SJ, Pfoh ER, Yang T, Pham JC, Dy SM. Rapid-response systems as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):417-425. PubMed
33. Jones DA, DeVita MA, Bellomo R. Rapid-response teams. N Engl J Med. 2011;365(2):139-146. PubMed

References

1. Buist MD, Jarmolowski E, Burton PR, Bernard SA, Waxman BP, Anderson J. Recognising clinical instability in hospital patients before cardiac arrest or unplanned admission to intensive care. A pilot study in a tertiary-care hospital. Med J Aust. 1999; 171(1):22-25. PubMed
2. Schein RM, Hazday N, Pena M, Ruben BH, Sprung CL. Clinical antecedents to in-hospital cardiopulmonary arrest. Chest. 1990;98(6):1388-1392. PubMed
3. Peberdy MA, Kaye W, Ornato JP, Larkin GL, Nadkarni V, Mancini ME, et al. Cardiopulmonary resuscitation of adults in the hospital: a report of 14720 cardiac arrests from the National Registry of Cardiopulmonary Resuscitation. Resuscitation. 2003; 58(3):297-308. PubMed
4. Nadkarni VM, Larkin GL, Peberdy MA, Carey SM, Kaye W, Mancini ME, et al. First documented rhythm and clinical outcome from in-hospital cardiac arrest among children and adults. JAMA. 2006;295(1):50-57. PubMed
5. Devita MA, Bellomo R, Hillman K, Kellum J, Rotondi A, Teres D, et al. Findings of the first consensus conference on medical emergency teams. Crit Care Med. 2006;34(9):2463-2478. PubMed
6. DeVita MA, Smith GB, Adam SK, Adams-Pizarro I, Buist M, Bellomo R, et al. “Identifying the hospitalised patient in crisis”--a consensus conference on the afferent limb of rapid response systems. Resuscitation. 2010;81(4):375-382. PubMed
7. Kawamoto K, Houlihan CA, Balas EA, Lobach DF. Improving clinical practice using clinical decision support systems: a systematic review of trials to identify features critical to success. BMJ. 2005;330(7494):765. PubMed
8. Romero-Brufau S, Huddleston JM, Naessens JM, Johnson MG, Hickman J, Morlan BW, et al. Widely used track and trigger scores: are they ready for automation in practice? Resuscitation. 2014;85(4):549-552. PubMed
9. Huddleston JM, Diedrich DA, Kinsey GC, Enzler MJ, Manning DM. Learning from every death. J Patient Saf. 2014;10(1):6-12. PubMed
10. Moriarty JP, Schiebel NE, Johnson MG, Jensen JB, Caples SM, Morlan BW, et al. Evaluating implementation of a rapid response team: considering alternative outcome measures. Int J Qual Health Care. 2014;26(1):49-57. PubMed
11. Kirkland LL, Malinchoc M, O’Byrne M, Benson JT, Kashiwagi DT, Burton MC, et al. A clinical deterioration prediction tool for internal medicine patients. Am J Med Qual. 2013;28(2):135-142. PubMed
12. Griffiths JR, Kidney EM. Current use of early warning scores in UK emergency departments. Emerg Med J. 2012;29(1):65-66. PubMed
13. Subbe CP, Kruger M, Rutherford P, Gemmel L. Validation of a modified Early Warning Score in medical admissions. QJM. 2001;94(10):521-526. PubMed
14. Paterson R, MacLeod DC, Thetford D, Beattie A, Graham C, Lam S, et al.. Prediction of in-hospital mortality and length of stay using an early warning scoring system: clinical audit. Clin Med (Lond). 2006;6(3):281-284. PubMed
15. Harrison GA, Jacques T, McLaws ML, Kilborn G. Combinations of early signs of critical illness predict in-hospital death–the SOCCER study (signs of critical conditions and emergency responses). Resuscitation. 2006;71(3):327-334. PubMed
16. Duckitt RW, Buxton-Thomas R, Walker J, Cheek E, Bewick V, Venn R, et al. Worthing physiological scoring system: derivation and validation of a physiological early-warning system for medical admissions. An observational, population-based single-centre study. Br J Anaesth. 2007; 98(6):769-774. PubMed
17. Smith GB, Prytherch DR, Meredith P, Schmidt PE, Featherstone PI. The ability of the National Early Warning Score (NEWS) to discriminate patients at risk of early cardiac arrest, unanticipated intensive care unit admission, and death. Resuscitation. 2013;84(4):465-470. PubMed
18. Prytherch DR, Smith GB, Schmidt PE, Featherstone PI. ViEWS--Towards a national early warning score for detecting adult inpatient deterioration. Resuscitation. 2010;81(8):932-937. PubMed
19. Romero-Brufau S, Huddleston JM. Reply to letter: widely used track and trigger scores: are they ready for automation in practice? Resuscitation. 2014;85(10):e159. PubMed
20. Efron B, Stein C. The jackknife estimate of variance. Annals of Statistics. 1981;586-596. 
21. Sessler CN, Gosnell MS, Grap MJ, Brophy GM, O’Neal PV, Keane KA, et al. The Richmond Agitation-Sedation Scale: validity and reliability in adult intensive care unit patients. Am J Respir Crit Care Med. 2002;166(10):1338-1344. PubMed
22. DeVita MA, Braithwaite RS, Mahidhara R, Stuart S, Foraida M, Simmons RL. Medical Emergency Response Improvement Team (MERIT). Use of medical emergency team responses to reduce hospital cardiopulmonary arrests. Qual Saf Health Care. 2004;13(4):251-254. PubMed
23. Goncales PD, Polessi JA, Bass LM, Santos Gde P, Yokota PK, Laselva CR, et al. Reduced frequency of cardiopulmonary arrests by rapid response teams. Einstein (Sao Paulo). 2012;10(4):442-448. PubMed
24. Cuthbertson BH, Boroujerdi M, McKie L, Aucott L, Prescott G. Can physiological variables and early warning scoring systems allow early recognition of the deteriorating surgical patient? Crit Care Med. 2007;35(2):402-409. PubMed
25. Gardner-Thorpe J, Love N, Wrightson J, Walsh S, Keeling N. The value of Modified Early Warning Score (MEWS) in surgical in-patients: a prospective observational study. Ann R Coll Surg Engl. 2006;88(6):571-575. PubMed
26. Stenhouse C, Coates S, Tivey M, Allsop P, Parker T. Prospective evaluation of a modified Early Warning Score to aid earlier detection of patients developing critical illness on a general surgical ward. British Journal of Anaesthesia. 2000;84(5):663-663. 
27. Smith GB, Prytherch DR, Schmidt PE, Featherstone PI. Review and performance evaluation of aggregate weighted ‘track and trigger’ systems. Resuscitation. 2008;77(2):170-179. PubMed
28. Romero-Brufau S, Huddleston JM, Escobar GJ, Liebow M. Why the C-statistic is not informative to evaluate early warning scores and what metrics to use. Crit Care. 2015; 19:285. PubMed
29. Hillman K, Chen J, Cretikos M, Bellomo R, Brown D, Doig G, et al. Introduction of the medical emergency team (MET) system: a cluster-randomised controlled trial. Lancet. 2005;365(9477):2091-2097. PubMed
30. Shearer B, Marshall S, Buist MD, Finnigan M, Kitto S, Hore T, et al. What stops hospital clinical staff from following protocols? An analysis of the incidence and factors behind the failure of bedside clinical staff to activate the rapid response system in a multi-campus Australian metropolitan healthcare service. BMJ Qual Saf. 2012;21(7):569-575. PubMed
31. Escobar GJ, LaGuardia JC, Turk BJ, Ragins A, Kipnis P, Draper D. Early detection of impending physiologic deterioration among patients who are not in intensive care: development of predictive models using data from an automated electronic medical record. J Hosp Med. 2012;7(5):388-395. PubMed
32. Winters BD, Weaver SJ, Pfoh ER, Yang T, Pham JC, Dy SM. Rapid-response systems as a patient safety strategy: a systematic review. Ann Intern Med. 2013;158(5 pt 2):417-425. PubMed
33. Jones DA, DeVita MA, Bellomo R. Rapid-response teams. N Engl J Med. 2011;365(2):139-146. PubMed

Issue
Journal of Hospital Medicine 12(4)
Issue
Journal of Hospital Medicine 12(4)
Page Number
217-223
Page Number
217-223
Topics
Article Type
Display Headline
Evaluating automated rules for rapid response system alarm triggers in medical and surgical patients
Display Headline
Evaluating automated rules for rapid response system alarm triggers in medical and surgical patients
Sections
Article Source

© 2017 Society of Hospital Medicine

Disallow All Ads
Correspondence Location
Address for correspondence and reprint requests: Lisa L. Kirkland MD, 200 First Street SW, Rochester, MN 55905; Telephone: 507-255-8715; Fax: 507-255-9189; E-mail: kirkland.lisa@mayo.edu
Content Gating
Gated (full article locked unless allowed per User)
Alternative CME
Gating Strategy
First Peek Free
Article PDF Media

Effect of Plate in Close Proximity to Empty External-Fixation Pin Site on Long-Bone Torsional Strength

Article Type
Changed
Thu, 09/19/2019 - 13:22
Display Headline
Effect of Plate in Close Proximity to Empty External-Fixation Pin Site on Long-Bone Torsional Strength

Take-Home Points

  • The location of a bicortical defect in proximity to a tibia plate does not appear to affect the torsional stiffness or torsional failure strength of the bone.
  • External fixator pin placement should be based on considerations other than the potential for creating a distal stress riser after definitive fracture management.

A stress riser in cortical bone may be considered any abrupt change in the contour or consistency of the hollow structure, such as a surface defect, that not only weakens the bone but concentrates stresses at that transition point.1 A cortical defect that is 20% of the bone diameter is associated with a 34% decrease in torsional strength, thus representing a “stress riser.”2 High-energy and complex tibia fractures are often provisionally stabilized with external fixation that gives the soft tissues time to recover before definitive fracture fixation. Pin diameter for a medium-size tibia external fixator typically is 5.0 mm, resulting in a 10-mm defect in bicortical placement. Therefore, any tibia with a diameter of <50 mm is at risk for a stress riser fracture.

Although it had been established that sizable cortical defects can decrease the torsional strength of long bone,2 the effect of a plate in close proximity to a defect secondary to an empty external-fixator pin site on torsional strength has not been determined. We conducted a study to evaluate this effect. The null hypothesis was there would be no difference in tibia torsional strength attributable to varying the proximity of a tibia midshaft plate to a 5.0-mm bicortical defect.

Methods

Forty fourth-generation, medium-size left composite tibias (Pacific Research Laboratories) were divided into 8 groups of 5 bones (Figure 1).

Figure 1.
To represent the stress riser created by the removal of a 5.0-mm Schantz external fixation pin, we produced distal tibia bicortical defects in 6 of the groups by creating anterior-to-posterior 5.0-mm bicortical drill holes. The longitudinal location of these drill holes was varied in relation to the distal end of a 4.5-mm × 121-mm 6-hole locking plate (PeriLoc; Smith & Nephew) fixed in a nonlocking configuration and positioned across the tibia midline on the anterior-medial aspect. In the experimental plated groups, the bicortical defects were created 3.0, 2.0, and 1.0 cm distal to the plate end, with 1 plated group without a defect. The control groups consisted of equivalent defects in the same distal longitudinal locations, without plates attached, as well as an unplated group without a defect.

Torsion testing to failure was performed for all specimens in a manner similar to that described by Gardner and colleagues.3 Impression molds for the composite tibia constructed from polymethylmethacrylate encased the superior and distal ends, leaving 25.5 cm of exposed midshaft. This allowed the composites to be rigidly clamped into a materials testing system (858 Mini-Bionix; MTS) equipped with a 100.0-Nm torsional load cell (Figure 2).

Figure 2.
The constructs were preconditioned by rotating the superior end internally up to 15.0 Nm at a rate of 0.25 Nm/s for 2 complete cycles. Next, the constructs were preloaded axially to 20.0 N and then rotated at 0.25°/s until failure. Torsional load and torsional displacement were recorded and used to determine construct stiffness and failure load. Stiffness was calculated as the slope of the linear elastic portion of the load versus displacement curves between 20.0 Nm and 40.0 Nm. Failure load was defined as the highest load achieved before fracture. One-way analysis of variance with Tukey adjustment for multiple comparisons and α set at 0.05 were used to detect differences in failure stiffness and failure load between the 8 constructs.

Results

Graphical results for torsional stiffness are presented in Figure 3. R2 for all stiffness calculations was >0.99.

Figure 3.
There were no statistical differences in torsional stiffness between any of the groups. Graphical results for torsional failure load are presented in Figure 4.
Figure 4.
During torsion-to-failure testing, both plated and unplated specimens without distal cortical defect holes nearly exceeded the torque capacity of the load cell without failing, stopping the tests. For the specimens that failed, there were no statistical differences in failure torque. A slight trend toward higher failure loads in plated specimens with a hole in close proximity was seen in the 1.0-cm distal defect hole groups, with the plated specimen achieving a higher mean (SD) failure load, 78.14 (7.58) Nm, than the unplated group, 66.75 (1.84) Nm, but this was not significant (P = .06). Another slight trend toward lower failure load in unplated specimens as the defect moved proximally was seen between the unplated 3.0-cm defect group, 77.91 (6.08) Nm, and the unplated 1.0-cm defect group, 66.75 (1.84) Nm; this was also not significant (P = .07). Mode of failure for all specimens with bicortical defects, with or without a plate, was a spiral fracture that bisected the axis of the defect (Figure 5).
Figure 5.
Post hoc power analysis for each measure indicated statistical power of 80% for stiffness and 75% for failure torque.

 

 

Discussion

Many tibia fractures require provisional stabilization with an external fixator that spans the knee, because of the high-energy nature of the injury or other, higher-priority polytrauma concerns. When the patient or injury is suitable for definitive fixation, the external fixator typically is removed in favor of internal fixation with a plate and screws. Depending on the nature and location of the fracture and the subsequent plate, the empty cortical pin-site defects, often lying at varying distances from the distal end of the plate, can potentially serve as stress risers for fracture.4

Other studies have evaluated long-bone cortical defects biomechanically1,2,4 and clinically,5-7 and multiple studies have been conducted on the effects of plates on long-bone strength for fracture stabilization.8-13 The present study evaluated the torsional strength of long bones in the presence of a bicortical defect and the proximity of the defect to a plate. There were no differences in stiffness or failure load between any of the groups of plated and unplated fourth-generation composite tibias tested to failure in torsion with varying distal bicortical defects. Hypothetically, one would expect the torsional stiffness of these specimens to increase with the mere addition of a metallic diaphyseal plate. However, this study demonstrated that the addition of a plate did not affect the torsional stiffness or strength of the tibias. Clinically, it is common practice to place external fixator pins as far as possible outside the planned incision site for definitive fracture fixation. Thus, we also hypothesized that the presence of a bicortical pin-site defect and its proximity to the plate would alter the torsional strength of the tibia specimens, and that the distal pin-site defect’s location farthest from the plate would exhibit greater strength, but this did not occur. Although other studies have shown that the presence of bicortical defects decreases the strength of long bones, we were unable to quantify this decrease because the 2 intact groups of composites, plated and unplated, survived failure testing.

This study had several limitations, first being the use of composite tibias as opposed to human cadaver bone. Although fourth-generation composite bone models have been validated as a suitable and accurate biomechanical substitute for cadaver specimens,14 anatomical variations in cadaver tibias may transfer forces differently through plates, screws, and distal pin sites. In order to test plated specimens against the unplated controls, we did not simulate a mid-shaft fracture in any of the tibias. The pin-site defects were intended to reflect the mechanical effects of bicortical defects immediately after pin removal and in the absence of any degree of bone healing. Finally, this study focused on pin-site defects that were distal to a midshaft plate and that may not represent the effects of bicortical pin-site defects proximal to the plate.

Given the results of this biomechanical study in composite tibias, varying the proximity of a bicortical defect to a plate does not affect the torsional stiffness or torsional failure strength of the bone. Placement of an intended bicortical defect should be based on considerations other than the potential for creating a distal stress riser after definitive fracture management.

Am J Orthop. 2017;46(2):E108-E111. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

References

1. Brooks DB, Burstein AH, Frankel VH. The biomechanics of torsional fractures. The stress concentration effect of a drill hole. J Bone Joint Surg Am. 1970;52(3):507-514.

2. Edgerton BC, An KN, Morrey BF. Torsional strength reduction due to cortical defects in bone. J Orthop Res. 1990;8(6):851-855.

3. Gardner MP, Chong AC, Pollock AG, Wooley PH. Mechanical evaluation of large-size fourth-generation composite femur and tibia models. Ann Biomed Eng. 2010;38(3):613-620.

4. Wysocki RW, Sheinkop MB, Virkus WW, Della Valle CJ. Femoral fracture through a previous pin site after computer-assisted total knee arthroplasty. J Arthroplasty. 2008;23(3):462-465.

5. Burstein AH, Currey J, Frankel VH, Heiple KG, Lunseth P, Vessely JC. Bone strength. The effect of screw holes. J Bone Joint Surg Am. 1972;54(6):1143-1156.

6. Clark CR, Morgan C, Sonstegard DA, Matthews LS. The effect of biopsy-hole shape and size on bone strength. J Bone Joint Surg Am. 1977;59(2):213-217.

7. Evans PE, Thomas WG. Tibial fracture through a traction-pin site. A report of two cases. J Bone Joint Surg Am. 1984;66(9):1475-1476.

8. Stoffel K, Dieter U, Stachowiak G, Gächter A, Kuster MS. Biomechanical testing of the LCP—how can stability in locked internal fixators be controlled? Injury. 2003;34(suppl 2):B11-B19.

9. Klaue K, Fengels I, Perren SM. Long-term effects of plate osteosynthesis: comparison of four different plates. Injury. 2000;31(suppl 2):B51-B62.

10. Uhthoff HK, Poitras P, Backman DS. Internal plate fixation of fractures: short history and recent developments. J Orthop Sci. 2006;11(2):118-126.

11. Takemoto RC, Sugi MT, Kummer F, Koval KJ, Egol KA. The effects of locked and unlocked neutralization plates on load bearing of fractures fixed with a lag screw. J Orthop Trauma. 2012;26(9):519-522.

12. Wagner M. General principles for the clinical use of the LCP. Injury. 2003;34(suppl 2):B31-B42.

13. Strauss EJ, Schwarzkopf R, Kummer F, Egol KA. The current status of locked plating: the good, the bad, and the ugly. J Orthop Trauma. 2008;22(7):479-486.

14. Elfar J, Menorca RM, Reed JD, Stanbury S. Composite bone models in orthopaedic surgery research and education. J Am Acad Orthop Surg. 2014;22(2):111-120.

Article PDF
Author and Disclosure Information

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Issue
The American Journal of Orthopedics - 46(2)
Publications
Topics
Page Number
E108-E111
Sections
Author and Disclosure Information

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Author and Disclosure Information

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Article PDF
Article PDF

Take-Home Points

  • The location of a bicortical defect in proximity to a tibia plate does not appear to affect the torsional stiffness or torsional failure strength of the bone.
  • External fixator pin placement should be based on considerations other than the potential for creating a distal stress riser after definitive fracture management.

A stress riser in cortical bone may be considered any abrupt change in the contour or consistency of the hollow structure, such as a surface defect, that not only weakens the bone but concentrates stresses at that transition point.1 A cortical defect that is 20% of the bone diameter is associated with a 34% decrease in torsional strength, thus representing a “stress riser.”2 High-energy and complex tibia fractures are often provisionally stabilized with external fixation that gives the soft tissues time to recover before definitive fracture fixation. Pin diameter for a medium-size tibia external fixator typically is 5.0 mm, resulting in a 10-mm defect in bicortical placement. Therefore, any tibia with a diameter of <50 mm is at risk for a stress riser fracture.

