Genomic Testing in the Management of Early-Stage Breast Cancer

Article Type
Changed
Thu, 02/15/2018 - 16:17
Display Headline
Genomic Testing in the Management of Early-Stage Breast Cancer

From the University of Arizona Cancer Center, Tucson, AZ (Dr. Ehsani), and University of Wisconsin Carbone Cancer Center and School of Medicine and Public Health, Madison, WI (Dr. Wisinski).

 

Abstract

  • Objectives: To describe common genomic tests being used clinically to assess prognosis and guide adjuvant chemotherapy and endocrine therapy decisions for early-stage breast cancer.
  • Methods: Case presentation and review of the literature.
  • Results: Hormone receptor–positive (HR-positive) breast cancers, which express the estrogen and/or progesterone receptor, account for the majority of breast cancers. Endocrine therapy can be highly effective for patients with these HR-positive tumors, and identification of HR-positive breast cancers that do not require the addition of chemotherapy is critical. Clinicopathological features of the breast cancer, including tumor size, nodal involvement, grading, and HR status, are insufficient in predicting the risk for recurrence or the need for chemotherapy. Furthermore, a portion of HR-positive breast cancers have an ongoing risk for late recurrence, and longer durations of endocrine therapy are being used to reduce this risk.
  • Conclusion: There is sufficient evidence for use of genomic testing in early-stage HR-positive breast cancer to aid in chemotherapy recommendations. Further confirmation of genomic assays for prediction of benefit from prolonged endocrine therapy is needed.

Key words: molecular testing; decision aids; HR-positive cancer; recurrence risk; adjuvant chemotherapy; endocrine therapy.

 

 

Despite the increase in incidence of breast cancer, breast cancer mortality has decreased over the past several decades. This is likely due to both early detection and advances in systemic therapy. However, with more widespread use of screening mammography, there are increasing concerns regarding potential overdiagnosis of cancer [1]. One key challenge is that breast cancer is a heterogeneous disease. Thus, improved tools for determining breast cancer biology can help physicians individualize treatments, with low-risk cancers approached with less aggressive treatments, thus preventing unnecessary toxicities, and higher-risk cancers treated appropriately.

Traditionally, adjuvant chemotherapy was recommended based on tumor features such as stage (tumor size, regional nodal involvement), grade, expression of hormone receptors (estrogen receptor [ER] and progesterone receptor [PR]) and human epidermal growth factor receptor-2 (HER2), and patient features (age, menopausal status). However, this approach is not accurate enough to guide individualized treatment recommendations, which are based on the risk for recurrence and the reduction in this risk that can be achieved with various systemic treatments. In particular, there are individuals with low-risk HR-positive, HER2-negative breast cancers who could be spared the toxicities of cytotoxic chemotherapies without compromising the prognosis.

Beyond chemotherapy, endocrine therapies also have risks, especially when given for extended durations. Recently, extended endocrine therapy has been shown to prevent late recurrences of HR-positive breast cancers. In the MA.17R study, extended endocrine therapy with letrozole for a total of 10 years (beyond 5 years of an aromatase inhibitor [AI]) decreased the risk for breast cancer recurrence or the occurrence of contralateral breast cancer by 34% [2]. However, the overall survival was similar between the 2 groups and the results were not confirmed in other studies [3–5]. Identifying the subgroup of patients who benefit from this extended AI therapy is important in the era of personalized medicine. Several tumor genomic assays have been developed to provide additional prognostic and predictive information with the goal of individualizing adjuvant therapies for breast cancer. Although assays are also being evaluated in HER2-positive and triple negative breast cancer, this review will focus on HR-positive, HER2-negative breast cancer.

Case Study

Initial Presentation

A 54-year-old postmenopausal woman with no significant past medical history presents with an abnormal screening mammogram, which shows a focal asymmetry in the 10 o’clock position at middle depth of the left breast. Further work-up with a diagnostic mammogram and ultrasound of the left breast shows a suspicious hypoechoic solid mass with irregular margins measuring 17 mm. The patient undergoes an ultrasound-guided core needle biopsy of the suspicious mass, the results of which are consistent with an invasive ductal carcinoma, Nottingham grade 2, ER strongly positive (95%), PR weakly positive (5%), HER2 negative, and Ki-67 of 15%. She undergoes a left partial mastectomy and sentinel lymph node biopsy, with final pathology demonstrating a single focus of invasive ductal carcinoma, measuring 2.2 cm in greatest dimension with no evidence of lymphovascular invasion. Margins are clear and 2 sentinel lymph nodes are negative for metastatic disease (final pathologic stage IIA, pT2 pN0 cM0). She is referred to medical oncology to discuss adjuvant systemic therapy.

  • Can additional testing be used to determine prognosis and guide systemic therapy rec-ommendations for early-stage HR-positive/HER2-negative breast cancer?

After a diagnosis of early-stage breast cancer, the key clinical question faced by the patient and medical oncologist is: what is the individual’s risk for a metastatic breast cancer recurrence and thus the risk for death due to breast cancer? Once the risk for recurrence is established, systemic adjuvant chemotherapy, endocrine therapy, and/or HER2-directed therapy are considered based on the receptor status (ER/PR and HER2) to reduce this risk. Hormone receptor (HR)–positive, HER2-negative breast cancer is the most common type of breast cancer. Although adjuvant endocrine therapy has significantly reduced the risk for recurrence and improved survival for HR-positive breast cancer [6], the role of adjuvant chemotherapy for this subset of breast cancer remains unclear. Prior to genomic testing, the recommendation for adjuvant chemotherapy for HR-positive/HER2-negative tumors was primarily based on patient age and tumor stage and grade. However, chemotherapy overtreatment remained a concern given the potential short- and long-term risks of chemotherapy. Further studies into HR-positive/HER2-negative tumors have shown that these tumors can be divided into 2 main subtypes, luminal A and luminal B [7]. These subtypes represent unique biology and differ in terms of prognosis and response to endocrine therapy and chemotherapy. Luminal A tumors are strongly endocrine responsive and have a good prognosis, while luminal B tumors are less endocrine responsive and are associated with a poorer prognosis; the addition of adjuvant chemotherapy is often considered for luminal B tumors [8]. Several tests, including tumor genomic assays, are now available to help with delineating the tumor subtype and aid in decision-making regarding adjuvant chemotherapy for HR-positive/HER2-negative breast cancers.