Although it had been established that sizable cortical defects can decrease the torsional strength of long bone,2 the effect of a plate in close proximity to a defect secondary to an empty external-fixator pin site on torsional strength has not been determined. We conducted a study to evaluate this effect. The null hypothesis was there would be no difference in tibia torsional strength attributable to varying the proximity of a tibia midshaft plate to a 5.0-mm bicortical defect.

Methods

Forty fourth-generation, medium-size left composite tibias (Pacific Research Laboratories) were divided into 8 groups of 5 bones (Figure 1).

Figure 1.
To represent the stress riser created by the removal of a 5.0-mm Schantz external fixation pin, we produced distal tibia bicortical defects in 6 of the groups by creating anterior-to-posterior 5.0-mm bicortical drill holes. The longitudinal location of these drill holes was varied in relation to the distal end of a 4.5-mm × 121-mm 6-hole locking plate (PeriLoc; Smith & Nephew) fixed in a nonlocking configuration and positioned across the tibia midline on the anterior-medial aspect. In the experimental plated groups, the bicortical defects were created 3.0, 2.0, and 1.0 cm distal to the plate end, with 1 plated group without a defect. The control groups consisted of equivalent defects in the same distal longitudinal locations, without plates attached, as well as an unplated group without a defect.

Torsion testing to failure was performed for all specimens in a manner similar to that described by Gardner and colleagues.3 Impression molds for the composite tibia constructed from polymethylmethacrylate encased the superior and distal ends, leaving 25.5 cm of exposed midshaft. This allowed the composites to be rigidly clamped into a materials testing system (858 Mini-Bionix; MTS) equipped with a 100.0-Nm torsional load cell (Figure 2).

Figure 2.
The constructs were preconditioned by rotating the superior end internally up to 15.0 Nm at a rate of 0.25 Nm/s for 2 complete cycles. Next, the constructs were preloaded axially to 20.0 N and then rotated at 0.25°/s until failure. Torsional load and torsional displacement were recorded and used to determine construct stiffness and failure load. Stiffness was calculated as the slope of the linear elastic portion of the load versus displacement curves between 20.0 Nm and 40.0 Nm. Failure load was defined as the highest load achieved before fracture. One-way analysis of variance with Tukey adjustment for multiple comparisons and α set at 0.05 were used to detect differences in failure stiffness and failure load between the 8 constructs.

Results

Graphical results for torsional stiffness are presented in Figure 3. R2 for all stiffness calculations was >0.99.

Figure 3.
There were no statistical differences in torsional stiffness between any of the groups. Graphical results for torsional failure load are presented in Figure 4.
Figure 4.
During torsion-to-failure testing, both plated and unplated specimens without distal cortical defect holes nearly exceeded the torque capacity of the load cell without failing, stopping the tests. For the specimens that failed, there were no statistical differences in failure torque. A slight trend toward higher failure loads in plated specimens with a hole in close proximity was seen in the 1.0-cm distal defect hole groups, with the plated specimen achieving a higher mean (SD) failure load, 78.14 (7.58) Nm, than the unplated group, 66.75 (1.84) Nm, but this was not significant (P = .06). Another slight trend toward lower failure load in unplated specimens as the defect moved proximally was seen between the unplated 3.0-cm defect group, 77.91 (6.08) Nm, and the unplated 1.0-cm defect group, 66.75 (1.84) Nm; this was also not significant (P = .07). Mode of failure for all specimens with bicortical defects, with or without a plate, was a spiral fracture that bisected the axis of the defect (Figure 5).
Figure 5.
Post hoc power analysis for each measure indicated statistical power of 80% for stiffness and 75% for failure torque.

 

 

Discussion

Many tibia fractures require provisional stabilization with an external fixator that spans the knee, because of the high-energy nature of the injury or other, higher-priority polytrauma concerns. When the patient or injury is suitable for definitive fixation, the external fixator typically is removed in favor of internal fixation with a plate and screws. Depending on the nature and location of the fracture and the subsequent plate, the empty cortical pin-site defects, often lying at varying distances from the distal end of the plate, can potentially serve as stress risers for fracture.4

Other studies have evaluated long-bone cortical defects biomechanically1,2,4 and clinically,5-7 and multiple studies have been conducted on the effects of plates on long-bone strength for fracture stabilization.8-13 The present study evaluated the torsional strength of long bones in the presence of a bicortical defect and the proximity of the defect to a plate. There were no differences in stiffness or failure load between any of the groups of plated and unplated fourth-generation composite tibias tested to failure in torsion with varying distal bicortical defects. Hypothetically, one would expect the torsional stiffness of these specimens to increase with the mere addition of a metallic diaphyseal plate. However, this study demonstrated that the addition of a plate did not affect the torsional stiffness or strength of the tibias. Clinically, it is common practice to place external fixator pins as far as possible outside the planned incision site for definitive fracture fixation. Thus, we also hypothesized that the presence of a bicortical pin-site defect and its proximity to the plate would alter the torsional strength of the tibia specimens, and that the distal pin-site defect’s location farthest from the plate would exhibit greater strength, but this did not occur. Although other studies have shown that the presence of bicortical defects decreases the strength of long bones, we were unable to quantify this decrease because the 2 intact groups of composites, plated and unplated, survived failure testing.

This study had several limitations, first being the use of composite tibias as opposed to human cadaver bone. Although fourth-generation composite bone models have been validated as a suitable and accurate biomechanical substitute for cadaver specimens,14 anatomical variations in cadaver tibias may transfer forces differently through plates, screws, and distal pin sites. In order to test plated specimens against the unplated controls, we did not simulate a mid-shaft fracture in any of the tibias. The pin-site defects were intended to reflect the mechanical effects of bicortical defects immediately after pin removal and in the absence of any degree of bone healing. Finally, this study focused on pin-site defects that were distal to a midshaft plate and that may not represent the effects of bicortical pin-site defects proximal to the plate.

Given the results of this biomechanical study in composite tibias, varying the proximity of a bicortical defect to a plate does not affect the torsional stiffness or torsional failure strength of the bone. Placement of an intended bicortical defect should be based on considerations other than the potential for creating a distal stress riser after definitive fracture management.

Am J Orthop. 2017;46(2):E108-E111. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

Take-Home Points

  • The location of a bicortical defect in proximity to a tibia plate does not appear to affect the torsional stiffness or torsional failure strength of the bone.
  • External fixator pin placement should be based on considerations other than the potential for creating a distal stress riser after definitive fracture management.

A stress riser in cortical bone may be considered any abrupt change in the contour or consistency of the hollow structure, such as a surface defect, that not only weakens the bone but concentrates stresses at that transition point.1 A cortical defect that is 20% of the bone diameter is associated with a 34% decrease in torsional strength, thus representing a “stress riser.”2 High-energy and complex tibia fractures are often provisionally stabilized with external fixation that gives the soft tissues time to recover before definitive fracture fixation. Pin diameter for a medium-size tibia external fixator typically is 5.0 mm, resulting in a 10-mm defect in bicortical placement. Therefore, any tibia with a diameter of <50 mm is at risk for a stress riser fracture.

Although it had been established that sizable cortical defects can decrease the torsional strength of long bone,2 the effect of a plate in close proximity to a defect secondary to an empty external-fixator pin site on torsional strength has not been determined. We conducted a study to evaluate this effect. The null hypothesis was there would be no difference in tibia torsional strength attributable to varying the proximity of a tibia midshaft plate to a 5.0-mm bicortical defect.

Methods

Forty fourth-generation, medium-size left composite tibias (Pacific Research Laboratories) were divided into 8 groups of 5 bones (Figure 1).

Figure 1.
To represent the stress riser created by the removal of a 5.0-mm Schantz external fixation pin, we produced distal tibia bicortical defects in 6 of the groups by creating anterior-to-posterior 5.0-mm bicortical drill holes. The longitudinal location of these drill holes was varied in relation to the distal end of a 4.5-mm × 121-mm 6-hole locking plate (PeriLoc; Smith & Nephew) fixed in a nonlocking configuration and positioned across the tibia midline on the anterior-medial aspect. In the experimental plated groups, the bicortical defects were created 3.0, 2.0, and 1.0 cm distal to the plate end, with 1 plated group without a defect. The control groups consisted of equivalent defects in the same distal longitudinal locations, without plates attached, as well as an unplated group without a defect.

Torsion testing to failure was performed for all specimens in a manner similar to that described by Gardner and colleagues.3 Impression molds for the composite tibia constructed from polymethylmethacrylate encased the superior and distal ends, leaving 25.5 cm of exposed midshaft. This allowed the composites to be rigidly clamped into a materials testing system (858 Mini-Bionix; MTS) equipped with a 100.0-Nm torsional load cell (Figure 2).

Figure 2.
The constructs were preconditioned by rotating the superior end internally up to 15.0 Nm at a rate of 0.25 Nm/s for 2 complete cycles. Next, the constructs were preloaded axially to 20.0 N and then rotated at 0.25°/s until failure. Torsional load and torsional displacement were recorded and used to determine construct stiffness and failure load. Stiffness was calculated as the slope of the linear elastic portion of the load versus displacement curves between 20.0 Nm and 40.0 Nm. Failure load was defined as the highest load achieved before fracture. One-way analysis of variance with Tukey adjustment for multiple comparisons and α set at 0.05 were used to detect differences in failure stiffness and failure load between the 8 constructs.

Results

Graphical results for torsional stiffness are presented in Figure 3. R2 for all stiffness calculations was >0.99.

Figure 3.
There were no statistical differences in torsional stiffness between any of the groups. Graphical results for torsional failure load are presented in Figure 4.
Figure 4.
During torsion-to-failure testing, both plated and unplated specimens without distal cortical defect holes nearly exceeded the torque capacity of the load cell without failing, stopping the tests. For the specimens that failed, there were no statistical differences in failure torque. A slight trend toward higher failure loads in plated specimens with a hole in close proximity was seen in the 1.0-cm distal defect hole groups, with the plated specimen achieving a higher mean (SD) failure load, 78.14 (7.58) Nm, than the unplated group, 66.75 (1.84) Nm, but this was not significant (P = .06). Another slight trend toward lower failure load in unplated specimens as the defect moved proximally was seen between the unplated 3.0-cm defect group, 77.91 (6.08) Nm, and the unplated 1.0-cm defect group, 66.75 (1.84) Nm; this was also not significant (P = .07). Mode of failure for all specimens with bicortical defects, with or without a plate, was a spiral fracture that bisected the axis of the defect (Figure 5).
Figure 5.
Post hoc power analysis for each measure indicated statistical power of 80% for stiffness and 75% for failure torque.

 

 

Discussion

Many tibia fractures require provisional stabilization with an external fixator that spans the knee, because of the high-energy nature of the injury or other, higher-priority polytrauma concerns. When the patient or injury is suitable for definitive fixation, the external fixator typically is removed in favor of internal fixation with a plate and screws. Depending on the nature and location of the fracture and the subsequent plate, the empty cortical pin-site defects, often lying at varying distances from the distal end of the plate, can potentially serve as stress risers for fracture.4

Other studies have evaluated long-bone cortical defects biomechanically1,2,4 and clinically,5-7 and multiple studies have been conducted on the effects of plates on long-bone strength for fracture stabilization.8-13 The present study evaluated the torsional strength of long bones in the presence of a bicortical defect and the proximity of the defect to a plate. There were no differences in stiffness or failure load between any of the groups of plated and unplated fourth-generation composite tibias tested to failure in torsion with varying distal bicortical defects. Hypothetically, one would expect the torsional stiffness of these specimens to increase with the mere addition of a metallic diaphyseal plate. However, this study demonstrated that the addition of a plate did not affect the torsional stiffness or strength of the tibias. Clinically, it is common practice to place external fixator pins as far as possible outside the planned incision site for definitive fracture fixation. Thus, we also hypothesized that the presence of a bicortical pin-site defect and its proximity to the plate would alter the torsional strength of the tibia specimens, and that the distal pin-site defect’s location farthest from the plate would exhibit greater strength, but this did not occur. Although other studies have shown that the presence of bicortical defects decreases the strength of long bones, we were unable to quantify this decrease because the 2 intact groups of composites, plated and unplated, survived failure testing.

This study had several limitations, first being the use of composite tibias as opposed to human cadaver bone. Although fourth-generation composite bone models have been validated as a suitable and accurate biomechanical substitute for cadaver specimens,14 anatomical variations in cadaver tibias may transfer forces differently through plates, screws, and distal pin sites. In order to test plated specimens against the unplated controls, we did not simulate a mid-shaft fracture in any of the tibias. The pin-site defects were intended to reflect the mechanical effects of bicortical defects immediately after pin removal and in the absence of any degree of bone healing. Finally, this study focused on pin-site defects that were distal to a midshaft plate and that may not represent the effects of bicortical pin-site defects proximal to the plate.

Given the results of this biomechanical study in composite tibias, varying the proximity of a bicortical defect to a plate does not affect the torsional stiffness or torsional failure strength of the bone. Placement of an intended bicortical defect should be based on considerations other than the potential for creating a distal stress riser after definitive fracture management.

Am J Orthop. 2017;46(2):E108-E111. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

References

1. Brooks DB, Burstein AH, Frankel VH. The biomechanics of torsional fractures. The stress concentration effect of a drill hole. J Bone Joint Surg Am. 1970;52(3):507-514.

2. Edgerton BC, An KN, Morrey BF. Torsional strength reduction due to cortical defects in bone. J Orthop Res. 1990;8(6):851-855.

3. Gardner MP, Chong AC, Pollock AG, Wooley PH. Mechanical evaluation of large-size fourth-generation composite femur and tibia models. Ann Biomed Eng. 2010;38(3):613-620.

4. Wysocki RW, Sheinkop MB, Virkus WW, Della Valle CJ. Femoral fracture through a previous pin site after computer-assisted total knee arthroplasty. J Arthroplasty. 2008;23(3):462-465.

5. Burstein AH, Currey J, Frankel VH, Heiple KG, Lunseth P, Vessely JC. Bone strength. The effect of screw holes. J Bone Joint Surg Am. 1972;54(6):1143-1156.

6. Clark CR, Morgan C, Sonstegard DA, Matthews LS. The effect of biopsy-hole shape and size on bone strength. J Bone Joint Surg Am. 1977;59(2):213-217.

7. Evans PE, Thomas WG. Tibial fracture through a traction-pin site. A report of two cases. J Bone Joint Surg Am. 1984;66(9):1475-1476.

8. Stoffel K, Dieter U, Stachowiak G, Gächter A, Kuster MS. Biomechanical testing of the LCP—how can stability in locked internal fixators be controlled? Injury. 2003;34(suppl 2):B11-B19.

9. Klaue K, Fengels I, Perren SM. Long-term effects of plate osteosynthesis: comparison of four different plates. Injury. 2000;31(suppl 2):B51-B62.

10. Uhthoff HK, Poitras P, Backman DS. Internal plate fixation of fractures: short history and recent developments. J Orthop Sci. 2006;11(2):118-126.

11. Takemoto RC, Sugi MT, Kummer F, Koval KJ, Egol KA. The effects of locked and unlocked neutralization plates on load bearing of fractures fixed with a lag screw. J Orthop Trauma. 2012;26(9):519-522.

12. Wagner M. General principles for the clinical use of the LCP. Injury. 2003;34(suppl 2):B31-B42.

13. Strauss EJ, Schwarzkopf R, Kummer F, Egol KA. The current status of locked plating: the good, the bad, and the ugly. J Orthop Trauma. 2008;22(7):479-486.

14. Elfar J, Menorca RM, Reed JD, Stanbury S. Composite bone models in orthopaedic surgery research and education. J Am Acad Orthop Surg. 2014;22(2):111-120.

References

1. Brooks DB, Burstein AH, Frankel VH. The biomechanics of torsional fractures. The stress concentration effect of a drill hole. J Bone Joint Surg Am. 1970;52(3):507-514.

2. Edgerton BC, An KN, Morrey BF. Torsional strength reduction due to cortical defects in bone. J Orthop Res. 1990;8(6):851-855.

3. Gardner MP, Chong AC, Pollock AG, Wooley PH. Mechanical evaluation of large-size fourth-generation composite femur and tibia models. Ann Biomed Eng. 2010;38(3):613-620.

4. Wysocki RW, Sheinkop MB, Virkus WW, Della Valle CJ. Femoral fracture through a previous pin site after computer-assisted total knee arthroplasty. J Arthroplasty. 2008;23(3):462-465.

5. Burstein AH, Currey J, Frankel VH, Heiple KG, Lunseth P, Vessely JC. Bone strength. The effect of screw holes. J Bone Joint Surg Am. 1972;54(6):1143-1156.

6. Clark CR, Morgan C, Sonstegard DA, Matthews LS. The effect of biopsy-hole shape and size on bone strength. J Bone Joint Surg Am. 1977;59(2):213-217.

7. Evans PE, Thomas WG. Tibial fracture through a traction-pin site. A report of two cases. J Bone Joint Surg Am. 1984;66(9):1475-1476.

8. Stoffel K, Dieter U, Stachowiak G, Gächter A, Kuster MS. Biomechanical testing of the LCP—how can stability in locked internal fixators be controlled? Injury. 2003;34(suppl 2):B11-B19.

9. Klaue K, Fengels I, Perren SM. Long-term effects of plate osteosynthesis: comparison of four different plates. Injury. 2000;31(suppl 2):B51-B62.

10. Uhthoff HK, Poitras P, Backman DS. Internal plate fixation of fractures: short history and recent developments. J Orthop Sci. 2006;11(2):118-126.

11. Takemoto RC, Sugi MT, Kummer F, Koval KJ, Egol KA. The effects of locked and unlocked neutralization plates on load bearing of fractures fixed with a lag screw. J Orthop Trauma. 2012;26(9):519-522.

12. Wagner M. General principles for the clinical use of the LCP. Injury. 2003;34(suppl 2):B31-B42.

13. Strauss EJ, Schwarzkopf R, Kummer F, Egol KA. The current status of locked plating: the good, the bad, and the ugly. J Orthop Trauma. 2008;22(7):479-486.

14. Elfar J, Menorca RM, Reed JD, Stanbury S. Composite bone models in orthopaedic surgery research and education. J Am Acad Orthop Surg. 2014;22(2):111-120.

Issue
The American Journal of Orthopedics - 46(2)
Issue
The American Journal of Orthopedics - 46(2)
Page Number
E108-E111
Page Number
E108-E111
Publications
Publications
Topics
Article Type
Display Headline
Effect of Plate in Close Proximity to Empty External-Fixation Pin Site on Long-Bone Torsional Strength
Display Headline
Effect of Plate in Close Proximity to Empty External-Fixation Pin Site on Long-Bone Torsional Strength
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

A Review of Psychostimulants for Adults With Depression

Article Type
Changed
Fri, 11/10/2017 - 11:04
A survey of medical literature suggests that for patients with depression who have not responded to other augmentation strategies, psychostimulants may offer improvements in mood, energy, and concentration.

Depression is a common condition that significantly impairs social and occupational functioning. Many patients do not respond to first-line pharmacotherapies and are considered to have treatment-resistant depression (TRD). These patients may benefit from augmentation of their antidepressant to reduce depression. Multiple medications have demonstrated various degrees of efficacy for augmentation, including psychostimulants. This article reviews studies of psychostimulants as augmentation agents for TRD and discusses risks, offers advice, and makes recommendations for clinicians who prescribe stimulants.