Tests for Guiding Adjuvant Chemotherapy Decisions

Ki-67 Assays, Including IHC4 and PEPI

Chronic proliferation is a hallmark of cancer cells [9]. Ki-67, a nuclear nonhistone protein whose expression varies in intensity throughout the cell cycle, has been used as a measurement of tumor cell proliferation [10]. Two large meta-analyses have demonstrated that high Ki-67 expression in breast tumors is independently associated with worse disease-free and overall survival rates [11,12]. Ki-67 expression has also been used to classify HR-positive tumors as luminal A or B. After classifying tumor subtypes based on intrinsic gene expression profiling, Cheang et al determined that a Ki-67 cut point of 13.25% differentiated luminal A and B tumors [13]. However, the ideal cut point for Ki-67 remains unclear, as the sensitivity and specificity in this study was 77% and 78%, respectively. Others have combined Ki-67 with standard ER, PR, and HER2 testing. This IHC4 score, which weighs each of these variables, was validated in postmenopausal patients from the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial who had ER-positive tumors and did not receive chemotherapy [14]. The prognostic information from the IHC4 was similar to that seen with the 21-gene recurrence score (Oncotype DX), which is discussed later in this article. The key challenge with Ki-67 testing currently is the lack of a validated test methodology, and intraobserver variability in interpreting the Ki-67 results [15]. Recent series have suggested that Ki-67 be considered as a continuous marker rather than a set cut point [16]. These issues continue to impact the clinical utility of Ki-67 for decision making for adjuvant chemotherapy.

 

 

Ki-67 and the preoperative endocrine prognostic index (PEPI) score have been explored in the neoadjuvant setting to separate postmenopausal women with endocrine-sensitive versus intrinsically resistant disease and identify patients at risk for recurrent disease [17]. The on-treatment levels of Ki-67 in response to endocrine therapy have been shown to be more prognostic than baseline values, and a decrease in Ki-67 as early as 2 weeks after initiation of neoadjuvant endocrine therapy is associated with endocrine-sensitive tumors and improved outcome. The PEPI score was developed through retrospective analysis of the P024 trial [18] to evaluate the relationship between post-neoadjuvant endocrine therapy tumor characteristics and risk for early relapse. This was subsequently validated in an independent data set from the IMPACT trial [19]. Patients with low pathological stage (0 or 1) and a favorable biomarker profile (PEPI score 0) at surgery had the best prognosis in the absence of chemotherapy. On the other hand, higher pathological stage at surgery and a poor biomarker profile with loss of ER positivity or persistently elevated Ki-67 (PEPI score of 3) identified de novo endocrine-resistant tumors which are at higher risk for early relapse [20]. The ongoing Alliance A011106 ALTERNATE trial (ALTernate approaches for clinical stage II or III Estrogen Receptor positive breast cancer NeoAdjuvant TrEatment in postmenopausal women, NCT01953588) is a phase 3 study to prospectively test this hypothesis.

21-Gene Recurrence Score (Oncotype DX Assay)

The 21-gene Oncotype DX assay is conducted on paraffin-embedded tumor tissue and measures the expression of 16 cancer-related genes and 5 reference genes using quantitative polymerase chain reaction. The genes included in this assay are mainly related to proliferation (including Ki-67), invasion, and HER2 or estrogen signaling [21]. Originally, the 21-gene recurrence score assay was analyzed as a prognostic biomarker tool in a prospective-retrospective biomarker substudy of the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14 clinical trial in which patients with node-negative, ER-positive tumors were randomly assigned to receive tamoxifen or placebo without chemotherapy [22]. Using the standard reported values of low risk (< 18), intermediate risk (18–30), or high risk (≥ 31) for recurrence, among the tamoxifen-treated patients, cancers with a high-risk recurrence score had a significantly worse rate of distant recurrence and overall survival [21]. Inferior breast cancer survival with a high recurrence score was also confirmed in other series of endocrine-treated patients with node-negative and node-positive disease [23–25].

The predictive utility of the 21-gene recurrence score for endocrine therapy has also been evaluated. A comparison of the placebo- and tamoxifen-treated patients from the NSABP B-14 trial demonstrated that the 21-gene recurrence score predicted benefit from tamoxifen in cancers with low- or intermediate-risk recurrence scores [26]. However, there was no benefit from the use of tamoxifen over placebo in cancers with high-risk recurrence scores. To date, this intriguing data has not been prospectively confirmed, and thus the 21-gene recurrence score is not used to avoid endocrine therapy.

The 21-gene recurrence score is primarily used by oncologists to aid in decision-making regarding adjuvant chemotherapy in patients with node-negative and node-positive (with up to 3 positive lymph nodes), HR-positive/HER2-negative breast cancers. The predictive utility of the 21-gene recurrence score for adjuvant chemotherapy was initially tested using tumor samples from the NSABP B-20 study. This study initially compared adjuvant tamoxifen alone with tamoxifen plus chemotherapy in patients with node-negative, HR-positive tumors. The prospective-retrospective biomarker analysis showed that the patients with high-risk 21-gene recurrence scores benefited from the addition of chemotherapy, whereas those with low- or intermediate-risk did not have an improved freedom from distant recurrence with chemotherapy [27]. Similarly, an analysis from the prospective phase 3 Southwest Oncology Group (SWOG) 8814 trial comparing tamoxifen to tamoxifen with chemotherapy showed that for node-positive tumors, chemotherapy benefit was only seen in those with high 21-gene recurrence scores [24].

Prospective studies are now starting to report results regarding the predictive role of the 21-gene recurrence score. The TAILORx (Trial Assigning Individualized Options for Treatment) trial includes women with node-negative, HR-positive and HER2-negative tumors measuring 0.6 to 5 cm. All patients were treated with standard of care endocrine therapy for at least 5 years. Chemotherapy was determined based on the 21-gene recurrence score results on the primary tumor. The 21-gene recurrence score cutoffs were changed to low (0–10), intermediate (11–25), and high (≥ 26). Patients with scores of 26 or higher were treated with chemotherapy, and those with intermediate scores were randomly assigned to hemotherapy or no chemotherapy; results from this cohort are still pending. However, excellent breast cancer outcomes with endocrine therapy alone were reported from the 1626 (15.9% of total cohort) prospectively followed patients with low-recurrence score tumors. The 5-year invasive disease-free survival was 93.8%, with overall survival of 98% [28]. Given that 5 years is appropriate follow-up to see any chemotherapy benefit, this data supports the recommendation for no chemotherapy in this cohort of patients with very low 21-gene recurrence scores.

The RxPONDER (Rx for Positive Node, Endocrine Responsive Breast Cancer) trial is evaluating women with 1 to 3 node-positive, HR-positive, HER2-negative tumors. In this trial, patients with 21-gene recurrence scores of 0 to 25 were assigned to adjuvant chemotherapy or none. Those with scores of 26 or higher were assigned to chemotherapy. All patients received standard adjuvant endocrine therapy. This study has completed accrual and results are pending. Of note, TAILORx and RxPONDER did not investigate the potential lack of benefit of endocrine therapy in cancers with high recurrence scores. Furthermore, despite data suggesting that chemotherapy may not even benefit women with 4 or more nodes involved but who have a low recurrence score [24], due to the lack of prospective data in this cohort and the quite high risk for distant recurrence, chemotherapy continues to be the standard of care for these patients.

PAM50 (Breast Cancer Prognostic Gene Signature)

Using microarray and quantitative reverse transcriptase PCR (RT-PCR) on formalin-fixed paraffin-embedded (FFPE) tissues, the Breast Cancer Prognostic Gene Signature (PAM50) assay was initially developed to identify intrinsic breast cancer subtypes, including luminal A, luminal B, HER2-enriched, and basal-like [7,29]. Based on the prediction analysis of microarray (PAM) method, the assay measures the expression levels of 50 genes, provides a risk category (low, intermediate, and high), and generates a numerical risk of recurrence score (ROR). The intrinsic subtype and ROR have been shown to add significant prognostic value to the clinicopathological characteristics of tumors. Clinical validity of PAM50 was evaluated in postmenopausal women with HR-positive, early-stage breast cancer treated in the prospective ATAC and ABCSG-8 (Austrian Breast and Colorectal Cancer Study Group 8) trials [30,31]. In 1017 patients with ER-positive breast cancer treated with anastrozole or tamoxifen in the ATAC trial, ROR added significant prognostic information beyond the clinical treatment score (integrated prognostic information from nodal status, tumor size, histopathologic grade, age, and anastrozole or tamoxifen treatment) in all patients. Also, compared with the 21-gene recurrence score, ROR provided more prognostic information in ER-positive, node-negative disease and better differentiation of intermediate- and higher-risk groups. Fewer patients were categorized as intermediate risk by ROR and more as high risk, which could reduce the uncertainty in the estimate of clinical benefit from chemotherapy [30]. The clinical utility of PAM50 as a prognostic model was also validated in 1478 postmenopausal women with ER-positive early-stage breast cancer enrolled in the ABCSG-8 trial. In this study, ROR assigned 47% of patients with node-negative disease to the low-risk category. In this low-risk group, the 10-year metastasis risk was less than 3.5 %, indicating lack of benefit from additional chemotherapy [31]. A key limitation of the PAM50 is the lack of any prospective studies with this assay.

PAM50 has been designed to be carried out in any qualified pathology laboratory. Moreover, the ROR score provides additional prognostic information about risk of late recurrence, which will be discussed in the next section.

 

 

70-Gene Breast Cancer Recurrence Assay (MammaPrint)

MammaPrint is a 70-gene assay that was initially developed using an unsupervised, hierarchical clustering algorithm on whole-genome expression arrays with early-stage breast cancer. Among 295 consecutive patients who had MammaPrint testing, those classified with a good-prognosis tumor signature (n = 115) had an excellent 10-year survival rate (94.5%) compared to those with a poor-prognosis signature (54.5%), and the signature remained prognostic upon multivariate analysis [32]. Subsequently, a pooled analysis comparing outcomes by MammaPrint score in patients with node-negative or 1 to 3 node-positive breast cancers treated as per discretion of their medical team with either adjuvant chemotherapy plus endocrine therapy or endocrine therapy alone reported that only those patients with a high-risk score benefited from chemotherapy [33]. Recently, a prospective phase 3 study (MINDACT [Microarray In Node negative Disease may Avoid ChemoTherapy]) evaluating the utility of MammaPrint for adjuvant chemotherapy decision-making reported results [34]. In this study, 6693 women with early-stage breast cancer were assessed by clinical risk and genomic risk using MammaPrint. Those with low clinical and genomic risk did not receive chemotherapy, while those with high clinical and genomic risk all received chemotherapy. The primary goal of the study was to assess whether forgoing chemotherapy would be associated with a low rate of recurrence in those patients with a low-risk prognostic MammaPrint signature but high clinical risk. A total of 1550 patients (23.2%) were in the discordant group, and the majority of these patients had HR-positive disease (98.1%). Without chemotherapy, the rate of survival without distant metastasis at 5 years in this group was 94.7% (95% confidence interval [CI] 92.5% to 96.2%), which met the primary endpoint. Of note, initially, MammaPrint was only available for fresh tissue analysis, but recent advances in RNA processing now allow for this analysis on FFPE tissue [35].