Background

Major depressive disorder (MDD) is a common psychiatric condition that significantly impairs quality of life.1 It is a recurrent illness, averaging 2 relapses per decade. The probability of recurrence increases with the number of depressive episodes.2,3 A patient who experiences major depressive episodes alternating with euthymia has unipolar depression; whereas one who experiences major depressive episodes alternating with episodes of mania or hypomania has bipolar depression.4

Despite adequate dose and duration of pharmacotherapy, many individuals with unipolar or bipolar depression do not achieve and sustain remission.5 Remission rates decrease and relapse rates increase with subsequent failed antidepressant trials.6 It is difficult to identify factors that predict treatment resistance, but one review of antidepressant studies found that patients who did not demonstrate a response within 3 weeks of medication initiation were less likely to respond after a longer duration.7

Treatment-resistant depression is commonly, but not universally, defined as lack of response after trials of 2 or more antidepressants with different mechanisms of action for sufficient duration.5 This definition will be used here as well. Other definitions have proposed stages of TRD, but these require further study to evaluate their reliability and predictive utility.8 Due to lack of consensus regarding the definition of TRD, it is not possible to determine the exact prevalence of TRD.

Patients with TRD may benefit from augmentation of their medication regimen. Augmentation with lithium has yielded conflicting results, and its efficacy with newer antidepressants is not well studied.9-12 Triiodothyronine, buspirone, and pindolol have demonstrated some efficacy when added to serotonin reuptake inhibitors (SRIs).10,12,13 Second-generation antipsychotic drugs, antidepressant drug combinations, omega-3 fatty acids, S-adenosyl methionine (SAMe), and L-methylfolate have demonstrated some efficacy in some studies as well.12,14-23 In patients with depression who have not responded to these strategies, psychostimulant augmentation may be appropriate.

Methods

A literature search was conducted following an algorithmic approach in the MEDLINE/PubMed database for studies in English from January 1985 to August 2014 of stimulants as augmenting agents for depression, using the Medical Subject Headings stimulant, depression, and augmentation, combined with an AND operator. The search was limited to adult humans and excluded case reports and letters, to identify studies with stronger evidence. Also excluded were studies using caffeine (to augment electroconvulsive therapy for depression) and pemoline as the sole augmenting stimulant as well as studies of patients with comorbid mental health diagnoses and studies that initiated stimulants and antidepressants simultaneously to assess antidepressant response.

This review organized results by stimulant rather than by depression type, even though some studies used > 1 stimulant or recruited patients with different types of depression. Although prevalence, prognosis, and monotherapy differ for unipolar and bipolar depression, psychostimulants target similar symptoms, despite augmenting different monotherapies in unipolar and bipolar depression. Therefore, no distinction is made between assessing studies of stimulants for unipolar and bipolar depression.

Results

A total of 70 articles were identified, and 31 studies met inclusion criteria (Figure). Of the studies included, 12 were double-blind, placebo-controlled (DBPC) trials and 19 were retrospective chart reviews or open studies. Most studies evaluated depression, using validated scales, such as the Hamilton Depression Rating Scale, Montgomery-Asberg Depression Rating Scale, Clinical Global Impressions of Severity, Inventory of Depressive Symptoms, Carroll Depression Rating Scale, Global Assessment of Functioning, Quick Inventory of Depressive Symptomatology, or the Psychiatric Symptom Assessment Scale. Study details are provided in Tables 1 to 4.

Dextroamphetamine and Methylphenidate

Dextroamphetamine and methylphenidate are indicated for the treatment of attention-deficit/hyperactivity disorder (ADHD) and exert their effects by inhibiting uptake of norepinephrine and dopamine.24 In one chart review, patients received dextroamphetamine or methylphenidate augmentation of monoamine oxidase inhibitors (MAOIs) alone or with concurrent tricyclic antidepressants; the majority reported decreased depression.25 In an openlabel trial, dextroamphetamine was titrated to efficacy in patients who were receiving an MAOI with or without pemoline.26 Nearly 80% of patients reported long-lasting improvement in depression. In an open-label trial, all patients reported decreased depression when methylphenidate was added to SRIs; however, no scales were used.27

In a case series, patients with both major depression and persistent depressive disorder (dysthymia) experienced a substantial, quick, and sustained response to dextroamphetamine or methylphenidate augmentation.28 Addition of lisdexamfetamine significantly reduced depressive symptoms in individuals with inadequate response to escitalopram.29 Patients with full or partial remission of depression noted improved executive function and residual depressive symptoms after lisdexamfetamine was added to SRI monotherapy.30 In a trial in which patients received dexamphetamine or methylphenidate as monotherapy or augmentation, 30% to 34% of patients reported mood improvement, but 36% reported no improvement.31 In an extension study, low-dose psychostimulants quickly diminished melancholia.32

Methylphenidate was safe and effective in patients with bipolar depression receiving treatment for 1 to 5 years; 44% evidenced significant improvement.33 When offered to patients with bipolar depression, patients receiving methylphenidate or dextroamphetamine reported less depression or sedation and did not develop tolerance, mania, or misuse.34 A case series concluded that methylphenidate addition to mood stabilizers was generally effective and safe.35

However, not all preparations of methylphenidate have demonstrated efficacy. In one study, osmotic controlledrelease oral system (OROS) methylphenidate improved apathy and fatigue but not overall depression.36 Although OROS methylphenidate similarly failed to demonstrate statistically significant efficacy in another study, more responders were documented in the treatment group.37

Although this review focuses on stimulants as augmenting agents in patients with depression, it is worth noting the limited number of studies evaluating stimulants’ effect on depression in patients with traumatic brain injury. This observation is of concern, as these conditions are frequently comorbid in returning veterans. One study noted that methylphenidate was an effective monotherapy for depression; whereas another study found that methylphenidate monotherapy reduced depression as well as sertraline, was better tolerated, and improved fatigue and cognition.38,39

Modafinil and Armodafinil

Modafinil and armodafinil (the R-enantiomer of modafinil) are indicated for improving wakefulness in individuals with narcolepsy, obstructive sleep apnea, and shift work sleep disorder by modulating glutamate, gamma amino-butyric acid, and histamine.40,41 Although they increase extracellular dopamine concentrations, they do not cause an increase in dopamine release and may have less misuse potential than that of dextroamphetamine and methylphenidate.40,41 In a study of 7 patients with unipolar or bipolar depression, all patients achieved full or partial remission with minimal adverse effects (AEs).42 In a prospective study, 41% of patients reported only mild depression or full remission with modafinil augmentation.43

Multiple trials and a pooled analysis noted decreased depression and fatigue and improved cognition in patients receiving modafinil augmentation compared with mood stabilizers or antidepressants.44-49 Modafinil is a useful adjunct for partial responders to SRIs, resulting in rapid mood improvement and decreased fatigue.50-54 However, in one study, modafinil did not demonstrate efficacy compared with placebo. This result was attributed to premature study termination after 2 modafinil-treated patients developed suicidal ideation.55 A post hoc analysis found no difference in frequency of suicidal ideation between groups.

Two DBPC studies evaluated armodafinil in patients with bipolar depression. In both studies it was added to a mood-stabilizing agent (lithium, valproate, aripiprazole, olanzapine, lamotrigine, risperidone, or ziprasidone), and patients receiving armodafinil reported significant reductions
in depression.56,57

Atomoxetine

Atomoxetine is a norepinephrine reuptake inhibitor indicated for the treatment of ADHD and is considered to have no misuse potential due to lack of dopamine modulation.58 In one study, 15 patients received atomoxetine added to their antidepressant, and 60% experienced significant symptom reduction.59 A chart review noted decreases in fatigue and depression when atomoxetine was added to an SRI, mirtazapine, or amitriptyline.60 However, in a DBPC trial, atomoxetine did not lead to significant changes in depression.61

Discussion

There is a limited amount of high-quality evidence to support psychostimulant augmentation, as noted by the relatively few DBPC trials, most of short duration. The evidence supports their efficacy primarily for unipolar depression, as 14 studies evaluated patients with unipolar depression, whereas only 7 studies evaluated patients with bipolar depression. The remaining studies recruited patients with both depression types. Collectively, modafinil and armodafinil have the most evidence in DBPC trials.

There are relatively few DBPC trials with high power and sufficient duration for dextroamphetamine and methylphenidate preparations. This discovery is surprising, considering the duration that these medications have been available. However, several chart reviews and open-label trials provided some evidence to support their use in patients without a history of substance misuse or cardiac conditions.62 Osmotic controlled- release oral system methylphenidate seems to be ineffective, and the efficacy of atomoxetine for augmentation
is uncertain.

Precautions

Prescribing physicians who offer stimulants should consider potential AEs, such as psychosis, anorexia, anxiety, insomnia, mood changes (eg, anger),  misuse, addiction, mania, and cardiovascular problems. Psychostimulants have been implicated in precipitating psychosis.63,64 However, in a 12-month study of 250 adults with ADHD, 73 reported AEs, and only 31 discontinued the stimulant. Adverse effects leading to discontinuation included mood instability (n = 7), agitation (n = 6), irritability (n = 4), or decreased appetite (n = 4).65

Although associated with the risks of anorexia and insomnia in patients with ADHD, methylphenidate rapidly improved daytime sleepiness and mood, and—paradoxically—appetite and nighttime sleep in medically ill elderly patients with depression.66 Misuse or abuse of methylphenidate and dextroamphetamine were noted in 23% of patients referred for substance misuse.67 Nonetheless, little evidence exists that these drugs possess significant misuse potential in patients taking them as prescribed. As a prodrug, lisdexamfetamine is hypothesized to have less abuse potential compared with dextroamphetamine and methylphenidate, but it carries the same prescribing and monitoring precautions.68 Risks related to stimulant usage extend to manic symptoms.69 Patients with bipolar disorder should not receive stimulants if they have a history of stimulant-induced mania, rapid cycling, or psychosis.70

Long-term cardiovascular safety data exist for dextroamphetamine and methylphenidate but are limited or unavailable for modafinil, armodafinil, and atomoxetine. A retrospective cohort study found no significant increase in the number of cardiac events in patients receiving dextroamphetamine,
methylphenidate, or atomoxetine for an average of 1 year compared with controls.71 Another cohort study of > 44,000 patients found that initiation of
methylphenidate was associated with increased risk of sudden death or arrhythmia, but the risk was attributed to an unmeasured confounding factor, as the authors found a negative correlation between methylphenidate dose and all cardiovascular events.72

Recent practice guidelines recommend that before prescribing stimulants, clinicians should perform a physical examination (including heart and lung auscultation), obtain vital signs and height and weight, and request an electrocardiogram in case of abnormal findings on a cardiovascular examination or in case of a personal or family history of heart disease. Before offering atomoxetine, clinicians should evaluate the patient for a history of liver disease (and check liver function studies in case of a positive history). Clinicians should also assess risk of self-harm prior to initiating psychostimulant therapy.73 Throughout treatment, clinicians should evaluate the patient for changes in blood pressure, pulse, weight or mood, as well as the development of dependence or misuse. Urine toxicology testing is recommended for dextroamphetamine and methylphenidate to screen for adherence and diversion.

Limitations

Using only PubMed and MEDLINE databases limited the search to articles published in English after 1985, excluding letters and case reports to identify studies with higher evidence (the studies were not weighted based on study design). In addition, the studies had certain limitations. These include a limited number of DBPC trials, most were of short duration. It is also difficult to compare studies due to various rating scales used and concurrent
medication regimens of study subjects. These limitations raise questions surrounding the long-term efficacy of stimulants, and there is no consensus for how long a stimulant should be continued if beneficial. Longer, higherpowered, DBPC trials are warranted to determine longterm efficacy and safety of stimulant augmentation.62

Conclusion

For patients with depression who have not responded to other augmentation strategies, psychostimulants may be offered to improve mood, energy, and concentration. For clinicians considering stimulant augmentation, modafinil and armodafinil are reasonable choices given their efficacy in double-blind, placebo-controlled trials and lower risk of misuse. Dextroamphetamine (particularly lisdexamphetamine) and methylphenidate may be appropriate for patients who have not benefited from or tolerated modafinil or armodafinil, provided these patients do not have a medical history of cardiac disease or current substance use.

Osmotic controlled-release oral system methylphenidate seems to be ineffective as an augmenting agent. The efficacy of atomoxetine for augmentation is questionable, but atomoxetine could be offered if other stimulants were contraindicated, ineffective, or poorly tolerated. Both OROS methylphenidate and atomoxetine should be evaluated in additional trials before they can be recommended as augmentation therapies. Certain psychostimulants may be appropriate and reasonable adjunctive pharmacotherapies for patients with unipolar or bipolar depression who have failed other augmentation strategies, for patients who have significant fatigue or cognitive complaints, or for elderly patients with melancholic or somatic features of depression.

Acknowledgements
The authors thank Maureen Humphrey-Shelton and Kathy Thomas for their help in obtaining references.

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

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

Click here to read the digital edition.

References

1. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distribution of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):593-602.

2. Solomon DA, Keller MB, Leon AC, et al. Multiple recurrences of major depressive disorder. Am J Psychiatry. 2000;157(2):229-233.

3. Katon WJ, Fan MY, Lin EH, Unützer J. Depressive symptom deterioration in a large
primary care-based elderly cohort. Am J Geriatr Psychiatry. 2006;14(3):246-254.

4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Arlington, VA: American Psychiatric Association; 2013.

5. McIntyre RS, Filteau M-J, Martin L, et al. Treatment-resistant depression: Definitions, review of the evidence, and algorithmic approach. J Affect Disord. 2014;156:1-7.

6. Gaynes BN, Warden D, Trivedi MH, Wisniewski SR, Fava M, Rush AJ. What did STAR*D teach us? Results from a large-scale, practical, clinical trial for patients with depression. Focus. 2012;10(4):510-517.

7. Kudlow PA, Cha DS, McIntyre RS. Predicting treatment response in major depressive disorder: The impact of early symptomatic improvement. Can J Psychiatry. 2012;57(12):782-788.

8. Ruhé HG, van Rooijen G, Spijker J, Peeters FP, Schene AH. Staging methods for treatment resistant depression. A systematic review. J Affect Disord. 2012;137(1-3):35-45.

9. Bauer M, Dopfmer S. Lithium augmentation treatment-resistant depression: Metaanalysis of placebo-controlled studies. J Clin Psychopharmacol. 1999;19(5):427-434.

10. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: A STAR*D report. Am J Psychiatry. 2006;163(9):1519-1530.

11. Nierenberg AA, Papakostas GI, Petersen T, et al. Lithium augmentation of nortriptyline
for subjects resistant to multiple antidepressants. J Clin Psychopharmacol. 2003;23(1):92-95.

12. Connolly KR, Thase ME. If at first you don’t succeed: A review of the evidence for antidepressant augmentation, combination, and switching strategies. Drugs. 2011;71(1):43-64.

13. Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team. Medication augmentation after the failure of SSRIs for depression. N Engl J Med. 2006;354(12):1243-1252.

14. Papakostas GI, Shelton RC, Smith J, Fava M. Augmentation of antidepressants with atypical antipsychotic medications for treatment resistant major depressive disorder: A meta-analysis. J Clin Psychiatry. 2007;68(6):826-831.

15. Mahmoud RA, Pandina GJ, Turkoz I, et al. Risperidone for treatment-refractory major depressive disorder: A randomized trial. Ann Intern Med. 2007;147(9):593-602.

16. Barbee JG, Conrad EJ, Jamhour NJ. The effectiveness of olanzapine, risperidone, quetiapine, and ziprasidone as augmentation agents in treatment resistant depressive disorder. J Clin Psychiatry. 2004;65(7):975-981.

17. Fatemi SH, Emamian ES, Kist DA. Venlafaxine and bupropion combination therapy in a case of treatment-resistant depression. Ann Pharmacother.1999;33(6):701-703.

18. Carpenter LL, Yasman S, Price LH. A double-blind, placebo-controlled study of antidepressant augmentation with mirtazapine. Biol Psychiatry. 2002;51(2):183-188.

19. Hannan N, Hamzah Z, Akinpeloye HO, Meagher D. Venlafaxine-mirtazapine combination therapy in the treatment of persistent depressive illness. J Psychopharmacol. 2007;21(2):161-164.

20. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: A STAR*D report. Am J Psychiatry. 2006;163(9):1531-1541.

21. Blier P, Ward HE, Tremblay P, Laberge L, Hébert C, Bergeron R. Combination of antidepressant medications from treatment initiation for major depressive disorder: A double-blind randomized study. Am J Psychiatry. 2010;167(3):281-288.

22. Papakostas GI, Mischoulon D, Shyu I, Alpert JE, Fava M. S-adenosyl methionine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: A double blind randomized clinical trial. Am J Psychiatry. 2010;167(8):942-948.

23. Papakostas GI, Shelton RC, Zajecka JM, et al. L-methylfolate as adjunctive therapy
for SSRI-resistant major depression: Results of two randomized, double-blind,
parallel-sequential trials. Am J Psychiatry. 2012;169(12):1267-1274.

24. Korston TR. Drugs of abuse. In: Katzung BG, ed. Basic and Clinical Pharmacology. 9th ed. New York, NY: McGraw-Hill; 2004:521-523.

25. Feighner JP, Herbstein J, Damlouji N. Combined MAOI, TCA, and direct stimulant therapy of treatment-resistant depression. J Clin Psychiatry. 1985;46(6):206-209.

26. Fawcett J, Kravitz HM, Zajecka JM, Schaff MR. CNS stimulant potentiation of monoamine oxidase inhibitors in treatment-refractory depression. J Clin Psychopharmacol. 1991;11(2):127-132.

27. Stoll AL, Pillay SS, Diamond L, Workum SB, Cole JO. Methylphenidate augmentation of serotonin selective reuptake inhibitors: A case series. J Clin Psychiatry. 1996;57(2):72-76.

28. Masand PS, Anand VS, Tanquary JF. Psychostimulant augmentation of second generation antidepressants: A case series. Depress Anxiety. 1998;7(2):89-91.

29. Trivedi MH, Cutler AJ, Richards C, et al. A randomized control trial of the efficacy and safety of lisdesxamfetamine dimesylate as augmentation therapy in adults with residual symptoms of major depressive disorder after treatment with escitalopram. J Clin Psychiatry. 2013;74(8):802-809.

30. Madhoo M, Keefe RS, Roth RM, et al. Lisdexamfetamine dimesylate augmentation in adults with persistent executive dysfunction after partial or full remission of major depressive disorder. Neuropsychopharmacology. 2014;39(6):1388-1398.

31. Parker G, Brotchie H. Do the old psychostimulant drugs have a role in managing treatment-resistant depression. Acta Psychiatr Scand. 2010;121(4):308-314.

32. Parker G, Brotchie H, McClure G, Fletcher K. Psychostimulants for managing unipolar and bipolar treatment-resistant melancholic depression: A medium term evaluation of cost benefits. J Affect Disord. 2013;151(1):360-364.

33. Lydon E, El-Mallakh RS. Naturalistic long-term use of methylphenidate in bipolar disorder. J Clin Psychopharmacol. 2006;26(5):516-518.

34. Carlson PJ, Merlock MC, Suppes T. Adjunctive stimulant use in patients with bipolar disorder: Treatment of residual depression and sedation. Bipolar Disord. 2004;6(5):416-420.

35. El-Mallakh RS. An open study of methylphenidate in bipolar depression. Bipolar Disord. 2000;2(1):56-59.

36. Ravindran AV, Kennedy SH, O’Donovan MC, Fallu A, Camacho F, Binder CE. Osmotic-release oral system methylphenidate augmentation of antidepressant monotherapy in major depressive disorder: Results of a double-blind, randomized, placebo-controlled trial. J Clin Psychiatry. 2008;69(1):87-94.