Summary

These genomic and biomarker assays can identify different subsets of HR-positive breast cancers, including those patients who have tumors with an excellent prognosis with endocrine therapies alone. Thus, we now have the tools to help avoid the toxicities of chemotherapy in many women with early-stage breast cancer. A summary of the genomic tests available is shown in Table 1.

 

 

Case Continued

The patient undergoes 21-gene recurrence score testing, which shows a low recurrence score of 10, estimating the 10-year risk of distant recurrence to be approximately 7% with 5 years of tamoxifen. Chemo-therapy is not recommended. The patient completes adjuvant whole breast radiation therapy, and then, based on data supporting AIs over tamoxifen in postmenopausal women, she is started on anastrozole [36]. She initially experiences mild side effects from treatment, including fatigue, arthralgia, and vaginal dryness, but her symptoms are able to be managed. As she approaches 5 years of adjuvant endocrine therapy with anastrozole, she is struggling with rotator cuff injury and is anxious about recurrence, but has no evidence of recurrent cancer. Her bone density scan in the beginning of her fourth year of therapy shows a decrease in bone mineral density, with the lowest T score of –1.5 at the left femoral neck, consistent with osteopenia. She has been treated with calcium and vitamin D supplements.

  • How long should this patient continue treatment with anastrozole?

The risk for recurrence is highest during the first 5 years after diagnosis for all patients with early breast cancer [37]. Although HR-positive breast cancers have a better prognosis than HR-negative disease, the pattern of recurrence is different between the 2 groups, and it is estimated that approximately half of the recurrences among patients with HR-positive early breast cancer occur after the first 5 years from diagnosis. Annualized hazard of recurrence in HR-positive breast cancer has been shown to remain elevated and fairly stable beyond 10 years, even for those with low tumor burden and node-negative disease [38]. Prospective trials showed that for women with HR-positive early breast cancer, 5 years of adjuvant tamoxifen could substantially reduce recurrence rates and improve survival, and this became the standard of care [39]. AIs are considered the standard of care for adjuvant endocrine therapy in most postmenopausal women, as they result in a significantly lower recurrence rate compared with tamoxifen, either as initial adjuvant therapy or sequentially following 2 to 3 years of tamoxifen [40].

Due to the risk for later recurrences with HR-positive breast cancer, more patients and oncologists are considering extended endocrine therapy. This is based on results from the ATLAS (Adjuvant Tamoxifen: Longer Against Shorter) and aTTOM (Adjuvant Tamoxifen–To Offer More?) studies (Table 2), both of which showed that women with HR-positive breast cancer who continued tamoxifen for 10 years had a lower late recurrence rate and a lower breast cancer mortality rate compared with those who stopped at 5 years [41,42]. Furthermore, the NCIC MA.17 trial evaluated extended endocrine therapy in postmenopausal women with 5 years of letrozole following 5 years of tamoxifen. Letrozole was shown to improve both disease-free and distant disease–free survival. The overall survival benefit was limited to patients with node-positive disease [43].

However, extending AI therapy from 5 years to 10 years is not clearly beneficial. In the MA.17R trial, although longer AI therapy resulted in significantly better disease-free survival (95% versus 91%, hazard ratio 0.66; P = 0.01), this was primarily due to a lower incidence of contralateral breast cancer in those taking the AI compared with placebo. The distant recurrence risks were similar and low (4.4% versus 5.5%), and there was no overall survival difference [2]. Also, the NSABP B-42 study, which was presented at the 2016 San Antonio Breast Cancer Symposium, did not meet its predefined endpoint for benefit from extending adjuvant AI therapy with letrozole beyond 5 years [3]. Thus, the absolute benefit from extended endocrine therapy has been modest across these studies. Although endocrine therapy is considered relatively safe and well tolerated, side effects can be significant and even associated with morbidity. Ideally, extended endocrine therapy should be offered to the subset of patients who would benefit the most. Several genomic diagnostic assays, including the EndoPredict test, PAM50, and the Breast Cancer Index (BCI) tests, specifically assess the risk for late recurrence in HR-positive cancers.

Tests for Assessing Risk for Late Recurrence

PAM50

Studies suggest that the ROR score also has value in predicting late recurrences. Analysis of data in patients enrolled in the ABCSG-8 trial showed that ROR could identify patients with endocrine-sensitive disease who are at low risk for late relapse and could be spared from unwanted toxicities of extended endocrine therapies. In 1246 ABCSG-8 patients between years 5 and 15, the PAM50 ROR demonstrated an absolute risk of distant recurrence of 2.4% in the low-risk group, as compared with 17.5% in the high-risk group [44]. Also, a combined analysis of patients from both the ATAC and ABCSG-8 trials demonstrated the utility of ROR in identifying this subgroup of patients with low risk for late relapse [45].

EndoPredict

EndoPredict (EP) is another quantitative RT-PCR–based assay which uses FFPE tissues to calculate a risk score based on 8 cancer-related and 3 reference genes. The score is combined with clinicopathological factors including tumor size and nodal status to make a comprehensive risk score (EPclin). EPclin is used to dichotomize patients into EP low- and EP high-risk groups. EP has been validated in 2 cohorts of patients enrolled in separate randomized studies, ABCSG-6 and ABCSG-8. EP provided prognostic information beyond clinicopathological variables to predict distant recurrence in patients with HR-positive, HER2-negative early breast cancer [46]. More important, EP has been shown to predict early (years 0–5) versus late (> 5 years after diagnosis) recurrences and identify a low-risk subset of patients who would not be expected to benefit from further treatment beyond 5 years of endocrine therapy [47]. Recently, EP and EPclin were compared with the 21-gene (Oncotype DX) recurrence score in a patient population from the TransATAC study. Both EP and EPclin provided more prognostic information compared to the 21-gene recurrence score and identified early and late relapse events [48]. EndoPredict is the first multigene expression assay that could be routinely performed in decentral molecular pathological laboratories with a short turnaround time [49].

Breast Cancer Index

The BCI is a RT-PCR–based gene expression assay that consists of 2 gene expression biomarkers: molecular grade index (MGI) and HOXB13/IL17BR (H/I). The BCI was developed as a prognostic test to assess risk for breast cancer recurrence using a cohort of ER-positive patients (n = 588) treated with adjuvant tamoxifen versus observation from the prospective randomized Stockholm trial [50]. In this blinded retrospective study, H/I and MGI were measured and a continuous risk model (BCI) was developed in the tamoxifen-treated group. More than 50% of the patients in this group were classified as having a low risk of recurrence. The rate of distant recurrence or death in this low-risk group at 10 years was less than 3%. The performance of the BCI model was then tested in the untreated arm of the Stockholm trial. In the untreated arm, BCI classified 53%, 27%, and 20% of patients as low, intermediate, and high risk, respectively. The rate of distant metastasis at 10 years in these risk groups was 8.3% (95% CI 4.7% to 14.4%), 22.9% (95% CI 14.5% to 35.2%), and 28.5% (95% CI 17.9% to 43.6%), respectively, and the rate of breast cancer–specific mortality was 5.1% (95% CI 1.3% to 8.7%), 19.8% (95% CI 10.0% to 28.6%), and 28.8% (95% CI 15.3% to 40.2%) [50].

 

 

The prognostic and predictive values of the BCI have been validated in other large, randomized studies and in patients with both node-negative and node-positive disease [51,52]. The predictive value of the endocrine-response biomarker, the H/I ratio, has been demonstrated in randomized studies. In the MA.17 trial, a high H/I ratio was associated with increased risk for late recurrence in the absence of letrozole. However, extended endocrine therapy with letrozole in patients with high H/I ratios predicted benefit from therapy and decreased the probability of late disease recurrence [53]. BCI was also compared to IHC4 and the 21-gene recurrence score in the TransATAC study and was the only test to show prognostic significance for both early (0–5 years) and late (5–10 year) recurrence [54].

The impact of the BCI results on physicians’ recommendations for extended endocrine therapy was assessed by a prospective study. This study showed that the test result had a significant effect on both physician treatment recommendation and patient satisfaction. BCI testing resulted in a change in physician recommendations for extended endocrine therapy, with an overall decrease in recommendations for extended endocrine therapy from 74% to 54%. Knowledge of the test result also led to improved patient satisfaction and decreased anxiety [55].

Summary

Due to the risk for late recurrence, extended endocrine therapy is being recommended for many patients with HR-positive breast cancers. Multiple genomic assays are being developed to better understand an individual’s risk for late recurrence and the potential for benefit from extended endocrine therapies. However, none of the assays have been validated in prospective randomized studies. Further validation is needed prior to routine use of these assays.

Case Continued

A BCI test is done and the result shows 4.3% BCI low-risk category in years 5–10; low likelihood of benefit from extended endocrine therapy. After discussing the results of the BCI test in the context of no survival benefit from extending AIs beyond 5 years, both the patient and her oncologist feel comfortable with discontinuing endocrine therapy at the end of 5 years.

Conclusion

Reduction in breast cancer mortality is mainly the result of improved systemic treatments. With advances in breast cancer screening tools in recent years, the rate of cancer detection has increased. This has raised concerns regarding overdiagnosis. To prevent unwanted toxicities associated with overtreatment, better treatment decision tools are needed. Several genomic assays are currently available and widely used to provide prognostic and predictive information and aid in decisions regarding appropriate use of adjuvant chemotherapy in HR-positive/HER2-negative early-stage breast cancer. Ongoing studies are refining the cutoffs for these assays and expanding the applicability to node-positive breast cancers. Furthermore, with several studies now showing benefit from the use of extended endocrine therapy, some of these assays may be able to identify the subset of patients who are at increased risk for late recurrence and who might benefit from extended endocrine therapy. Advances in molecular testing has enabled clinicians to offer more personalized treatments to their patients, improve patient’s compliance, and decrease anxiety and conflict associated with management decisions. Although small numbers of patients with HER2-positive and triple negative breast cancers were also included in some of these studies, use of genomic assays in this subset of patients is very limited and currently not recommended.

 

Corresponding author: Kari Braun Wisinski, MD, 1111 Highland Avenue, 6033 Wisconsin Institute for Medical Research, Madison, WI 53705-2275, [email protected].

Financial disclosures: This work was supported by the NCI Cancer Center Support Grant P30 CA014520.

References

1. Welch HG, Prorok PC, O'Malley AJ, Kramer BS. Breast-cancer tumor size, overdiagnosis, and mammography screening effectiveness. N Engl J Med 2016;375:1438–47.

2. Goss PE, Ingle JN, Pritchard KI, et al. Extending aromatase-inhibitor adjuvant therapy to 10 years. N Engl J Med 2016;375:209–19.

3. Mamounas E, Bandos H, Lembersky B. A randomized, double-blinded, placebo-controlled clinical trial of extended adjuvant endocrine therapy with letrozole in postmenopausal women with hormone-receptor-positive breast cancer who have completed previous adjuvant treatment with an aromatase inhibitor. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-05.

4. Tjan-Heijnen VC, Van Hellemond IE, Peer PG, et al: First results from the multicenter phase III DATA study comparing 3 versus 6 years of anastrozole after 2-3 years of tamoxifen in postmenopausal women with hormone receptor-positive early breast cancer. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-03.

5. Blok EJ, Van de Velde CJH, Meershoek-Klein Kranenbarg EM, et al: Optimal duration of extended letrozole treatment after 5 years of adjuvant endocrine therapy. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-04.

6. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 2005;365:1687–717.

7. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature 2000;406:747–52.

8. Coates AS, Winer EP, Goldhirsch A, et al. Tailoring therapies--improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol 2015;26:1533–46.

9. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.