37. Patkar AA, Masand PS, Pae CU, et al. A randomized, double-blind, placebocontrolled
trial of augmentation with an extended release formulation of methylphenidate in outpatients with treatment-resistant depression. J Clin Psychopharmacol. 2006;26(6):653-656.

38. Lee H, Kim SW, Kim JM, Shin IS, Yang SJ, Yoon JS Comparing effects of methylphenidate, sertraline, and placebo on neuropsychiatric sequelae in patients with
traumatic brain injury. Hum Psychopharmacol. 2005;20(2):97-104.

39. Gualtieri CT, Evans RW. Stimulant treatment for the neurobehavioural sequelae of traumatic brain injury. Brain Inj. 1988;2(4):273-290.

40. Provigil [package insert]. North Wales, PA: Cephalon Inc; 2015.

41. Nuvigil [package insert]. Frazer, PA: Cephalon, Inc; 2013.

42. Menza MA, Kaufman KR, Castellanos A. Modafinil augmentation of antidepressant treatment in depression. J Clin Psychiatry. 2000;61(5):378-381.

43. Markovitz PJ, Wagner S. An open-label trial of modafinil augmentation in patients with partial response to antidepressant therapy. J Clin Psychopharmacol. 2003;23(2):207-209.

44. Fernandes PP, Petty F. Modafinil for remitted bipolar depression with hypersomnia. Ann Pharmacother. 2003;37(12):1807-1809.

45. Nasr S. Modafinil as adjunctive therapy in depressed outpatients. Ann Clin Psychiatry. 2004;16(3):133-138.

46. DeBattista C, Lembke A, Solvason HB, Ghebremichael R, Poirier J. A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol. 2004;24(1):87-90.

47. Nasr S, Wendt B, Steiner K. Absence of mood switch with and tolerance to modafinil: A replication study from a large private practice. J Affect Disord. 2006;95(1-3):111-114.

48. DeBattista C, Doghramji K, Menza MA, Rosenthal MH, Fieve RR; Modafinil in Depression Study Group. Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: A preliminary doubleblind, placebo-controlled study. J Clin Psychiatry. 2003;64(9):1057-1064.

49. Frye MA, Grunze H, Suppes T, et al. A placebo-controlled evaluation of adjunctive modafinil in the treatment of bipolar depression. Am J Psychiatry. 2007;164(8):1242-1249.

50. Fava M, Thase ME, DeBattista C, Doghramji K, Arora S, Hughes RJ. Modafinil augmentation of selective serotonin reuptake inhibitor therapy in MDD partial responders with persistent fatigue and sleepiness. Ann Clin Psychiatry. 2007;19(3):153-159.

51. Thase ME, Fava M, DeBattista C, Arora S, Hughes RJ. Modafinil augmentation of SSRI therapy in patients with major depressive disorder and excessive sleepiness and fatigue: A 12-week, open-label, extension study. CNS Spectr. 2006;11(2):93-102.

52. Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry. 2005;66(1):85-93.

53. Abolfazli R, Hosseini M, Ghanizadeh A, et al. Double-blind randomized parallelgroup clinical trial of efficacy of the combination fluoxetine plus modafinil versus fluoxetine plus placebo in the treatment of major depression. Depress Anxiety. 2011;28(4):297-302.

54. Rasmussen NA, Schrøder P, Olsen LR, Brødsgaard M, Undén M, Bech P. Modafinil augmentation in depressed patients with partial response to antidepressants: A pilot study on self-reported symptoms covered by the Major Depression Inventory (MDI) and the Symptom Checklist (SCL-92). Nord J Psychiatry. 2005;59(3):173-178.

55. Dunlop BW, Crits-Christoph P, Evans DL, et al. Coadministration of modafinil and a selective serotonin reuptake inhibitor from the initiation of treatment of major depressive disorder with fatigue and sleepiness: A double-blind, placebocontrolled study. J Clin Psychopharmacol. 2007;27(6):614-619.

56. Calabrese JR, Ketter TA, Youakim JM, Tiller JM, Yang R, Frye MA. Adjunctive armodafinil
for major depressive episodes associated with bipolar I disorder: A randomized multicenter, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2010;71(10):1363-1370.

57. Calabrese JR, Frye MA, Yang R, Ketter TA; Armodafinil Treatment Trial Study Network. Efficacy and safety of adjunctive armodafinil in adults with major depressive episodes associated with bipolar I disorder: A randomized, double-blind, placebo-controlled, multicenter trial. J Clin Psychiatry. 2014;75(10):1054-1061.

58. Strattera [package insert]. Indianapolis, IN. Lilly; 2015.

59. Carpenter LL, Milosavljevic N, Schecter JM, Tyrka AR, Price LH. Augmentation with open-label atomoxetine for partial or nonresponse to antidepressants. J Clin Psychiatry. 2005;66(10):1234-1238.

60. Papakostas GI, Petersen TJ, Burns AM, Fava M. Adjunctive atomoxetine for residual
fatigue in major depressive disorder. J Psychiatr Res. 2006;40(4):370-373.

61. Michelson D, Adler LA, Amsterdam JD, et al. Addition of atomoxetine for depression
incompletely responsive to sertraline: A randomized, double-blind, placebocontrolled study. J Clin Psychiatry. 2007;68(4):582-587.

62. Corp SA, Gitlin MJ, Altshuler LL. A review of the use of stimulants and stimulant alternatives in treating bipolar depression and major depressive disorder. J Clin Psychiatry. 2014;75(9):1010-1018.

63. Kraemer M, Uekermann J, Wiltfang J, Kis B. Methylphenidate-induced psychosis in adult attention-deficit/hyperactivity disorder: Report of 3 new cases and review of the literature. Clin Neuropharmacol. 2010;33(4):204-206.

64. Berman SM, Kuczenski R, McCracken JT, London ED. Potential adverse effects of amphetamine treatment on brain and behavior: A review. Mol Psychiatry. 2009;14(2):123-142.

65. Fredriksen M, Dahl AA, Martinsen EW, Klungsøyr O, Haavik J, Peleikis DE. Effectiveness of one-year pharmacological treatment of adult attention-deficit/hyperactivity disorder (ADHD): An open-label prospective study of time in treatment, dose, side-effects and comorbidity. Eur Neuropsychopharmacol. 2014;24(12):1873-1874.

66. Hardy SE. Methylphenidate for the treatment of depressive symptoms, including fatigue and apathy, in medically ill older adults and terminally ill adults. Am J Geriatr Pharmacother. 2009;7(1):34-59.

67. Williams RJ, Goodale LA, Shay-Fiddler MA, Gloster SP, Chang SY. Methylphenidate and dextroamphetamine abuse in substance-abusing adolescents. Am J Addict. 2004;13(4):381-389.

68. Madaan V, Kolli V, Bestha DP, Shah MJ. Update on optimal use of lisdexamfetamine in the treatment of ADHD. Neuropsychiatr Dis Treat. 2013;9:977-983.

69. Ross RG. Psychotic and manic-like symptoms during stimulant treatment of attention deficit hyperactivity disorder. Am J Psychiatry. 2006;163(7):1149-1152.

70. Dell’Osso B, Ketter TA. Use of adjunctive stimulants in adult bipolar depression. Int J Neuropsychopharmacol. 2013;16(1):55-68.

71. Habel LA, Cooper WO, Sox CM, et al. ADHD medications and risk of serious cardiovascular events in young and middle-aged adults. JAMA. 2011;306(24):2673-2683.

72. Schelleman H, Bilker WB, Kimmel SE, et al. Methylphenidate and risk of serious cardiovascular events in adults. Am J Psychiatry. 2012;169(2):178-185.

73. Bolea-Alamañac B, Nutt DJ, Adamou M, et al; British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological management of attention deficit hyperactivity disorder: Update on recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(3):179-203.

74. Moher D, Liberati A, Tetzlaff J, Altman DG; The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009;6(6):e1000097.

Author and Disclosure Information

Drs. Pary, Jijakli, and Tobias are psychiatrists at the Robley Rex VAMC in Louisville, Kentucky. Dr. Scarff is a psychiatrist at the William Jennings Bryan Dorn Veterans Affairs Community Based Outpatient Clinic in Spartanburg, South Carolina. Dr. Lippmann is professor emeritus in the Department of Psychiatry and Behavioral Sciences at the University of Louisville School of Medicine in Kentucky.

Issue
Federal Practitioner - 32(3)s
Publications
Topics
Sections
Author and Disclosure Information

Drs. Pary, Jijakli, and Tobias are psychiatrists at the Robley Rex VAMC in Louisville, Kentucky. Dr. Scarff is a psychiatrist at the William Jennings Bryan Dorn Veterans Affairs Community Based Outpatient Clinic in Spartanburg, South Carolina. Dr. Lippmann is professor emeritus in the Department of Psychiatry and Behavioral Sciences at the University of Louisville School of Medicine in Kentucky.

Author and Disclosure Information

Drs. Pary, Jijakli, and Tobias are psychiatrists at the Robley Rex VAMC in Louisville, Kentucky. Dr. Scarff is a psychiatrist at the William Jennings Bryan Dorn Veterans Affairs Community Based Outpatient Clinic in Spartanburg, South Carolina. Dr. Lippmann is professor emeritus in the Department of Psychiatry and Behavioral Sciences at the University of Louisville School of Medicine in Kentucky.

A survey of medical literature suggests that for patients with depression who have not responded to other augmentation strategies, psychostimulants may offer improvements in mood, energy, and concentration.
A survey of medical literature suggests that for patients with depression who have not responded to other augmentation strategies, psychostimulants may offer improvements in mood, energy, and concentration.

Depression is a common condition that significantly impairs social and occupational functioning. Many patients do not respond to first-line pharmacotherapies and are considered to have treatment-resistant depression (TRD). These patients may benefit from augmentation of their antidepressant to reduce depression. Multiple medications have demonstrated various degrees of efficacy for augmentation, including psychostimulants. This article reviews studies of psychostimulants as augmentation agents for TRD and discusses risks, offers advice, and makes recommendations for clinicians who prescribe stimulants.

Background

Major depressive disorder (MDD) is a common psychiatric condition that significantly impairs quality of life.1 It is a recurrent illness, averaging 2 relapses per decade. The probability of recurrence increases with the number of depressive episodes.2,3 A patient who experiences major depressive episodes alternating with euthymia has unipolar depression; whereas one who experiences major depressive episodes alternating with episodes of mania or hypomania has bipolar depression.4

Despite adequate dose and duration of pharmacotherapy, many individuals with unipolar or bipolar depression do not achieve and sustain remission.5 Remission rates decrease and relapse rates increase with subsequent failed antidepressant trials.6 It is difficult to identify factors that predict treatment resistance, but one review of antidepressant studies found that patients who did not demonstrate a response within 3 weeks of medication initiation were less likely to respond after a longer duration.7

Treatment-resistant depression is commonly, but not universally, defined as lack of response after trials of 2 or more antidepressants with different mechanisms of action for sufficient duration.5 This definition will be used here as well. Other definitions have proposed stages of TRD, but these require further study to evaluate their reliability and predictive utility.8 Due to lack of consensus regarding the definition of TRD, it is not possible to determine the exact prevalence of TRD.

Patients with TRD may benefit from augmentation of their medication regimen. Augmentation with lithium has yielded conflicting results, and its efficacy with newer antidepressants is not well studied.9-12 Triiodothyronine, buspirone, and pindolol have demonstrated some efficacy when added to serotonin reuptake inhibitors (SRIs).10,12,13 Second-generation antipsychotic drugs, antidepressant drug combinations, omega-3 fatty acids, S-adenosyl methionine (SAMe), and L-methylfolate have demonstrated some efficacy in some studies as well.12,14-23 In patients with depression who have not responded to these strategies, psychostimulant augmentation may be appropriate.

Methods

A literature search was conducted following an algorithmic approach in the MEDLINE/PubMed database for studies in English from January 1985 to August 2014 of stimulants as augmenting agents for depression, using the Medical Subject Headings stimulant, depression, and augmentation, combined with an AND operator. The search was limited to adult humans and excluded case reports and letters, to identify studies with stronger evidence. Also excluded were studies using caffeine (to augment electroconvulsive therapy for depression) and pemoline as the sole augmenting stimulant as well as studies of patients with comorbid mental health diagnoses and studies that initiated stimulants and antidepressants simultaneously to assess antidepressant response.

This review organized results by stimulant rather than by depression type, even though some studies used > 1 stimulant or recruited patients with different types of depression. Although prevalence, prognosis, and monotherapy differ for unipolar and bipolar depression, psychostimulants target similar symptoms, despite augmenting different monotherapies in unipolar and bipolar depression. Therefore, no distinction is made between assessing studies of stimulants for unipolar and bipolar depression.

Results

A total of 70 articles were identified, and 31 studies met inclusion criteria (Figure). Of the studies included, 12 were double-blind, placebo-controlled (DBPC) trials and 19 were retrospective chart reviews or open studies. Most studies evaluated depression, using validated scales, such as the Hamilton Depression Rating Scale, Montgomery-Asberg Depression Rating Scale, Clinical Global Impressions of Severity, Inventory of Depressive Symptoms, Carroll Depression Rating Scale, Global Assessment of Functioning, Quick Inventory of Depressive Symptomatology, or the Psychiatric Symptom Assessment Scale. Study details are provided in Tables 1 to 4.

Dextroamphetamine and Methylphenidate

Dextroamphetamine and methylphenidate are indicated for the treatment of attention-deficit/hyperactivity disorder (ADHD) and exert their effects by inhibiting uptake of norepinephrine and dopamine.24 In one chart review, patients received dextroamphetamine or methylphenidate augmentation of monoamine oxidase inhibitors (MAOIs) alone or with concurrent tricyclic antidepressants; the majority reported decreased depression.25 In an openlabel trial, dextroamphetamine was titrated to efficacy in patients who were receiving an MAOI with or without pemoline.26 Nearly 80% of patients reported long-lasting improvement in depression. In an open-label trial, all patients reported decreased depression when methylphenidate was added to SRIs; however, no scales were used.27

In a case series, patients with both major depression and persistent depressive disorder (dysthymia) experienced a substantial, quick, and sustained response to dextroamphetamine or methylphenidate augmentation.28 Addition of lisdexamfetamine significantly reduced depressive symptoms in individuals with inadequate response to escitalopram.29 Patients with full or partial remission of depression noted improved executive function and residual depressive symptoms after lisdexamfetamine was added to SRI monotherapy.30 In a trial in which patients received dexamphetamine or methylphenidate as monotherapy or augmentation, 30% to 34% of patients reported mood improvement, but 36% reported no improvement.31 In an extension study, low-dose psychostimulants quickly diminished melancholia.32

Methylphenidate was safe and effective in patients with bipolar depression receiving treatment for 1 to 5 years; 44% evidenced significant improvement.33 When offered to patients with bipolar depression, patients receiving methylphenidate or dextroamphetamine reported less depression or sedation and did not develop tolerance, mania, or misuse.34 A case series concluded that methylphenidate addition to mood stabilizers was generally effective and safe.35

However, not all preparations of methylphenidate have demonstrated efficacy. In one study, osmotic controlledrelease oral system (OROS) methylphenidate improved apathy and fatigue but not overall depression.36 Although OROS methylphenidate similarly failed to demonstrate statistically significant efficacy in another study, more responders were documented in the treatment group.37

Although this review focuses on stimulants as augmenting agents in patients with depression, it is worth noting the limited number of studies evaluating stimulants’ effect on depression in patients with traumatic brain injury. This observation is of concern, as these conditions are frequently comorbid in returning veterans. One study noted that methylphenidate was an effective monotherapy for depression; whereas another study found that methylphenidate monotherapy reduced depression as well as sertraline, was better tolerated, and improved fatigue and cognition.38,39

Modafinil and Armodafinil

Modafinil and armodafinil (the R-enantiomer of modafinil) are indicated for improving wakefulness in individuals with narcolepsy, obstructive sleep apnea, and shift work sleep disorder by modulating glutamate, gamma amino-butyric acid, and histamine.40,41 Although they increase extracellular dopamine concentrations, they do not cause an increase in dopamine release and may have less misuse potential than that of dextroamphetamine and methylphenidate.40,41 In a study of 7 patients with unipolar or bipolar depression, all patients achieved full or partial remission with minimal adverse effects (AEs).42 In a prospective study, 41% of patients reported only mild depression or full remission with modafinil augmentation.43

Multiple trials and a pooled analysis noted decreased depression and fatigue and improved cognition in patients receiving modafinil augmentation compared with mood stabilizers or antidepressants.44-49 Modafinil is a useful adjunct for partial responders to SRIs, resulting in rapid mood improvement and decreased fatigue.50-54 However, in one study, modafinil did not demonstrate efficacy compared with placebo. This result was attributed to premature study termination after 2 modafinil-treated patients developed suicidal ideation.55 A post hoc analysis found no difference in frequency of suicidal ideation between groups.

Two DBPC studies evaluated armodafinil in patients with bipolar depression. In both studies it was added to a mood-stabilizing agent (lithium, valproate, aripiprazole, olanzapine, lamotrigine, risperidone, or ziprasidone), and patients receiving armodafinil reported significant reductions
in depression.56,57

Atomoxetine

Atomoxetine is a norepinephrine reuptake inhibitor indicated for the treatment of ADHD and is considered to have no misuse potential due to lack of dopamine modulation.58 In one study, 15 patients received atomoxetine added to their antidepressant, and 60% experienced significant symptom reduction.59 A chart review noted decreases in fatigue and depression when atomoxetine was added to an SRI, mirtazapine, or amitriptyline.60 However, in a DBPC trial, atomoxetine did not lead to significant changes in depression.61

Discussion

There is a limited amount of high-quality evidence to support psychostimulant augmentation, as noted by the relatively few DBPC trials, most of short duration. The evidence supports their efficacy primarily for unipolar depression, as 14 studies evaluated patients with unipolar depression, whereas only 7 studies evaluated patients with bipolar depression. The remaining studies recruited patients with both depression types. Collectively, modafinil and armodafinil have the most evidence in DBPC trials.

There are relatively few DBPC trials with high power and sufficient duration for dextroamphetamine and methylphenidate preparations. This discovery is surprising, considering the duration that these medications have been available. However, several chart reviews and open-label trials provided some evidence to support their use in patients without a history of substance misuse or cardiac conditions.62 Osmotic controlled- release oral system methylphenidate seems to be ineffective, and the efficacy of atomoxetine for augmentation
is uncertain.