10. Urruticoechea A, Smith IE, Dowsett M. Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 2005;23:7212–20.

11. de Azambuja E, Cardoso F, de Castro G Jr, et al. Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer 2007;96:1504–13.

12. Petrelli F, Viale G, Cabiddu M, Barni S. Prognostic value of different cut-off levels of Ki-67 in breast cancer: a systematic review and meta-analysis of 64,196 patients. Breast Cancer Res Treat 2015;153:477–91.

13. Cheang MC, Chia SK, Voduc D, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 2009;101:736–50.

14. Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and com-parison with the Genomic Health recurrence score in early breast cancer. J Clin Oncol 2011;29:4273–8.

15. Pathmanathan N, Balleine RL. Ki67 and proliferation in breast cancer. J Clin Pathol 2013;66:512–6.

16. Denkert C, Budczies J, von Minckwitz G, et al. Strategies for developing Ki67 as a useful biomarker in breast cancer. Breast 2015; 24 Suppl 2:S67–72.

17. Ma CX, Bose R, Ellis MJ. Prognostic and predictive biomarkers of endocrine responsiveness for estrogen receptor positive breast cancer. Adv Exp Med Biol 2016;882:125–54.

18. Eiermann W, Paepke S, Appfelstaedt J, et al. Preoperative treatment of postmenopausal breast cancer patients with letrozole: a randomized double-blind multicenter study. Ann Oncol 2001;12:1527–32.

19. Smith IE, Dowsett M, Ebbs SR, et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anas-trozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin Oncol 2005;23:5108–16.

20. Ellis MJ, Tao Y, Luo J, et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst 2008;100:1380–8.

21. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 2004;351:2817–26.

22. Fisher B, Jeong JH, Bryant J, et al. Treatment of lymph-node-negative, oestrogen-receptor-positive breast cancer: long-term findings from National Surgical Adjuvant Breast and Bowel Project randomised clinical trials. Lancet 2004;364:858–68.

23. Habel LA, Shak S, Jacobs MK, et al. A population-based study of tumor gene expression and risk of breast cancer death among lymph node-negative patients. Breast Cancer Res 2006;8:R25.

24. Albain KS, Barlow WE, Shak S, et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol 2010;11:55–65.

25. Dowsett M, Cuzick J, Wale C, et al. Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study. J Clin Oncol 2010;28:1829–34.

26. Paik S, Shak S, Tang G, et al. Expression of the 21 genes in the recurrence score assay and tamoxifen clinical benefit in the NSABP study B-14 of node negative, estrogen receptor positive breast cancer. J Clin Oncol 2005;23: suppl:510.

27. Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol2006;24:3726–34.

28. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med 2015;373:2005–14.

29. Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 2009;27:1160–7.

30. Dowsett M, Sestak I, Lopez-Knowles E, et al. Comparison of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant recurrence after endocrine therapy. J Clin Oncol 2013;31:2783–90.

31. Gnant M, Filipits M, Greil R, et al. Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 post-menopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann Oncol 2014;25:339–45.

32. van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002;347:1999–2009.

33. Knauer M, Mook S, Rutgers EJ, et al. The predictive value of the 70-gene signature for adjuvant chemotherapy in early breast cancer. Breast Cancer Res Treat 2010;120:655–61.

34. Cardoso F, van't Veer LJ, Bogaerts J, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med 2016;375:717–29.

35. Sapino A, Roepman P, Linn SC, et al. MammaPrint molecular diagnostics on formalin-fixed, paraffin-embedded tissue. J Mol Diagn 2014;16:190–7.

36. Burstein HJ, Griggs JJ, Prestrud AA, Temin S. American society of clinical oncology clinical practice guideline update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Oncol Pract 2010;6:243–6.

37. Saphner T, Tormey DC, Gray R. Annual hazard rates of recurrence for breast cancer after primary therapy. J Clin Oncol 1996;14:2738–46.

38. Colleoni M, Sun Z, Price KN, et al. Annual hazard rates of recurrence for breast cancer during 24 years of follow-up: results from the International Breast Cancer Study Group Trials I to V. J Clin Oncol 2016;34:927–35.

39. Davies C, Godwin J, Gray R, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 2011;378:771–84.

40. Dowsett M, Forbes JF, Bradley R, et al. Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomised trials. Lancet 2015;386:1341–52.

41. Davies C, Pan H, Godwin J, et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013;381:805–16.

42. Gray R, Rea D, Handley K, et al. aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013;31 (suppl):5.

43. Goss PE, Ingle JN, Martino S, et al. Randomized trial of letrozole following tamoxifen as extended adjuvant therapy in receptor-positive breast cancer: updated findings from NCIC CTG MA.17. J Natl Can-cer Inst 2005;97:1262–71.

44. Filipits M, Nielsen TO, Rudas M, et al. The PAM50 risk-of-recurrence score predicts risk for late distant recurrence after endocrine therapy in postmenopausal women with endocrine-responsive early breast cancer. Clin Cancer Res 2014;20:1298–305.

45. Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant recurrence after 5 years of endocrine treatment: a combined analysis of patients from the Austrian breast and colorectal cancer study group 8 and arimidex, tamoxifen alone or in combination randomized trials using the PAM50 risk of recurrence score. J Clin Oncol 2015;33:916–22.

46. Filipits M, Rudas M, Jakesz R, et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res 2011;17:6012–20.

47. Dubsky P, Brase JC, Jakesz R, et al. The EndoPredict score provides prognostic information on late distant metastases in ER+/HER2- breast cancer patients. Br J Cancer 2013;109:2959–64.

48. Buus R, Sestak I, Kronenwett R, et al. Comparison of EndoPredict and EPclin with Oncotype DX Recurrence Score for prediction of risk of distant recurrence after endocrine therapy. J Natl Cancer Inst 2016;108:djw149.

49. Muller BM, Keil E, Lehmann A, et al. The EndoPredict gene-expression assay in clinical practice - performance and impact on clinical decisions. PLoS One 2013;8:e68252.

50. Jerevall PL, Ma XJ, Li H, et al. Prognostic utility of HOXB13:IL17BR and molecular grade index in early-stage breast cancer patients from the Stockholm trial. Br J Cancer 2011;104:1762–9.

51. Sgroi DC, Chapman JA, Badovinac-Crnjevic T, et al. Assessment of the prognostic and predictive utility of the Breast Cancer Index (BCI): an NCIC CTG MA.14 study. Breast Cancer Res 2016;18:1.

52. Zhang Y, Schnabel CA, Schroeder BE, et al. Breast cancer index identifies early-stage estrogen receptor-positive breast cancer patients at risk for early- and late-distant recurrence. Clin Cancer Res 2013;19:4196–205.

53. Sgroi DC, Carney E, Zarrella E, et al. Prediction of late disease recurrence and extended adjuvant letrozole benefit by the HOXB13/IL17BR biomarker. J Natl Cancer Inst 2013;105:1036–42.

54. Sgroi DC, Sestak I, Cuzick J, et al. Prediction of late distant recurrence in patients with oestrogen-receptor-positive breast cancer: a prospective comparison of the breast-cancer index (BCI) assay, 21-gene recurrence score, and IHC4 in the TransATAC study population. Lancet Oncol 2013;14:1067–76.

55. Sanft T, Aktas B, Schroeder B, et al. Prospective assessment of the decision-making impact of the Breast Cancer Index in recommending extended adjuvant endocrine therapy for patients with early-stage ER-positive breast cancer. Breast Cancer Res Treat 2015;154:533–41.

56. Nielsen TO, Parker JS, Leung S, et al. A comparison of PAM50 Insrinsic Subtyping with Immunohistochemistry and Clinical Prognostic Factors in Tamoxifen-Treated Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res 2010;16:5222–32.

57. Mamounas EP, Jeong JH, Wickerham DL, et al. Benefit from exemestane as extended adjuvant therapy after 5 years of adjuvant tamoxifen: intention-to-treat analysis of the National Surgical Adjuvant Breast And Bowel Project B-33 trial. J Clin Oncol 2008;26:1965–71.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Topics
Sections

From the University of Arizona Cancer Center, Tucson, AZ (Dr. Ehsani), and University of Wisconsin Carbone Cancer Center and School of Medicine and Public Health, Madison, WI (Dr. Wisinski).

 

Abstract

  • Objectives: To describe common genomic tests being used clinically to assess prognosis and guide adjuvant chemotherapy and endocrine therapy decisions for early-stage breast cancer.
  • Methods: Case presentation and review of the literature.
  • Results: Hormone receptor–positive (HR-positive) breast cancers, which express the estrogen and/or progesterone receptor, account for the majority of breast cancers. Endocrine therapy can be highly effective for patients with these HR-positive tumors, and identification of HR-positive breast cancers that do not require the addition of chemotherapy is critical. Clinicopathological features of the breast cancer, including tumor size, nodal involvement, grading, and HR status, are insufficient in predicting the risk for recurrence or the need for chemotherapy. Furthermore, a portion of HR-positive breast cancers have an ongoing risk for late recurrence, and longer durations of endocrine therapy are being used to reduce this risk.
  • Conclusion: There is sufficient evidence for use of genomic testing in early-stage HR-positive breast cancer to aid in chemotherapy recommendations. Further confirmation of genomic assays for prediction of benefit from prolonged endocrine therapy is needed.

Key words: molecular testing; decision aids; HR-positive cancer; recurrence risk; adjuvant chemotherapy; endocrine therapy.

 

 

Despite the increase in incidence of breast cancer, breast cancer mortality has decreased over the past several decades. This is likely due to both early detection and advances in systemic therapy. However, with more widespread use of screening mammography, there are increasing concerns regarding potential overdiagnosis of cancer [1]. One key challenge is that breast cancer is a heterogeneous disease. Thus, improved tools for determining breast cancer biology can help physicians individualize treatments, with low-risk cancers approached with less aggressive treatments, thus preventing unnecessary toxicities, and higher-risk cancers treated appropriately.

Traditionally, adjuvant chemotherapy was recommended based on tumor features such as stage (tumor size, regional nodal involvement), grade, expression of hormone receptors (estrogen receptor [ER] and progesterone receptor [PR]) and human epidermal growth factor receptor-2 (HER2), and patient features (age, menopausal status). However, this approach is not accurate enough to guide individualized treatment recommendations, which are based on the risk for recurrence and the reduction in this risk that can be achieved with various systemic treatments. In particular, there are individuals with low-risk HR-positive, HER2-negative breast cancers who could be spared the toxicities of cytotoxic chemotherapies without compromising the prognosis.

Beyond chemotherapy, endocrine therapies also have risks, especially when given for extended durations. Recently, extended endocrine therapy has been shown to prevent late recurrences of HR-positive breast cancers. In the MA.17R study, extended endocrine therapy with letrozole for a total of 10 years (beyond 5 years of an aromatase inhibitor [AI]) decreased the risk for breast cancer recurrence or the occurrence of contralateral breast cancer by 34% [2]. However, the overall survival was similar between the 2 groups and the results were not confirmed in other studies [3–5]. Identifying the subgroup of patients who benefit from this extended AI therapy is important in the era of personalized medicine. Several tumor genomic assays have been developed to provide additional prognostic and predictive information with the goal of individualizing adjuvant therapies for breast cancer. Although assays are also being evaluated in HER2-positive and triple negative breast cancer, this review will focus on HR-positive, HER2-negative breast cancer.