Precautions

Prescribing physicians who offer stimulants should consider potential AEs, such as psychosis, anorexia, anxiety, insomnia, mood changes (eg, anger),  misuse, addiction, mania, and cardiovascular problems. Psychostimulants have been implicated in precipitating psychosis.63,64 However, in a 12-month study of 250 adults with ADHD, 73 reported AEs, and only 31 discontinued the stimulant. Adverse effects leading to discontinuation included mood instability (n = 7), agitation (n = 6), irritability (n = 4), or decreased appetite (n = 4).65

Although associated with the risks of anorexia and insomnia in patients with ADHD, methylphenidate rapidly improved daytime sleepiness and mood, and—paradoxically—appetite and nighttime sleep in medically ill elderly patients with depression.66 Misuse or abuse of methylphenidate and dextroamphetamine were noted in 23% of patients referred for substance misuse.67 Nonetheless, little evidence exists that these drugs possess significant misuse potential in patients taking them as prescribed. As a prodrug, lisdexamfetamine is hypothesized to have less abuse potential compared with dextroamphetamine and methylphenidate, but it carries the same prescribing and monitoring precautions.68 Risks related to stimulant usage extend to manic symptoms.69 Patients with bipolar disorder should not receive stimulants if they have a history of stimulant-induced mania, rapid cycling, or psychosis.70

Long-term cardiovascular safety data exist for dextroamphetamine and methylphenidate but are limited or unavailable for modafinil, armodafinil, and atomoxetine. A retrospective cohort study found no significant increase in the number of cardiac events in patients receiving dextroamphetamine,
methylphenidate, or atomoxetine for an average of 1 year compared with controls.71 Another cohort study of > 44,000 patients found that initiation of
methylphenidate was associated with increased risk of sudden death or arrhythmia, but the risk was attributed to an unmeasured confounding factor, as the authors found a negative correlation between methylphenidate dose and all cardiovascular events.72

Recent practice guidelines recommend that before prescribing stimulants, clinicians should perform a physical examination (including heart and lung auscultation), obtain vital signs and height and weight, and request an electrocardiogram in case of abnormal findings on a cardiovascular examination or in case of a personal or family history of heart disease. Before offering atomoxetine, clinicians should evaluate the patient for a history of liver disease (and check liver function studies in case of a positive history). Clinicians should also assess risk of self-harm prior to initiating psychostimulant therapy.73 Throughout treatment, clinicians should evaluate the patient for changes in blood pressure, pulse, weight or mood, as well as the development of dependence or misuse. Urine toxicology testing is recommended for dextroamphetamine and methylphenidate to screen for adherence and diversion.

Limitations

Using only PubMed and MEDLINE databases limited the search to articles published in English after 1985, excluding letters and case reports to identify studies with higher evidence (the studies were not weighted based on study design). In addition, the studies had certain limitations. These include a limited number of DBPC trials, most were of short duration. It is also difficult to compare studies due to various rating scales used and concurrent
medication regimens of study subjects. These limitations raise questions surrounding the long-term efficacy of stimulants, and there is no consensus for how long a stimulant should be continued if beneficial. Longer, higherpowered, DBPC trials are warranted to determine longterm efficacy and safety of stimulant augmentation.62

Conclusion

For patients with depression who have not responded to other augmentation strategies, psychostimulants may be offered to improve mood, energy, and concentration. For clinicians considering stimulant augmentation, modafinil and armodafinil are reasonable choices given their efficacy in double-blind, placebo-controlled trials and lower risk of misuse. Dextroamphetamine (particularly lisdexamphetamine) and methylphenidate may be appropriate for patients who have not benefited from or tolerated modafinil or armodafinil, provided these patients do not have a medical history of cardiac disease or current substance use.

Osmotic controlled-release oral system methylphenidate seems to be ineffective as an augmenting agent. The efficacy of atomoxetine for augmentation is questionable, but atomoxetine could be offered if other stimulants were contraindicated, ineffective, or poorly tolerated. Both OROS methylphenidate and atomoxetine should be evaluated in additional trials before they can be recommended as augmentation therapies. Certain psychostimulants may be appropriate and reasonable adjunctive pharmacotherapies for patients with unipolar or bipolar depression who have failed other augmentation strategies, for patients who have significant fatigue or cognitive complaints, or for elderly patients with melancholic or somatic features of depression.

Acknowledgements
The authors thank Maureen Humphrey-Shelton and Kathy Thomas for their help in obtaining references.

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

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

Click here to read the digital edition.

Depression is a common condition that significantly impairs social and occupational functioning. Many patients do not respond to first-line pharmacotherapies and are considered to have treatment-resistant depression (TRD). These patients may benefit from augmentation of their antidepressant to reduce depression. Multiple medications have demonstrated various degrees of efficacy for augmentation, including psychostimulants. This article reviews studies of psychostimulants as augmentation agents for TRD and discusses risks, offers advice, and makes recommendations for clinicians who prescribe stimulants.

Background

Major depressive disorder (MDD) is a common psychiatric condition that significantly impairs quality of life.1 It is a recurrent illness, averaging 2 relapses per decade. The probability of recurrence increases with the number of depressive episodes.2,3 A patient who experiences major depressive episodes alternating with euthymia has unipolar depression; whereas one who experiences major depressive episodes alternating with episodes of mania or hypomania has bipolar depression.4

Despite adequate dose and duration of pharmacotherapy, many individuals with unipolar or bipolar depression do not achieve and sustain remission.5 Remission rates decrease and relapse rates increase with subsequent failed antidepressant trials.6 It is difficult to identify factors that predict treatment resistance, but one review of antidepressant studies found that patients who did not demonstrate a response within 3 weeks of medication initiation were less likely to respond after a longer duration.7

Treatment-resistant depression is commonly, but not universally, defined as lack of response after trials of 2 or more antidepressants with different mechanisms of action for sufficient duration.5 This definition will be used here as well. Other definitions have proposed stages of TRD, but these require further study to evaluate their reliability and predictive utility.8 Due to lack of consensus regarding the definition of TRD, it is not possible to determine the exact prevalence of TRD.

Patients with TRD may benefit from augmentation of their medication regimen. Augmentation with lithium has yielded conflicting results, and its efficacy with newer antidepressants is not well studied.9-12 Triiodothyronine, buspirone, and pindolol have demonstrated some efficacy when added to serotonin reuptake inhibitors (SRIs).10,12,13 Second-generation antipsychotic drugs, antidepressant drug combinations, omega-3 fatty acids, S-adenosyl methionine (SAMe), and L-methylfolate have demonstrated some efficacy in some studies as well.12,14-23 In patients with depression who have not responded to these strategies, psychostimulant augmentation may be appropriate.

Methods

A literature search was conducted following an algorithmic approach in the MEDLINE/PubMed database for studies in English from January 1985 to August 2014 of stimulants as augmenting agents for depression, using the Medical Subject Headings stimulant, depression, and augmentation, combined with an AND operator. The search was limited to adult humans and excluded case reports and letters, to identify studies with stronger evidence. Also excluded were studies using caffeine (to augment electroconvulsive therapy for depression) and pemoline as the sole augmenting stimulant as well as studies of patients with comorbid mental health diagnoses and studies that initiated stimulants and antidepressants simultaneously to assess antidepressant response.

This review organized results by stimulant rather than by depression type, even though some studies used > 1 stimulant or recruited patients with different types of depression. Although prevalence, prognosis, and monotherapy differ for unipolar and bipolar depression, psychostimulants target similar symptoms, despite augmenting different monotherapies in unipolar and bipolar depression. Therefore, no distinction is made between assessing studies of stimulants for unipolar and bipolar depression.

Results

A total of 70 articles were identified, and 31 studies met inclusion criteria (Figure). Of the studies included, 12 were double-blind, placebo-controlled (DBPC) trials and 19 were retrospective chart reviews or open studies. Most studies evaluated depression, using validated scales, such as the Hamilton Depression Rating Scale, Montgomery-Asberg Depression Rating Scale, Clinical Global Impressions of Severity, Inventory of Depressive Symptoms, Carroll Depression Rating Scale, Global Assessment of Functioning, Quick Inventory of Depressive Symptomatology, or the Psychiatric Symptom Assessment Scale. Study details are provided in Tables 1 to 4.

Dextroamphetamine and Methylphenidate

Dextroamphetamine and methylphenidate are indicated for the treatment of attention-deficit/hyperactivity disorder (ADHD) and exert their effects by inhibiting uptake of norepinephrine and dopamine.24 In one chart review, patients received dextroamphetamine or methylphenidate augmentation of monoamine oxidase inhibitors (MAOIs) alone or with concurrent tricyclic antidepressants; the majority reported decreased depression.25 In an openlabel trial, dextroamphetamine was titrated to efficacy in patients who were receiving an MAOI with or without pemoline.26 Nearly 80% of patients reported long-lasting improvement in depression. In an open-label trial, all patients reported decreased depression when methylphenidate was added to SRIs; however, no scales were used.27

In a case series, patients with both major depression and persistent depressive disorder (dysthymia) experienced a substantial, quick, and sustained response to dextroamphetamine or methylphenidate augmentation.28 Addition of lisdexamfetamine significantly reduced depressive symptoms in individuals with inadequate response to escitalopram.29 Patients with full or partial remission of depression noted improved executive function and residual depressive symptoms after lisdexamfetamine was added to SRI monotherapy.30 In a trial in which patients received dexamphetamine or methylphenidate as monotherapy or augmentation, 30% to 34% of patients reported mood improvement, but 36% reported no improvement.31 In an extension study, low-dose psychostimulants quickly diminished melancholia.32

Methylphenidate was safe and effective in patients with bipolar depression receiving treatment for 1 to 5 years; 44% evidenced significant improvement.33 When offered to patients with bipolar depression, patients receiving methylphenidate or dextroamphetamine reported less depression or sedation and did not develop tolerance, mania, or misuse.34 A case series concluded that methylphenidate addition to mood stabilizers was generally effective and safe.35

However, not all preparations of methylphenidate have demonstrated efficacy. In one study, osmotic controlledrelease oral system (OROS) methylphenidate improved apathy and fatigue but not overall depression.36 Although OROS methylphenidate similarly failed to demonstrate statistically significant efficacy in another study, more responders were documented in the treatment group.37

Although this review focuses on stimulants as augmenting agents in patients with depression, it is worth noting the limited number of studies evaluating stimulants’ effect on depression in patients with traumatic brain injury. This observation is of concern, as these conditions are frequently comorbid in returning veterans. One study noted that methylphenidate was an effective monotherapy for depression; whereas another study found that methylphenidate monotherapy reduced depression as well as sertraline, was better tolerated, and improved fatigue and cognition.38,39

Modafinil and Armodafinil

Modafinil and armodafinil (the R-enantiomer of modafinil) are indicated for improving wakefulness in individuals with narcolepsy, obstructive sleep apnea, and shift work sleep disorder by modulating glutamate, gamma amino-butyric acid, and histamine.40,41 Although they increase extracellular dopamine concentrations, they do not cause an increase in dopamine release and may have less misuse potential than that of dextroamphetamine and methylphenidate.40,41 In a study of 7 patients with unipolar or bipolar depression, all patients achieved full or partial remission with minimal adverse effects (AEs).42 In a prospective study, 41% of patients reported only mild depression or full remission with modafinil augmentation.43

Multiple trials and a pooled analysis noted decreased depression and fatigue and improved cognition in patients receiving modafinil augmentation compared with mood stabilizers or antidepressants.44-49 Modafinil is a useful adjunct for partial responders to SRIs, resulting in rapid mood improvement and decreased fatigue.50-54 However, in one study, modafinil did not demonstrate efficacy compared with placebo. This result was attributed to premature study termination after 2 modafinil-treated patients developed suicidal ideation.55 A post hoc analysis found no difference in frequency of suicidal ideation between groups.

Two DBPC studies evaluated armodafinil in patients with bipolar depression. In both studies it was added to a mood-stabilizing agent (lithium, valproate, aripiprazole, olanzapine, lamotrigine, risperidone, or ziprasidone), and patients receiving armodafinil reported significant reductions
in depression.56,57

Atomoxetine

Atomoxetine is a norepinephrine reuptake inhibitor indicated for the treatment of ADHD and is considered to have no misuse potential due to lack of dopamine modulation.58 In one study, 15 patients received atomoxetine added to their antidepressant, and 60% experienced significant symptom reduction.59 A chart review noted decreases in fatigue and depression when atomoxetine was added to an SRI, mirtazapine, or amitriptyline.60 However, in a DBPC trial, atomoxetine did not lead to significant changes in depression.61

Discussion

There is a limited amount of high-quality evidence to support psychostimulant augmentation, as noted by the relatively few DBPC trials, most of short duration. The evidence supports their efficacy primarily for unipolar depression, as 14 studies evaluated patients with unipolar depression, whereas only 7 studies evaluated patients with bipolar depression. The remaining studies recruited patients with both depression types. Collectively, modafinil and armodafinil have the most evidence in DBPC trials.

There are relatively few DBPC trials with high power and sufficient duration for dextroamphetamine and methylphenidate preparations. This discovery is surprising, considering the duration that these medications have been available. However, several chart reviews and open-label trials provided some evidence to support their use in patients without a history of substance misuse or cardiac conditions.62 Osmotic controlled- release oral system methylphenidate seems to be ineffective, and the efficacy of atomoxetine for augmentation
is uncertain.

Precautions

Prescribing physicians who offer stimulants should consider potential AEs, such as psychosis, anorexia, anxiety, insomnia, mood changes (eg, anger),  misuse, addiction, mania, and cardiovascular problems. Psychostimulants have been implicated in precipitating psychosis.63,64 However, in a 12-month study of 250 adults with ADHD, 73 reported AEs, and only 31 discontinued the stimulant. Adverse effects leading to discontinuation included mood instability (n = 7), agitation (n = 6), irritability (n = 4), or decreased appetite (n = 4).65

Although associated with the risks of anorexia and insomnia in patients with ADHD, methylphenidate rapidly improved daytime sleepiness and mood, and—paradoxically—appetite and nighttime sleep in medically ill elderly patients with depression.66 Misuse or abuse of methylphenidate and dextroamphetamine were noted in 23% of patients referred for substance misuse.67 Nonetheless, little evidence exists that these drugs possess significant misuse potential in patients taking them as prescribed. As a prodrug, lisdexamfetamine is hypothesized to have less abuse potential compared with dextroamphetamine and methylphenidate, but it carries the same prescribing and monitoring precautions.68 Risks related to stimulant usage extend to manic symptoms.69 Patients with bipolar disorder should not receive stimulants if they have a history of stimulant-induced mania, rapid cycling, or psychosis.70

Long-term cardiovascular safety data exist for dextroamphetamine and methylphenidate but are limited or unavailable for modafinil, armodafinil, and atomoxetine. A retrospective cohort study found no significant increase in the number of cardiac events in patients receiving dextroamphetamine,
methylphenidate, or atomoxetine for an average of 1 year compared with controls.71 Another cohort study of > 44,000 patients found that initiation of
methylphenidate was associated with increased risk of sudden death or arrhythmia, but the risk was attributed to an unmeasured confounding factor, as the authors found a negative correlation between methylphenidate dose and all cardiovascular events.72

Recent practice guidelines recommend that before prescribing stimulants, clinicians should perform a physical examination (including heart and lung auscultation), obtain vital signs and height and weight, and request an electrocardiogram in case of abnormal findings on a cardiovascular examination or in case of a personal or family history of heart disease. Before offering atomoxetine, clinicians should evaluate the patient for a history of liver disease (and check liver function studies in case of a positive history). Clinicians should also assess risk of self-harm prior to initiating psychostimulant therapy.73 Throughout treatment, clinicians should evaluate the patient for changes in blood pressure, pulse, weight or mood, as well as the development of dependence or misuse. Urine toxicology testing is recommended for dextroamphetamine and methylphenidate to screen for adherence and diversion.

Limitations

Using only PubMed and MEDLINE databases limited the search to articles published in English after 1985, excluding letters and case reports to identify studies with higher evidence (the studies were not weighted based on study design). In addition, the studies had certain limitations. These include a limited number of DBPC trials, most were of short duration. It is also difficult to compare studies due to various rating scales used and concurrent
medication regimens of study subjects. These limitations raise questions surrounding the long-term efficacy of stimulants, and there is no consensus for how long a stimulant should be continued if beneficial. Longer, higherpowered, DBPC trials are warranted to determine longterm efficacy and safety of stimulant augmentation.62

Conclusion

For patients with depression who have not responded to other augmentation strategies, psychostimulants may be offered to improve mood, energy, and concentration. For clinicians considering stimulant augmentation, modafinil and armodafinil are reasonable choices given their efficacy in double-blind, placebo-controlled trials and lower risk of misuse. Dextroamphetamine (particularly lisdexamphetamine) and methylphenidate may be appropriate for patients who have not benefited from or tolerated modafinil or armodafinil, provided these patients do not have a medical history of cardiac disease or current substance use.

Osmotic controlled-release oral system methylphenidate seems to be ineffective as an augmenting agent. The efficacy of atomoxetine for augmentation is questionable, but atomoxetine could be offered if other stimulants were contraindicated, ineffective, or poorly tolerated. Both OROS methylphenidate and atomoxetine should be evaluated in additional trials before they can be recommended as augmentation therapies. Certain psychostimulants may be appropriate and reasonable adjunctive pharmacotherapies for patients with unipolar or bipolar depression who have failed other augmentation strategies, for patients who have significant fatigue or cognitive complaints, or for elderly patients with melancholic or somatic features of depression.

Acknowledgements
The authors thank Maureen Humphrey-Shelton and Kathy Thomas for their help in obtaining references.

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

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

Click here to read the digital edition.

References

1. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distribution of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):593-602.

2. Solomon DA, Keller MB, Leon AC, et al. Multiple recurrences of major depressive disorder. Am J Psychiatry. 2000;157(2):229-233.

3. Katon WJ, Fan MY, Lin EH, Unützer J. Depressive symptom deterioration in a large
primary care-based elderly cohort. Am J Geriatr Psychiatry. 2006;14(3):246-254.

4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Arlington, VA: American Psychiatric Association; 2013.

5. McIntyre RS, Filteau M-J, Martin L, et al. Treatment-resistant depression: Definitions, review of the evidence, and algorithmic approach. J Affect Disord. 2014;156:1-7.

6. Gaynes BN, Warden D, Trivedi MH, Wisniewski SR, Fava M, Rush AJ. What did STAR*D teach us? Results from a large-scale, practical, clinical trial for patients with depression. Focus. 2012;10(4):510-517.

7. Kudlow PA, Cha DS, McIntyre RS. Predicting treatment response in major depressive disorder: The impact of early symptomatic improvement. Can J Psychiatry. 2012;57(12):782-788.

8. Ruhé HG, van Rooijen G, Spijker J, Peeters FP, Schene AH. Staging methods for treatment resistant depression. A systematic review. J Affect Disord. 2012;137(1-3):35-45.

9. Bauer M, Dopfmer S. Lithium augmentation treatment-resistant depression: Metaanalysis of placebo-controlled studies. J Clin Psychopharmacol. 1999;19(5):427-434.

10. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: A STAR*D report. Am J Psychiatry. 2006;163(9):1519-1530.

11. Nierenberg AA, Papakostas GI, Petersen T, et al. Lithium augmentation of nortriptyline
for subjects resistant to multiple antidepressants. J Clin Psychopharmacol. 2003;23(1):92-95.

12. Connolly KR, Thase ME. If at first you don’t succeed: A review of the evidence for antidepressant augmentation, combination, and switching strategies. Drugs. 2011;71(1):43-64.

13. Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team. Medication augmentation after the failure of SSRIs for depression. N Engl J Med. 2006;354(12):1243-1252.

14. Papakostas GI, Shelton RC, Smith J, Fava M. Augmentation of antidepressants with atypical antipsychotic medications for treatment resistant major depressive disorder: A meta-analysis. J Clin Psychiatry. 2007;68(6):826-831.

15. Mahmoud RA, Pandina GJ, Turkoz I, et al. Risperidone for treatment-refractory major depressive disorder: A randomized trial. Ann Intern Med. 2007;147(9):593-602.

16. Barbee JG, Conrad EJ, Jamhour NJ. The effectiveness of olanzapine, risperidone, quetiapine, and ziprasidone as augmentation agents in treatment resistant depressive disorder. J Clin Psychiatry. 2004;65(7):975-981.

17. Fatemi SH, Emamian ES, Kist DA. Venlafaxine and bupropion combination therapy in a case of treatment-resistant depression. Ann Pharmacother.1999;33(6):701-703.