Case Study

Initial Presentation

A 54-year-old postmenopausal woman with no significant past medical history presents with an abnormal screening mammogram, which shows a focal asymmetry in the 10 o’clock position at middle depth of the left breast. Further work-up with a diagnostic mammogram and ultrasound of the left breast shows a suspicious hypoechoic solid mass with irregular margins measuring 17 mm. The patient undergoes an ultrasound-guided core needle biopsy of the suspicious mass, the results of which are consistent with an invasive ductal carcinoma, Nottingham grade 2, ER strongly positive (95%), PR weakly positive (5%), HER2 negative, and Ki-67 of 15%. She undergoes a left partial mastectomy and sentinel lymph node biopsy, with final pathology demonstrating a single focus of invasive ductal carcinoma, measuring 2.2 cm in greatest dimension with no evidence of lymphovascular invasion. Margins are clear and 2 sentinel lymph nodes are negative for metastatic disease (final pathologic stage IIA, pT2 pN0 cM0). She is referred to medical oncology to discuss adjuvant systemic therapy.

  • Can additional testing be used to determine prognosis and guide systemic therapy rec-ommendations for early-stage HR-positive/HER2-negative breast cancer?

After a diagnosis of early-stage breast cancer, the key clinical question faced by the patient and medical oncologist is: what is the individual’s risk for a metastatic breast cancer recurrence and thus the risk for death due to breast cancer? Once the risk for recurrence is established, systemic adjuvant chemotherapy, endocrine therapy, and/or HER2-directed therapy are considered based on the receptor status (ER/PR and HER2) to reduce this risk. Hormone receptor (HR)–positive, HER2-negative breast cancer is the most common type of breast cancer. Although adjuvant endocrine therapy has significantly reduced the risk for recurrence and improved survival for HR-positive breast cancer [6], the role of adjuvant chemotherapy for this subset of breast cancer remains unclear. Prior to genomic testing, the recommendation for adjuvant chemotherapy for HR-positive/HER2-negative tumors was primarily based on patient age and tumor stage and grade. However, chemotherapy overtreatment remained a concern given the potential short- and long-term risks of chemotherapy. Further studies into HR-positive/HER2-negative tumors have shown that these tumors can be divided into 2 main subtypes, luminal A and luminal B [7]. These subtypes represent unique biology and differ in terms of prognosis and response to endocrine therapy and chemotherapy. Luminal A tumors are strongly endocrine responsive and have a good prognosis, while luminal B tumors are less endocrine responsive and are associated with a poorer prognosis; the addition of adjuvant chemotherapy is often considered for luminal B tumors [8]. Several tests, including tumor genomic assays, are now available to help with delineating the tumor subtype and aid in decision-making regarding adjuvant chemotherapy for HR-positive/HER2-negative breast cancers.

Tests for Guiding Adjuvant Chemotherapy Decisions

Ki-67 Assays, Including IHC4 and PEPI

Chronic proliferation is a hallmark of cancer cells [9]. Ki-67, a nuclear nonhistone protein whose expression varies in intensity throughout the cell cycle, has been used as a measurement of tumor cell proliferation [10]. Two large meta-analyses have demonstrated that high Ki-67 expression in breast tumors is independently associated with worse disease-free and overall survival rates [11,12]. Ki-67 expression has also been used to classify HR-positive tumors as luminal A or B. After classifying tumor subtypes based on intrinsic gene expression profiling, Cheang et al determined that a Ki-67 cut point of 13.25% differentiated luminal A and B tumors [13]. However, the ideal cut point for Ki-67 remains unclear, as the sensitivity and specificity in this study was 77% and 78%, respectively. Others have combined Ki-67 with standard ER, PR, and HER2 testing. This IHC4 score, which weighs each of these variables, was validated in postmenopausal patients from the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial who had ER-positive tumors and did not receive chemotherapy [14]. The prognostic information from the IHC4 was similar to that seen with the 21-gene recurrence score (Oncotype DX), which is discussed later in this article. The key challenge with Ki-67 testing currently is the lack of a validated test methodology, and intraobserver variability in interpreting the Ki-67 results [15]. Recent series have suggested that Ki-67 be considered as a continuous marker rather than a set cut point [16]. These issues continue to impact the clinical utility of Ki-67 for decision making for adjuvant chemotherapy.

 

 

Ki-67 and the preoperative endocrine prognostic index (PEPI) score have been explored in the neoadjuvant setting to separate postmenopausal women with endocrine-sensitive versus intrinsically resistant disease and identify patients at risk for recurrent disease [17]. The on-treatment levels of Ki-67 in response to endocrine therapy have been shown to be more prognostic than baseline values, and a decrease in Ki-67 as early as 2 weeks after initiation of neoadjuvant endocrine therapy is associated with endocrine-sensitive tumors and improved outcome. The PEPI score was developed through retrospective analysis of the P024 trial [18] to evaluate the relationship between post-neoadjuvant endocrine therapy tumor characteristics and risk for early relapse. This was subsequently validated in an independent data set from the IMPACT trial [19]. Patients with low pathological stage (0 or 1) and a favorable biomarker profile (PEPI score 0) at surgery had the best prognosis in the absence of chemotherapy. On the other hand, higher pathological stage at surgery and a poor biomarker profile with loss of ER positivity or persistently elevated Ki-67 (PEPI score of 3) identified de novo endocrine-resistant tumors which are at higher risk for early relapse [20]. The ongoing Alliance A011106 ALTERNATE trial (ALTernate approaches for clinical stage II or III Estrogen Receptor positive breast cancer NeoAdjuvant TrEatment in postmenopausal women, NCT01953588) is a phase 3 study to prospectively test this hypothesis.

21-Gene Recurrence Score (Oncotype DX Assay)

The 21-gene Oncotype DX assay is conducted on paraffin-embedded tumor tissue and measures the expression of 16 cancer-related genes and 5 reference genes using quantitative polymerase chain reaction. The genes included in this assay are mainly related to proliferation (including Ki-67), invasion, and HER2 or estrogen signaling [21]. Originally, the 21-gene recurrence score assay was analyzed as a prognostic biomarker tool in a prospective-retrospective biomarker substudy of the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14 clinical trial in which patients with node-negative, ER-positive tumors were randomly assigned to receive tamoxifen or placebo without chemotherapy [22]. Using the standard reported values of low risk (< 