18. Carpenter LL, Yasman S, Price LH. A double-blind, placebo-controlled study of antidepressant augmentation with mirtazapine. Biol Psychiatry. 2002;51(2):183-188.

19. Hannan N, Hamzah Z, Akinpeloye HO, Meagher D. Venlafaxine-mirtazapine combination therapy in the treatment of persistent depressive illness. J Psychopharmacol. 2007;21(2):161-164.

20. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: A STAR*D report. Am J Psychiatry. 2006;163(9):1531-1541.

21. Blier P, Ward HE, Tremblay P, Laberge L, Hébert C, Bergeron R. Combination of antidepressant medications from treatment initiation for major depressive disorder: A double-blind randomized study. Am J Psychiatry. 2010;167(3):281-288.

22. Papakostas GI, Mischoulon D, Shyu I, Alpert JE, Fava M. S-adenosyl methionine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: A double blind randomized clinical trial. Am J Psychiatry. 2010;167(8):942-948.

23. Papakostas GI, Shelton RC, Zajecka JM, et al. L-methylfolate as adjunctive therapy
for SSRI-resistant major depression: Results of two randomized, double-blind,
parallel-sequential trials. Am J Psychiatry. 2012;169(12):1267-1274.

24. Korston TR. Drugs of abuse. In: Katzung BG, ed. Basic and Clinical Pharmacology. 9th ed. New York, NY: McGraw-Hill; 2004:521-523.

25. Feighner JP, Herbstein J, Damlouji N. Combined MAOI, TCA, and direct stimulant therapy of treatment-resistant depression. J Clin Psychiatry. 1985;46(6):206-209.

26. Fawcett J, Kravitz HM, Zajecka JM, Schaff MR. CNS stimulant potentiation of monoamine oxidase inhibitors in treatment-refractory depression. J Clin Psychopharmacol. 1991;11(2):127-132.

27. Stoll AL, Pillay SS, Diamond L, Workum SB, Cole JO. Methylphenidate augmentation of serotonin selective reuptake inhibitors: A case series. J Clin Psychiatry. 1996;57(2):72-76.

28. Masand PS, Anand VS, Tanquary JF. Psychostimulant augmentation of second generation antidepressants: A case series. Depress Anxiety. 1998;7(2):89-91.

29. Trivedi MH, Cutler AJ, Richards C, et al. A randomized control trial of the efficacy and safety of lisdesxamfetamine dimesylate as augmentation therapy in adults with residual symptoms of major depressive disorder after treatment with escitalopram. J Clin Psychiatry. 2013;74(8):802-809.

30. Madhoo M, Keefe RS, Roth RM, et al. Lisdexamfetamine dimesylate augmentation in adults with persistent executive dysfunction after partial or full remission of major depressive disorder. Neuropsychopharmacology. 2014;39(6):1388-1398.

31. Parker G, Brotchie H. Do the old psychostimulant drugs have a role in managing treatment-resistant depression. Acta Psychiatr Scand. 2010;121(4):308-314.

32. Parker G, Brotchie H, McClure G, Fletcher K. Psychostimulants for managing unipolar and bipolar treatment-resistant melancholic depression: A medium term evaluation of cost benefits. J Affect Disord. 2013;151(1):360-364.

33. Lydon E, El-Mallakh RS. Naturalistic long-term use of methylphenidate in bipolar disorder. J Clin Psychopharmacol. 2006;26(5):516-518.

34. Carlson PJ, Merlock MC, Suppes T. Adjunctive stimulant use in patients with bipolar disorder: Treatment of residual depression and sedation. Bipolar Disord. 2004;6(5):416-420.

35. El-Mallakh RS. An open study of methylphenidate in bipolar depression. Bipolar Disord. 2000;2(1):56-59.

36. Ravindran AV, Kennedy SH, O’Donovan MC, Fallu A, Camacho F, Binder CE. Osmotic-release oral system methylphenidate augmentation of antidepressant monotherapy in major depressive disorder: Results of a double-blind, randomized, placebo-controlled trial. J Clin Psychiatry. 2008;69(1):87-94.

37. Patkar AA, Masand PS, Pae CU, et al. A randomized, double-blind, placebocontrolled
trial of augmentation with an extended release formulation of methylphenidate in outpatients with treatment-resistant depression. J Clin Psychopharmacol. 2006;26(6):653-656.

38. Lee H, Kim SW, Kim JM, Shin IS, Yang SJ, Yoon JS Comparing effects of methylphenidate, sertraline, and placebo on neuropsychiatric sequelae in patients with
traumatic brain injury. Hum Psychopharmacol. 2005;20(2):97-104.

39. Gualtieri CT, Evans RW. Stimulant treatment for the neurobehavioural sequelae of traumatic brain injury. Brain Inj. 1988;2(4):273-290.

40. Provigil [package insert]. North Wales, PA: Cephalon Inc; 2015.

41. Nuvigil [package insert]. Frazer, PA: Cephalon, Inc; 2013.

42. Menza MA, Kaufman KR, Castellanos A. Modafinil augmentation of antidepressant treatment in depression. J Clin Psychiatry. 2000;61(5):378-381.

43. Markovitz PJ, Wagner S. An open-label trial of modafinil augmentation in patients with partial response to antidepressant therapy. J Clin Psychopharmacol. 2003;23(2):207-209.

44. Fernandes PP, Petty F. Modafinil for remitted bipolar depression with hypersomnia. Ann Pharmacother. 2003;37(12):1807-1809.

45. Nasr S. Modafinil as adjunctive therapy in depressed outpatients. Ann Clin Psychiatry. 2004;16(3):133-138.

46. DeBattista C, Lembke A, Solvason HB, Ghebremichael R, Poirier J. A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol. 2004;24(1):87-90.

47. Nasr S, Wendt B, Steiner K. Absence of mood switch with and tolerance to modafinil: A replication study from a large private practice. J Affect Disord. 2006;95(1-3):111-114.

48. DeBattista C, Doghramji K, Menza MA, Rosenthal MH, Fieve RR; Modafinil in Depression Study Group. Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: A preliminary doubleblind, placebo-controlled study. J Clin Psychiatry. 2003;64(9):1057-1064.

49. Frye MA, Grunze H, Suppes T, et al. A placebo-controlled evaluation of adjunctive modafinil in the treatment of bipolar depression. Am J Psychiatry. 2007;164(8):1242-1249.

50. Fava M, Thase ME, DeBattista C, Doghramji K, Arora S, Hughes RJ. Modafinil augmentation of selective serotonin reuptake inhibitor therapy in MDD partial responders with persistent fatigue and sleepiness. Ann Clin Psychiatry. 2007;19(3):153-159.

51. Thase ME, Fava M, DeBattista C, Arora S, Hughes RJ. Modafinil augmentation of SSRI therapy in patients with major depressive disorder and excessive sleepiness and fatigue: A 12-week, open-label, extension study. CNS Spectr. 2006;11(2):93-102.

52. Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry. 2005;66(1):85-93.

53. Abolfazli R, Hosseini M, Ghanizadeh A, et al. Double-blind randomized parallelgroup clinical trial of efficacy of the combination fluoxetine plus modafinil versus fluoxetine plus placebo in the treatment of major depression. Depress Anxiety. 2011;28(4):297-302.

54. Rasmussen NA, Schrøder P, Olsen LR, Brødsgaard M, Undén M, Bech P. Modafinil augmentation in depressed patients with partial response to antidepressants: A pilot study on self-reported symptoms covered by the Major Depression Inventory (MDI) and the Symptom Checklist (SCL-92). Nord J Psychiatry. 2005;59(3):173-178.

55. Dunlop BW, Crits-Christoph P, Evans DL, et al. Coadministration of modafinil and a selective serotonin reuptake inhibitor from the initiation of treatment of major depressive disorder with fatigue and sleepiness: A double-blind, placebocontrolled study. J Clin Psychopharmacol. 2007;27(6):614-619.

56. Calabrese JR, Ketter TA, Youakim JM, Tiller JM, Yang R, Frye MA. Adjunctive armodafinil
for major depressive episodes associated with bipolar I disorder: A randomized multicenter, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2010;71(10):1363-1370.

57. Calabrese JR, Frye MA, Yang R, Ketter TA; Armodafinil Treatment Trial Study Network. Efficacy and safety of adjunctive armodafinil in adults with major depressive episodes associated with bipolar I disorder: A randomized, double-blind, placebo-controlled, multicenter trial. J Clin Psychiatry. 2014;75(10):1054-1061.

58. Strattera [package insert]. Indianapolis, IN. Lilly; 2015.

59. Carpenter LL, Milosavljevic N, Schecter JM, Tyrka AR, Price LH. Augmentation with open-label atomoxetine for partial or nonresponse to antidepressants. J Clin Psychiatry. 2005;66(10):1234-1238.

60. Papakostas GI, Petersen TJ, Burns AM, Fava M. Adjunctive atomoxetine for residual
fatigue in major depressive disorder. J Psychiatr Res. 2006;40(4):370-373.

61. Michelson D, Adler LA, Amsterdam JD, et al. Addition of atomoxetine for depression
incompletely responsive to sertraline: A randomized, double-blind, placebocontrolled study. J Clin Psychiatry. 2007;68(4):582-587.

62. Corp SA, Gitlin MJ, Altshuler LL. A review of the use of stimulants and stimulant alternatives in treating bipolar depression and major depressive disorder. J Clin Psychiatry. 2014;75(9):1010-1018.

63. Kraemer M, Uekermann J, Wiltfang J, Kis B. Methylphenidate-induced psychosis in adult attention-deficit/hyperactivity disorder: Report of 3 new cases and review of the literature. Clin Neuropharmacol. 2010;33(4):204-206.

64. Berman SM, Kuczenski R, McCracken JT, London ED. Potential adverse effects of amphetamine treatment on brain and behavior: A review. Mol Psychiatry. 2009;14(2):123-142.

65. Fredriksen M, Dahl AA, Martinsen EW, Klungsøyr O, Haavik J, Peleikis DE. Effectiveness of one-year pharmacological treatment of adult attention-deficit/hyperactivity disorder (ADHD): An open-label prospective study of time in treatment, dose, side-effects and comorbidity. Eur Neuropsychopharmacol. 2014;24(12):1873-1874.

66. Hardy SE. Methylphenidate for the treatment of depressive symptoms, including fatigue and apathy, in medically ill older adults and terminally ill adults. Am J Geriatr Pharmacother. 2009;7(1):34-59.

67. Williams RJ, Goodale LA, Shay-Fiddler MA, Gloster SP, Chang SY. Methylphenidate and dextroamphetamine abuse in substance-abusing adolescents. Am J Addict. 2004;13(4):381-389.

68. Madaan V, Kolli V, Bestha DP, Shah MJ. Update on optimal use of lisdexamfetamine in the treatment of ADHD. Neuropsychiatr Dis Treat. 2013;9:977-983.

69. Ross RG. Psychotic and manic-like symptoms during stimulant treatment of attention deficit hyperactivity disorder. Am J Psychiatry. 2006;163(7):1149-1152.

70. Dell’Osso B, Ketter TA. Use of adjunctive stimulants in adult bipolar depression. Int J Neuropsychopharmacol. 2013;16(1):55-68.

71. Habel LA, Cooper WO, Sox CM, et al. ADHD medications and risk of serious cardiovascular events in young and middle-aged adults. JAMA. 2011;306(24):2673-2683.

72. Schelleman H, Bilker WB, Kimmel SE, et al. Methylphenidate and risk of serious cardiovascular events in adults. Am J Psychiatry. 2012;169(2):178-185.

73. Bolea-Alamañac B, Nutt DJ, Adamou M, et al; British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological management of attention deficit hyperactivity disorder: Update on recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(3):179-203.

74. Moher D, Liberati A, Tetzlaff J, Altman DG; The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009;6(6):e1000097.

References

1. Kessler RC, Berglund P, Demler O, Jin R, Merikangas KR, Walters EE. Lifetime prevalence and age-of-onset distribution of DSM-IV disorders in the National Comorbidity Survey Replication. Arch Gen Psychiatry. 2005;62(6):593-602.

2. Solomon DA, Keller MB, Leon AC, et al. Multiple recurrences of major depressive disorder. Am J Psychiatry. 2000;157(2):229-233.

3. Katon WJ, Fan MY, Lin EH, Unützer J. Depressive symptom deterioration in a large
primary care-based elderly cohort. Am J Geriatr Psychiatry. 2006;14(3):246-254.

4. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Arlington, VA: American Psychiatric Association; 2013.

5. McIntyre RS, Filteau M-J, Martin L, et al. Treatment-resistant depression: Definitions, review of the evidence, and algorithmic approach. J Affect Disord. 2014;156:1-7.

6. Gaynes BN, Warden D, Trivedi MH, Wisniewski SR, Fava M, Rush AJ. What did STAR*D teach us? Results from a large-scale, practical, clinical trial for patients with depression. Focus. 2012;10(4):510-517.

7. Kudlow PA, Cha DS, McIntyre RS. Predicting treatment response in major depressive disorder: The impact of early symptomatic improvement. Can J Psychiatry. 2012;57(12):782-788.

8. Ruhé HG, van Rooijen G, Spijker J, Peeters FP, Schene AH. Staging methods for treatment resistant depression. A systematic review. J Affect Disord. 2012;137(1-3):35-45.

9. Bauer M, Dopfmer S. Lithium augmentation treatment-resistant depression: Metaanalysis of placebo-controlled studies. J Clin Psychopharmacol. 1999;19(5):427-434.

10. Nierenberg AA, Fava M, Trivedi MH, et al. A comparison of lithium and T(3) augmentation following two failed medication treatments for depression: A STAR*D report. Am J Psychiatry. 2006;163(9):1519-1530.

11. Nierenberg AA, Papakostas GI, Petersen T, et al. Lithium augmentation of nortriptyline
for subjects resistant to multiple antidepressants. J Clin Psychopharmacol. 2003;23(1):92-95.

12. Connolly KR, Thase ME. If at first you don’t succeed: A review of the evidence for antidepressant augmentation, combination, and switching strategies. Drugs. 2011;71(1):43-64.

13. Trivedi MH, Fava M, Wisniewski SR, et al; STAR*D Study Team. Medication augmentation after the failure of SSRIs for depression. N Engl J Med. 2006;354(12):1243-1252.

14. Papakostas GI, Shelton RC, Smith J, Fava M. Augmentation of antidepressants with atypical antipsychotic medications for treatment resistant major depressive disorder: A meta-analysis. J Clin Psychiatry. 2007;68(6):826-831.

15. Mahmoud RA, Pandina GJ, Turkoz I, et al. Risperidone for treatment-refractory major depressive disorder: A randomized trial. Ann Intern Med. 2007;147(9):593-602.

16. Barbee JG, Conrad EJ, Jamhour NJ. The effectiveness of olanzapine, risperidone, quetiapine, and ziprasidone as augmentation agents in treatment resistant depressive disorder. J Clin Psychiatry. 2004;65(7):975-981.

17. Fatemi SH, Emamian ES, Kist DA. Venlafaxine and bupropion combination therapy in a case of treatment-resistant depression. Ann Pharmacother.1999;33(6):701-703.

18. Carpenter LL, Yasman S, Price LH. A double-blind, placebo-controlled study of antidepressant augmentation with mirtazapine. Biol Psychiatry. 2002;51(2):183-188.

19. Hannan N, Hamzah Z, Akinpeloye HO, Meagher D. Venlafaxine-mirtazapine combination therapy in the treatment of persistent depressive illness. J Psychopharmacol. 2007;21(2):161-164.

20. McGrath PJ, Stewart JW, Fava M, et al. Tranylcypromine versus venlafaxine plus mirtazapine following three failed antidepressant medication trials for depression: A STAR*D report. Am J Psychiatry. 2006;163(9):1531-1541.

21. Blier P, Ward HE, Tremblay P, Laberge L, Hébert C, Bergeron R. Combination of antidepressant medications from treatment initiation for major depressive disorder: A double-blind randomized study. Am J Psychiatry. 2010;167(3):281-288.

22. Papakostas GI, Mischoulon D, Shyu I, Alpert JE, Fava M. S-adenosyl methionine (SAMe) augmentation of serotonin reuptake inhibitors for antidepressant nonresponders with major depressive disorder: A double blind randomized clinical trial. Am J Psychiatry. 2010;167(8):942-948.

23. Papakostas GI, Shelton RC, Zajecka JM, et al. L-methylfolate as adjunctive therapy
for SSRI-resistant major depression: Results of two randomized, double-blind,
parallel-sequential trials. Am J Psychiatry. 2012;169(12):1267-1274.

24. Korston TR. Drugs of abuse. In: Katzung BG, ed. Basic and Clinical Pharmacology. 9th ed. New York, NY: McGraw-Hill; 2004:521-523.

25. Feighner JP, Herbstein J, Damlouji N. Combined MAOI, TCA, and direct stimulant therapy of treatment-resistant depression. J Clin Psychiatry. 1985;46(6):206-209.

26. Fawcett J, Kravitz HM, Zajecka JM, Schaff MR. CNS stimulant potentiation of monoamine oxidase inhibitors in treatment-refractory depression. J Clin Psychopharmacol. 1991;11(2):127-132.

27. Stoll AL, Pillay SS, Diamond L, Workum SB, Cole JO. Methylphenidate augmentation of serotonin selective reuptake inhibitors: A case series. J Clin Psychiatry. 1996;57(2):72-76.

28. Masand PS, Anand VS, Tanquary JF. Psychostimulant augmentation of second generation antidepressants: A case series. Depress Anxiety. 1998;7(2):89-91.

29. Trivedi MH, Cutler AJ, Richards C, et al. A randomized control trial of the efficacy and safety of lisdesxamfetamine dimesylate as augmentation therapy in adults with residual symptoms of major depressive disorder after treatment with escitalopram. J Clin Psychiatry. 2013;74(8):802-809.

30. Madhoo M, Keefe RS, Roth RM, et al. Lisdexamfetamine dimesylate augmentation in adults with persistent executive dysfunction after partial or full remission of major depressive disorder. Neuropsychopharmacology. 2014;39(6):1388-1398.

31. Parker G, Brotchie H. Do the old psychostimulant drugs have a role in managing treatment-resistant depression. Acta Psychiatr Scand. 2010;121(4):308-314.

32. Parker G, Brotchie H, McClure G, Fletcher K. Psychostimulants for managing unipolar and bipolar treatment-resistant melancholic depression: A medium term evaluation of cost benefits. J Affect Disord. 2013;151(1):360-364.

33. Lydon E, El-Mallakh RS. Naturalistic long-term use of methylphenidate in bipolar disorder. J Clin Psychopharmacol. 2006;26(5):516-518.

34. Carlson PJ, Merlock MC, Suppes T. Adjunctive stimulant use in patients with bipolar disorder: Treatment of residual depression and sedation. Bipolar Disord. 2004;6(5):416-420.

35. El-Mallakh RS. An open study of methylphenidate in bipolar depression. Bipolar Disord. 2000;2(1):56-59.

36. Ravindran AV, Kennedy SH, O’Donovan MC, Fallu A, Camacho F, Binder CE. Osmotic-release oral system methylphenidate augmentation of antidepressant monotherapy in major depressive disorder: Results of a double-blind, randomized, placebo-controlled trial. J Clin Psychiatry. 2008;69(1):87-94.

37. Patkar AA, Masand PS, Pae CU, et al. A randomized, double-blind, placebocontrolled
trial of augmentation with an extended release formulation of methylphenidate in outpatients with treatment-resistant depression. J Clin Psychopharmacol. 2006;26(6):653-656.

38. Lee H, Kim SW, Kim JM, Shin IS, Yang SJ, Yoon JS Comparing effects of methylphenidate, sertraline, and placebo on neuropsychiatric sequelae in patients with
traumatic brain injury. Hum Psychopharmacol. 2005;20(2):97-104.

39. Gualtieri CT, Evans RW. Stimulant treatment for the neurobehavioural sequelae of traumatic brain injury. Brain Inj. 1988;2(4):273-290.

40. Provigil [package insert]. North Wales, PA: Cephalon Inc; 2015.

41. Nuvigil [package insert]. Frazer, PA: Cephalon, Inc; 2013.

42. Menza MA, Kaufman KR, Castellanos A. Modafinil augmentation of antidepressant treatment in depression. J Clin Psychiatry. 2000;61(5):378-381.

43. Markovitz PJ, Wagner S. An open-label trial of modafinil augmentation in patients with partial response to antidepressant therapy. J Clin Psychopharmacol. 2003;23(2):207-209.

44. Fernandes PP, Petty F. Modafinil for remitted bipolar depression with hypersomnia. Ann Pharmacother. 2003;37(12):1807-1809.

45. Nasr S. Modafinil as adjunctive therapy in depressed outpatients. Ann Clin Psychiatry. 2004;16(3):133-138.

46. DeBattista C, Lembke A, Solvason HB, Ghebremichael R, Poirier J. A prospective trial of modafinil as an adjunctive treatment of major depression. J Clin Psychopharmacol. 2004;24(1):87-90.

47. Nasr S, Wendt B, Steiner K. Absence of mood switch with and tolerance to modafinil: A replication study from a large private practice. J Affect Disord. 2006;95(1-3):111-114.

48. DeBattista C, Doghramji K, Menza MA, Rosenthal MH, Fieve RR; Modafinil in Depression Study Group. Adjunct modafinil for the short-term treatment of fatigue and sleepiness in patients with major depressive disorder: A preliminary doubleblind, placebo-controlled study. J Clin Psychiatry. 2003;64(9):1057-1064.

49. Frye MA, Grunze H, Suppes T, et al. A placebo-controlled evaluation of adjunctive modafinil in the treatment of bipolar depression. Am J Psychiatry. 2007;164(8):1242-1249.

50. Fava M, Thase ME, DeBattista C, Doghramji K, Arora S, Hughes RJ. Modafinil augmentation of selective serotonin reuptake inhibitor therapy in MDD partial responders with persistent fatigue and sleepiness. Ann Clin Psychiatry. 2007;19(3):153-159.

51. Thase ME, Fava M, DeBattista C, Arora S, Hughes RJ. Modafinil augmentation of SSRI therapy in patients with major depressive disorder and excessive sleepiness and fatigue: A 12-week, open-label, extension study. CNS Spectr. 2006;11(2):93-102.

52. Fava M, Thase ME, DeBattista C. A multicenter, placebo-controlled study of modafinil augmentation in partial responders to selective serotonin reuptake inhibitors with persistent fatigue and sleepiness. J Clin Psychiatry. 2005;66(1):85-93.

53. Abolfazli R, Hosseini M, Ghanizadeh A, et al. Double-blind randomized parallelgroup clinical trial of efficacy of the combination fluoxetine plus modafinil versus fluoxetine plus placebo in the treatment of major depression. Depress Anxiety. 2011;28(4):297-302.

54. Rasmussen NA, Schrøder P, Olsen LR, Brødsgaard M, Undén M, Bech P. Modafinil augmentation in depressed patients with partial response to antidepressants: A pilot study on self-reported symptoms covered by the Major Depression Inventory (MDI) and the Symptom Checklist (SCL-92). Nord J Psychiatry. 2005;59(3):173-178.

55. Dunlop BW, Crits-Christoph P, Evans DL, et al. Coadministration of modafinil and a selective serotonin reuptake inhibitor from the initiation of treatment of major depressive disorder with fatigue and sleepiness: A double-blind, placebocontrolled study. J Clin Psychopharmacol. 2007;27(6):614-619.

56. Calabrese JR, Ketter TA, Youakim JM, Tiller JM, Yang R, Frye MA. Adjunctive armodafinil
for major depressive episodes associated with bipolar I disorder: A randomized multicenter, double-blind, placebo-controlled, proof-of-concept study. J Clin Psychiatry. 2010;71(10):1363-1370.

57. Calabrese JR, Frye MA, Yang R, Ketter TA; Armodafinil Treatment Trial Study Network. Efficacy and safety of adjunctive armodafinil in adults with major depressive episodes associated with bipolar I disorder: A randomized, double-blind, placebo-controlled, multicenter trial. J Clin Psychiatry. 2014;75(10):1054-1061.

58. Strattera [package insert]. Indianapolis, IN. Lilly; 2015.

59. Carpenter LL, Milosavljevic N, Schecter JM, Tyrka AR, Price LH. Augmentation with open-label atomoxetine for partial or nonresponse to antidepressants. J Clin Psychiatry. 2005;66(10):1234-1238.

60. Papakostas GI, Petersen TJ, Burns AM, Fava M. Adjunctive atomoxetine for residual
fatigue in major depressive disorder. J Psychiatr Res. 2006;40(4):370-373.

61. Michelson D, Adler LA, Amsterdam JD, et al. Addition of atomoxetine for depression
incompletely responsive to sertraline: A randomized, double-blind, placebocontrolled study. J Clin Psychiatry. 2007;68(4):582-587.

62. Corp SA, Gitlin MJ, Altshuler LL. A review of the use of stimulants and stimulant alternatives in treating bipolar depression and major depressive disorder. J Clin Psychiatry. 2014;75(9):1010-1018.

63. Kraemer M, Uekermann J, Wiltfang J, Kis B. Methylphenidate-induced psychosis in adult attention-deficit/hyperactivity disorder: Report of 3 new cases and review of the literature. Clin Neuropharmacol. 2010;33(4):204-206.

64. Berman SM, Kuczenski R, McCracken JT, London ED. Potential adverse effects of amphetamine treatment on brain and behavior: A review. Mol Psychiatry. 2009;14(2):123-142.

65. Fredriksen M, Dahl AA, Martinsen EW, Klungsøyr O, Haavik J, Peleikis DE. Effectiveness of one-year pharmacological treatment of adult attention-deficit/hyperactivity disorder (ADHD): An open-label prospective study of time in treatment, dose, side-effects and comorbidity. Eur Neuropsychopharmacol. 2014;24(12):1873-1874.

66. Hardy SE. Methylphenidate for the treatment of depressive symptoms, including fatigue and apathy, in medically ill older adults and terminally ill adults. Am J Geriatr Pharmacother. 2009;7(1):34-59.

67. Williams RJ, Goodale LA, Shay-Fiddler MA, Gloster SP, Chang SY. Methylphenidate and dextroamphetamine abuse in substance-abusing adolescents. Am J Addict. 2004;13(4):381-389.

68. Madaan V, Kolli V, Bestha DP, Shah MJ. Update on optimal use of lisdexamfetamine in the treatment of ADHD. Neuropsychiatr Dis Treat. 2013;9:977-983.

69. Ross RG. Psychotic and manic-like symptoms during stimulant treatment of attention deficit hyperactivity disorder. Am J Psychiatry. 2006;163(7):1149-1152.

70. Dell’Osso B, Ketter TA. Use of adjunctive stimulants in adult bipolar depression. Int J Neuropsychopharmacol. 2013;16(1):55-68.

71. Habel LA, Cooper WO, Sox CM, et al. ADHD medications and risk of serious cardiovascular events in young and middle-aged adults. JAMA. 2011;306(24):2673-2683.

72. Schelleman H, Bilker WB, Kimmel SE, et al. Methylphenidate and risk of serious cardiovascular events in adults. Am J Psychiatry. 2012;169(2):178-185.

73. Bolea-Alamañac B, Nutt DJ, Adamou M, et al; British Association for Psychopharmacology. Evidence-based guidelines for the pharmacological management of attention deficit hyperactivity disorder: Update on recommendations from the British Association for Psychopharmacology. J Psychopharmacol. 2014;28(3):179-203.

74. Moher D, Liberati A, Tetzlaff J, Altman DG; The PRISMA Group. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. PLoS Med. 2009;6(6):e1000097.

Issue
Federal Practitioner - 32(3)s
Issue
Federal Practitioner - 32(3)s
Publications
Publications
Topics
Article Type
Sections
Citation Override
Fed Pract. 2015 April;32(suppl 3):30S-37S
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME

Multidisciplinary Management of a Patient With Multiple Sclerosis:Part 3. Psychologists’ Perspective

Article Type
Changed
Fri, 11/10/2017 - 11:02

Multiple sclerosis (MS) poses a host of cognitive and psychosocial challenges that may contribute to functioning and quality of life (QOL). Although each patient’s experience with MS is different, some challenges are more common than are others, including cognitive changes, depression, and maintaining positive health behaviors. William’s case study illustrates some of these challenges as well as the resources and strategies of the psychologists at the MS
Centers of Excellence (MSCoE) to help patients with MS adapt and thrive.

Cognition

Difficulties with cognitive functioning are common in patients with MS. About half of patients with MS will develop cognitive impairments in one or more areas during their lifetime.1 Although cognitive difficulties tend to worsen over the course of the disease, they can appear at any point in the illness, differ greatly from patient to patient, and are only modestly correlated with physical symptoms.1 Impairments are most common in the areas of information processing speed and memory, as well as in complex attention and mental flexibility. These impairments can impact activities of daily living, sustained employment, driving, and social relationships.

Early in the disease, William, who admitted to difficulties with cognition and the impact of cognitive impairment on his life, benefited from neuropsychological testing. Typically, an MSCoE will use a battery of tests tailored to patients with MS. For patients, the results of these tests can be used to clarify areas of relative strength and weakness and inform team decisions related to treatment and future life activities, such as whether the patient will need accommodations at work. Test results may also be used to guide how the MS treatment team interacts with the patient.

An initial clinic screening from neurology indicated William had below average cognitive processing speed, and he agreed to neurocognitive testing. The psycholopsychologist asked William about his functioning at home. William noted poor attention and memory made it difficult to advocate for himself. He recounted an episode of taking his car to be repaired and only later realizing that he had been charged twice for the same part. He also disclosed that he had gotten lost while driving through familiar places. On several occasions while cooking, he had become distracted and started another task only to find he had burned his food.

Cognitive rehabilitation has shown considerable promise in helping patients work through difficulties with memory, attention, and problem solving by developing compensatory strategies.2 Skill training is available in individual and group formats. There is also promising but very preliminary evidence that some psychosocial interventions might improve memory performance for patients with MS.3

William’s neuropsychological testing confirmed impairments in information processing speed, attention, and memory, and William was diagnosed with cognitive disorder not otherwise specified. His cognitive impairment correlated with his magnetic resonance imaging (MRI) findings, which included lesions on the corpus callosum and in multiple subcortical areas.

During a feedback session, William was encouraged to use compensatory strategies, including memory aids, visual cueing, and self-pacing. He was given a referral to speech and language pathology to develop and practice these strategies. The psychologist also reminded William’s health care team to speak slowly and repeat important information to him, write down important instructions, and cue him when asking questions in the clinic. William was provided with a kitchen timer and instructed to set it whenever he began cooking, so that even if he got distracted, the alarm would remind him to return to his cooking. William was asked whether he would like to participate in the MSCoE cognitive compensatory training program.4 The program, offered through a research protocol, involved a series of classes that taught strategies to manage cognitive symptoms and improve patients’ ability to function independently. William agreed to participate and reported feeling hopeful that his situation could improve.

Depression

Multiple sclerosis brings many variable and unpredictable challenges and can be a source of distress. Often these challenges occur with the onset of new disease milestones, such as the diagnosis or an increased disability. Given the physical, cognitive, and social stresses, it is not surprising that depression is extremely common, appearing in about half of patients with MS over their lifetime.5 During the course of ordinary MS care, the majority of patients with depression can be identified by a brief screening and referred for additional assessment and treatment.

Fortunately, there are many available treatment options. Antidepressant medications have shown some efficacy.6 Cognitive behavioral therapy (CBT), a counseling strategy that helps individuals become more active, connects them with rewarding activities, and challenges maladaptive thought patterns, has been shown to be effective in individual and group counseling settings via in-person or telephone-based delivery.7,8 Anxiety is also common experience among patients with MS and is treated with many of the same types of psychotherapy intervention.9

Focusing on the psychological and social needs of patients with MS has obvious implications for holistic care and QOL, but in some instances, MS may also contribute to safety concerns. Nearly one-third of veterans with MS admit to suicidal ideation, and the ultimate risk of suicide is about twice that of similar individuals without MS.10,11 For this reason, screening for risk of self-harm should be routinely incorporated into MS care.

A quick look at William’s Computerized Patient Record System (CPRS) record revealed that he had called the VA suicide prevention hotline. During the conversation he had noted that although he originally thought he would be able to deal with MS on his own, he realized he couldn’t. When the psychologist asked William about life at home, he disclosed that some days he never left his bed except to go to the bathroom. He stated he had given up on dating, and asked “who would want me?” He reported little appetite or interest in sex.

William was anxious about the problems he faced from day to day and grieving about the future that he no longer believed was possible. His distress was generally related to the MS diagnosis, and he spent a significant amount of time minimizing his disability, avoiding his family for this reason.

The psychologist diagnosed William with adjustment disorder with mixed anxiety and depressed mood and initiated individual CBT. The psychologist suggested that William attend the MS social work support group and the MS education group to get to know other veterans with MS and learn about managing symptoms. William agreed to attend the groups and admitted it would be good to have a reason to leave the house.

Health Behavior

Recognizing that MS is a chronic illness that requires coordinated efforts, the MS team helped William manage his disease and maintain his health. The psychological and social components of this process were considerable. For most newly diagnosed patients with MS, diseasemodifying therapies (DMTs) are important tools to decrease relapses and short-term disability. Although the benefits of these medications are well known, many patients are nonadherent. Contributing to poor adherence are adverse effects, cognitive challenges, anxiety, depression, and lack of belief in their efficacy.12,13 Brief
counseling, problem solving, and clinical monitoring have all been shown to reduce missed doses and improve DMT use.13 Both the MS Assessment Tool and the pharmacy database within CPRS are helpful for tracking patient adherence over time.

Other health behaviors may contribute not only to overall health, but also to the disease course. Patients who smoke have accelerated progression of their MS disease process and greater mortality than that of nonsmokers.14 Likewise, patients who engage in regular physical activity experience not only greater strength and endurance, but also less fatigue, depression, and better QOL.15 As part of his chronic illness care, the MS team provided William with information about the potential impact of health behaviors on MS progression.

William’s emotional and cognitive symptoms of MS presented important challenges to the management of his MS care. Initially, he ignored his diagnosis, delayed care, and refused to take a DMT. Once he agreed to taking a DMT, he often forgot to take it. He frequently missed medical appointments, because he did not remember them. Depression, fatigue, and stress decreased his motivation to follow through with recommendations from his health care providers.

Given William’s multiple psychosocial needs and transportation challenges, VA psychologists initiated telehealth visits with William in addition to clinic visits to provide many services, including psychotherapy and health behavior counseling. This continued support, along with the coordination of the rest of his health care team has been vital to maintaining William’s adherence to his treatment plan and QOL.

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

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

Click here for the digital edition.

References

1. Rao SM, Leo GJ, Bernardin L, Unverzagt F. Cognitive dysfunction in multiple sclerosis. I. Frequency, patterns, and prediction. Neurology. 1991;41(5):685-691.

2. Cicerone KD, Langenbahn DM, Braden C, et al. Evidence-based cognitive rehabilitation: Updated review of the literature from 2003 through 2008. Arch Phys Med Rehabil. 2011;92(4):519-530.

3. Chiaravalloti ND, Moore NB, Nikelshpur OM, DeLuca J. An RCT to treat learning impairment in multiple sclerosis: The MEMREHAB trial. Neurology. 2013;81(24):2066-2072.

4. Huckans M, Pavawalla S, Demadura T, et al. A pilot study examining effects of group-based Cognitive Strategy Training treatment on self-reported cognitive problems, psychiatric symptoms, functioning, and compensatory strategy use in OIF/OEF combat veterans with persistent mild cognitive disorder and history of traumatic brain injury. J Rehabil Res Dev. 2010;47(1):43-60.

5. Sadovnick AD, Remick RA, Allen J, et al. Depression and multiple sclerosis. Neurology. 1996;46(3):628-632.

6. Wallin MT, Wilken JA, Turner AP, Williams RM, Kane R. Depression and multiple
sclerosis: Review of a lethal combination. J Rehabil Res Dev. 2006;43(1):45-62.

7. Mohr DC, Boudewyn AC, Goodkin DE, Bostrom A, Epstein L. Comparative outcomes for individual cognitive-behavior therapy, supportive-expressive group psychotherapy, and sertraline for the treatment of depression in multiple sclerosis. J Consult Clin Psychol. 2001;69(6):942-949.

8. Mohr DC, Hart SL, Julian L, et al. Telephone-administered psychotherapy for depression. Arch Gen Psychiatry. 2005;62(9):1007-1014.

9. Korostil M, Feinstein A. Anxiety disorders and their clinical correlates in multiple sclerosis patients. Mult Scler. 2007;13(1):67-72.

10. Turner AP, Williams RM, Bowen JD, Kivlahan DR, Haselkorn JK. Suicidal ideation in multiple sclerosis. Arch Phys Med Rehabil. 2006;87(8):1073-1078.

11. Stenager EN, Stenager E. Suicide and patients with neurologic diseases. Methodologic problems. Arch Neurol. 1992;49(12):1296-1303.

12. Bruce JM, Hancock LM, Arnett P, Lynch S. Treatment adherence in multiple sclerosis:
Association with emotional status, personality, and cognition. J Behav Med. 2010;33(3):219-227.

13. Turner AP, Kivlahan DR, Sloan AP, Haselkorn JK. Predicting ongoing adherence to disease modifying therapies in multiple sclerosis: Utility of the health beliefs model. Mult Scler. 2007;13(9):1146-1152.

14. Overs S, Hughes CM, Haselkorn JK, Turner AP. Modifiable comorbidities and disability in multiple sclerosis. Curr Neurol Neurosci Rep. 2012;12(5):610-617.

15. Motl RW, Pilutti LA. The benefits of exercise training in multiple sclerosis. Nat Rev Neurol. 2012;8(9):487-497.

Author and Disclosure Information

Ms. Settle is a research coordinator at the Washington, DC VAMC. Dr. Turner is director of Rehabilitation Psychology, VA Puget Sound Health Care System and associate professor at the University of Washington's Department of Rehabilitation Medicine, both in Seattle.

Issue
Federal Practitioner - 32(3)s
Publications
Topics
Sections
Author and Disclosure Information

Ms. Settle is a research coordinator at the Washington, DC VAMC. Dr. Turner is director of Rehabilitation Psychology, VA Puget Sound Health Care System and associate professor at the University of Washington's Department of Rehabilitation Medicine, both in Seattle.

Author and Disclosure Information

Ms. Settle is a research coordinator at the Washington, DC VAMC. Dr. Turner is director of Rehabilitation Psychology, VA Puget Sound Health Care System and associate professor at the University of Washington's Department of Rehabilitation Medicine, both in Seattle.

Multiple sclerosis (MS) poses a host of cognitive and psychosocial challenges that may contribute to functioning and quality of life (QOL). Although each patient’s experience with MS is different, some challenges are more common than are others, including cognitive changes, depression, and maintaining positive health behaviors. William’s case study illustrates some of these challenges as well as the resources and strategies of the psychologists at the MS
Centers of Excellence (MSCoE) to help patients with MS adapt and thrive.

Cognition

Difficulties with cognitive functioning are common in patients with MS. About half of patients with MS will develop cognitive impairments in one or more areas during their lifetime.1 Although cognitive difficulties tend to worsen over the course of the disease, they can appear at any point in the illness, differ greatly from patient to patient, and are only modestly correlated with physical symptoms.1 Impairments are most common in the areas of information processing speed and memory, as well as in complex attention and mental flexibility. These impairments can impact activities of daily living, sustained employment, driving, and social relationships.

Early in the disease, William, who admitted to difficulties with cognition and the impact of cognitive impairment on his life, benefited from neuropsychological testing. Typically, an MSCoE will use a battery of tests tailored to patients with MS. For patients, the results of these tests can be used to clarify areas of relative strength and weakness and inform team decisions related to treatment and future life activities, such as whether the patient will need accommodations at work. Test results may also be used to guide how the MS treatment team interacts with the patient.

An initial clinic screening from neurology indicated William had below average cognitive processing speed, and he agreed to neurocognitive testing. The psycholopsychologist asked William about his functioning at home. William noted poor attention and memory made it difficult to advocate for himself. He recounted an episode of taking his car to be repaired and only later realizing that he had been charged twice for the same part. He also disclosed that he had gotten lost while driving through familiar places. On several occasions while cooking, he had become distracted and started another task only to find he had burned his food.

Cognitive rehabilitation has shown considerable promise in helping patients work through difficulties with memory, attention, and problem solving by developing compensatory strategies.2 Skill training is available in individual and group formats. There is also promising but very preliminary evidence that some psychosocial interventions might improve memory performance for patients with MS.3

William’s neuropsychological testing confirmed impairments in information processing speed, attention, and memory, and William was diagnosed with cognitive disorder not otherwise specified. His cognitive impairment correlated with his magnetic resonance imaging (MRI) findings, which included lesions on the corpus callosum and in multiple subcortical areas.

During a feedback session, William was encouraged to use compensatory strategies, including memory aids, visual cueing, and self-pacing. He was given a referral to speech and language pathology to develop and practice these strategies. The psychologist also reminded William’s health care team to speak slowly and repeat important information to him, write down important instructions, and cue him when asking questions in the clinic. William was provided with a kitchen timer and instructed to set it whenever he began cooking, so that even if he got distracted, the alarm would remind him to return to his cooking. William was asked whether he would like to participate in the MSCoE cognitive compensatory training program.4 The program, offered through a research protocol, involved a series of classes that taught strategies to manage cognitive symptoms and improve patients’ ability to function independently. William agreed to participate and reported feeling hopeful that his situation could improve.

Depression

Multiple sclerosis brings many variable and unpredictable challenges and can be a source of distress. Often these challenges occur with the onset of new disease milestones, such as the diagnosis or an increased disability. Given the physical, cognitive, and social stresses, it is not surprising that depression is extremely common, appearing in about half of patients with MS over their lifetime.5 During the course of ordinary MS care, the majority of patients with depression can be identified by a brief screening and referred for additional assessment and treatment.

Fortunately, there are many available treatment options. Antidepressant medications have shown some efficacy.6 Cognitive behavioral therapy (CBT), a counseling strategy that helps individuals become more active, connects them with rewarding activities, and challenges maladaptive thought patterns, has been shown to be effective in individual and group counseling settings via in-person or telephone-based delivery.7,8 Anxiety is also common experience among patients with MS and is treated with many of the same types of psychotherapy intervention.9

Focusing on the psychological and social needs of patients with MS has obvious implications for holistic care and QOL, but in some instances, MS may also contribute to safety concerns. Nearly one-third of veterans with MS admit to suicidal ideation, and the ultimate risk of suicide is about twice that of similar individuals without MS.10,11 For this reason, screening for risk of self-harm should be routinely incorporated into MS care.

A quick look at William’s Computerized Patient Record System (CPRS) record revealed that he had called the VA suicide prevention hotline. During the conversation he had noted that although he originally thought he would be able to deal with MS on his own, he realized he couldn’t. When the psychologist asked William about life at home, he disclosed that some days he never left his bed except to go to the bathroom. He stated he had given up on dating, and asked “who would want me?” He reported little appetite or interest in sex.

William was anxious about the problems he faced from day to day and grieving about the future that he no longer believed was possible. His distress was generally related to the MS diagnosis, and he spent a significant amount of time minimizing his disability, avoiding his family for this reason.

The psychologist diagnosed William with adjustment disorder with mixed anxiety and depressed mood and initiated individual CBT. The psychologist suggested that William attend the MS social work support group and the MS education group to get to know other veterans with MS and learn about managing symptoms. William agreed to attend the groups and admitted it would be good to have a reason to leave the house.

Health Behavior

Recognizing that MS is a chronic illness that requires coordinated efforts, the MS team helped William manage his disease and maintain his health. The psychological and social components of this process were considerable. For most newly diagnosed patients with MS, diseasemodifying therapies (DMTs) are important tools to decrease relapses and short-term disability. Although the benefits of these medications are well known, many patients are nonadherent. Contributing to poor adherence are adverse effects, cognitive challenges, anxiety, depression, and lack of belief in their efficacy.12,13 Brief
counseling, problem solving, and clinical monitoring have all been shown to reduce missed doses and improve DMT use.13 Both the MS Assessment Tool and the pharmacy database within CPRS are helpful for tracking patient adherence over time.

Other health behaviors may contribute not only to overall health, but also to the disease course. Patients who smoke have accelerated progression of their MS disease process and greater mortality than that of nonsmokers.14 Likewise, patients who engage in regular physical activity experience not only greater strength and endurance, but also less fatigue, depression, and better QOL.15 As part of his chronic illness care, the MS team provided William with information about the potential impact of health behaviors on MS progression.

William’s emotional and cognitive symptoms of MS presented important challenges to the management of his MS care. Initially, he ignored his diagnosis, delayed care, and refused to take a DMT. Once he agreed to taking a DMT, he often forgot to take it. He frequently missed medical appointments, because he did not remember them. Depression, fatigue, and stress decreased his motivation to follow through with recommendations from his health care providers.

Given William’s multiple psychosocial needs and transportation challenges, VA psychologists initiated telehealth visits with William in addition to clinic visits to provide many services, including psychotherapy and health behavior counseling. This continued support, along with the coordination of the rest of his health care team has been vital to maintaining William’s adherence to his treatment plan and QOL.

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

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

Click here for the digital edition.

Multiple sclerosis (MS) poses a host of cognitive and psychosocial challenges that may contribute to functioning and quality of life (QOL). Although each patient’s experience with MS is different, some challenges are more common than are others, including cognitive changes, depression, and maintaining positive health behaviors. William’s case study illustrates some of these challenges as well as the resources and strategies of the psychologists at the MS
Centers of Excellence (MSCoE) to help patients with MS adapt and thrive.

Cognition

Difficulties with cognitive functioning are common in patients with MS. About half of patients with MS will develop cognitive impairments in one or more areas during their lifetime.1 Although cognitive difficulties tend to worsen over the course of the disease, they can appear at any point in the illness, differ greatly from patient to patient, and are only modestly correlated with physical symptoms.1 Impairments are most common in the areas of information processing speed and memory, as well as in complex attention and mental flexibility. These impairments can impact activities of daily living, sustained employment, driving, and social relationships.

Early in the disease, William, who admitted to difficulties with cognition and the impact of cognitive impairment on his life, benefited from neuropsychological testing. Typically, an MSCoE will use a battery of tests tailored to patients with MS. For patients, the results of these tests can be used to clarify areas of relative strength and weakness and inform team decisions related to treatment and future life activities, such as whether the patient will need accommodations at work. Test results may also be used to guide how the MS treatment team interacts with the patient.

An initial clinic screening from neurology indicated William had below average cognitive processing speed, and he agreed to neurocognitive testing. The psycholopsychologist asked William about his functioning at home. William noted poor attention and memory made it difficult to advocate for himself. He recounted an episode of taking his car to be repaired and only later realizing that he had been charged twice for the same part. He also disclosed that he had gotten lost while driving through familiar places. On several occasions while cooking, he had become distracted and started another task only to find he had burned his food.

Cognitive rehabilitation has shown considerable promise in helping patients work through difficulties with memory, attention, and problem solving by developing compensatory strategies.2 Skill training is available in individual and group formats. There is also promising but very preliminary evidence that some psychosocial interventions might improve memory performance for patients with MS.3

William’s neuropsychological testing confirmed impairments in information processing speed, attention, and memory, and William was diagnosed with cognitive disorder not otherwise specified. His cognitive impairment correlated with his magnetic resonance imaging (MRI) findings, which included lesions on the corpus callosum and in multiple subcortical areas.

During a feedback session, William was encouraged to use compensatory strategies, including memory aids, visual cueing, and self-pacing. He was given a referral to speech and language pathology to develop and practice these strategies. The psychologist also reminded William’s health care team to speak slowly and repeat important information to him, write down important instructions, and cue him when asking questions in the clinic. William was provided with a kitchen timer and instructed to set it whenever he began cooking, so that even if he got distracted, the alarm would remind him to return to his cooking. William was asked whether he would like to participate in the MSCoE cognitive compensatory training program.4 The program, offered through a research protocol, involved a series of classes that taught strategies to manage cognitive symptoms and improve patients’ ability to function independently. William agreed to participate and reported feeling hopeful that his situation could improve.

Depression

Multiple sclerosis brings many variable and unpredictable challenges and can be a source of distress. Often these challenges occur with the onset of new disease milestones, such as the diagnosis or an increased disability. Given the physical, cognitive, and social stresses, it is not surprising that depression is extremely common, appearing in about half of patients with MS over their lifetime.5 During the course of ordinary MS care, the majority of patients with depression can be identified by a brief screening and referred for additional assessment and treatment.

Fortunately, there are many available treatment options. Antidepressant medications have shown some efficacy.6 Cognitive behavioral therapy (CBT), a counseling strategy that helps individuals become more active, connects them with rewarding activities, and challenges maladaptive thought patterns, has been shown to be effective in individual and group counseling settings via in-person or telephone-based delivery.7,8 Anxiety is also common experience among patients with MS and is treated with many of the same types of psychotherapy intervention.9

Focusing on the psychological and social needs of patients with MS has obvious implications for holistic care and QOL, but in some instances, MS may also contribute to safety concerns. Nearly one-third of veterans with MS admit to suicidal ideation, and the ultimate risk of suicide is about twice that of similar individuals without MS.10,11 For this reason, screening for risk of self-harm should be routinely incorporated into MS care.

A quick look at William’s Computerized Patient Record System (CPRS) record revealed that he had called the VA suicide prevention hotline. During the conversation he had noted that although he originally thought he would be able to deal with MS on his own, he realized he couldn’t. When the psychologist asked William about life at home, he disclosed that some days he never left his bed except to go to the bathroom. He stated he had given up on dating, and asked “who would want me?” He reported little appetite or interest in sex.

William was anxious about the problems he faced from day to day and grieving about the future that he no longer believed was possible. His distress was generally related to the MS diagnosis, and he spent a significant amount of time minimizing his disability, avoiding his family for this reason.

The psychologist diagnosed William with adjustment disorder with mixed anxiety and depressed mood and initiated individual CBT. The psychologist suggested that William attend the MS social work support group and the MS education group to get to know other veterans with MS and learn about managing symptoms. William agreed to attend the groups and admitted it would be good to have a reason to leave the house.

Health Behavior

Recognizing that MS is a chronic illness that requires coordinated efforts, the MS team helped William manage his disease and maintain his health. The psychological and social components of this process were considerable. For most newly diagnosed patients with MS, diseasemodifying therapies (DMTs) are important tools to decrease relapses and short-term disability. Although the benefits of these medications are well known, many patients are nonadherent. Contributing to poor adherence are adverse effects, cognitive challenges, anxiety, depression, and lack of belief in their efficacy.12,13 Brief
counseling, problem solving, and clinical monitoring have all been shown to reduce missed doses and improve DMT use.13 Both the MS Assessment Tool and the pharmacy database within CPRS are helpful for tracking patient adherence over time.

Other health behaviors may contribute not only to overall health, but also to the disease course. Patients who smoke have accelerated progression of their MS disease process and greater mortality than that of nonsmokers.14 Likewise, patients who engage in regular physical activity experience not only greater strength and endurance, but also less fatigue, depression, and better QOL.15 As part of his chronic illness care, the MS team provided William with information about the potential impact of health behaviors on MS progression.

William’s emotional and cognitive symptoms of MS presented important challenges to the management of his MS care. Initially, he ignored his diagnosis, delayed care, and refused to take a DMT. Once he agreed to taking a DMT, he often forgot to take it. He frequently missed medical appointments, because he did not remember them. Depression, fatigue, and stress decreased his motivation to follow through with recommendations from his health care providers.

Given William’s multiple psychosocial needs and transportation challenges, VA psychologists initiated telehealth visits with William in addition to clinic visits to provide many services, including psychotherapy and health behavior counseling. This continued support, along with the coordination of the rest of his health care team has been vital to maintaining William’s adherence to his treatment plan and QOL.

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

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

Click here for the digital edition.

References

1. Rao SM, Leo GJ, Bernardin L, Unverzagt F. Cognitive dysfunction in multiple sclerosis. I. Frequency, patterns, and prediction. Neurology. 1991;41(5):685-691.

2. Cicerone KD, Langenbahn DM, Braden C, et al. Evidence-based cognitive rehabilitation: Updated review of the literature from 2003 through 2008. Arch Phys Med Rehabil. 2011;92(4):519-530.

3. Chiaravalloti ND, Moore NB, Nikelshpur OM, DeLuca J. An RCT to treat learning impairment in multiple sclerosis: The MEMREHAB trial. Neurology. 2013;81(24):2066-2072.

4. Huckans M, Pavawalla S, Demadura T, et al. A pilot study examining effects of group-based Cognitive Strategy Training treatment on self-reported cognitive problems, psychiatric symptoms, functioning, and compensatory strategy use in OIF/OEF combat veterans with persistent mild cognitive disorder and history of traumatic brain injury. J Rehabil Res Dev. 2010;47(1):43-60.

5. Sadovnick AD, Remick RA, Allen J, et al. Depression and multiple sclerosis. Neurology. 1996;46(3):628-632.

6. Wallin MT, Wilken JA, Turner AP, Williams RM, Kane R. Depression and multiple
sclerosis: Review of a lethal combination. J Rehabil Res Dev. 2006;43(1):45-62.

7. Mohr DC, Boudewyn AC, Goodkin DE, Bostrom A, Epstein L. Comparative outcomes for individual cognitive-behavior therapy, supportive-expressive group psychotherapy, and sertraline for the treatment of depression in multiple sclerosis. J Consult Clin Psychol. 2001;69(6):942-949.

8. Mohr DC, Hart SL, Julian L, et al. Telephone-administered psychotherapy for depression. Arch Gen Psychiatry. 2005;62(9):1007-1014.

9. Korostil M, Feinstein A. Anxiety disorders and their clinical correlates in multiple sclerosis patients. Mult Scler. 2007;13(1):67-72.

10. Turner AP, Williams RM, Bowen JD, Kivlahan DR, Haselkorn JK. Suicidal ideation in multiple sclerosis. Arch Phys Med Rehabil. 2006;87(8):1073-1078.

11. Stenager EN, Stenager E. Suicide and patients with neurologic diseases. Methodologic problems. Arch Neurol. 1992;49(12):1296-1303.

12. Bruce JM, Hancock LM, Arnett P, Lynch S. Treatment adherence in multiple sclerosis:
Association with emotional status, personality, and cognition. J Behav Med. 2010;33(3):219-227.

13. Turner AP, Kivlahan DR, Sloan AP, Haselkorn JK. Predicting ongoing adherence to disease modifying therapies in multiple sclerosis: Utility of the health beliefs model. Mult Scler. 2007;13(9):1146-1152.

14. Overs S, Hughes CM, Haselkorn JK, Turner AP. Modifiable comorbidities and disability in multiple sclerosis. Curr Neurol Neurosci Rep. 2012;12(5):610-617.

15. Motl RW, Pilutti LA. The benefits of exercise training in multiple sclerosis. Nat Rev Neurol. 2012;8(9):487-497.

References

1. Rao SM, Leo GJ, Bernardin L, Unverzagt F. Cognitive dysfunction in multiple sclerosis. I. Frequency, patterns, and prediction. Neurology. 1991;41(5):685-691.

2. Cicerone KD, Langenbahn DM, Braden C, et al. Evidence-based cognitive rehabilitation: Updated review of the literature from 2003 through 2008. Arch Phys Med Rehabil. 2011;92(4):519-530.

3. Chiaravalloti ND, Moore NB, Nikelshpur OM, DeLuca J. An RCT to treat learning impairment in multiple sclerosis: The MEMREHAB trial. Neurology. 2013;81(24):2066-2072.

4. Huckans M, Pavawalla S, Demadura T, et al. A pilot study examining effects of group-based Cognitive Strategy Training treatment on self-reported cognitive problems, psychiatric symptoms, functioning, and compensatory strategy use in OIF/OEF combat veterans with persistent mild cognitive disorder and history of traumatic brain injury. J Rehabil Res Dev. 2010;47(1):43-60.

5. Sadovnick AD, Remick RA, Allen J, et al. Depression and multiple sclerosis. Neurology. 1996;46(3):628-632.

6. Wallin MT, Wilken JA, Turner AP, Williams RM, Kane R. Depression and multiple
sclerosis: Review of a lethal combination. J Rehabil Res Dev. 2006;43(1):45-62.

7. Mohr DC, Boudewyn AC, Goodkin DE, Bostrom A, Epstein L. Comparative outcomes for individual cognitive-behavior therapy, supportive-expressive group psychotherapy, and sertraline for the treatment of depression in multiple sclerosis. J Consult Clin Psychol. 2001;69(6):942-949.

8. Mohr DC, Hart SL, Julian L, et al. Telephone-administered psychotherapy for depression. Arch Gen Psychiatry. 2005;62(9):1007-1014.

9. Korostil M, Feinstein A. Anxiety disorders and their clinical correlates in multiple sclerosis patients. Mult Scler. 2007;13(1):67-72.

10. Turner AP, Williams RM, Bowen JD, Kivlahan DR, Haselkorn JK. Suicidal ideation in multiple sclerosis. Arch Phys Med Rehabil. 2006;87(8):1073-1078.

11. Stenager EN, Stenager E. Suicide and patients with neurologic diseases. Methodologic problems. Arch Neurol. 1992;49(12):1296-1303.

12. Bruce JM, Hancock LM, Arnett P, Lynch S. Treatment adherence in multiple sclerosis:
Association with emotional status, personality, and cognition. J Behav Med. 2010;33(3):219-227.

13. Turner AP, Kivlahan DR, Sloan AP, Haselkorn JK. Predicting ongoing adherence to disease modifying therapies in multiple sclerosis: Utility of the health beliefs model. Mult Scler. 2007;13(9):1146-1152.

14. Overs S, Hughes CM, Haselkorn JK, Turner AP. Modifiable comorbidities and disability in multiple sclerosis. Curr Neurol Neurosci Rep. 2012;12(5):610-617.

15. Motl RW, Pilutti LA. The benefits of exercise training in multiple sclerosis. Nat Rev Neurol. 2012;8(9):487-497.

Issue
Federal Practitioner - 32(3)s
Issue
Federal Practitioner - 32(3)s
Publications
Publications
Topics
Article Type
Sections
Citation Override
Fed Pract. 2015 April;32(suppl 3):20S-22S
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME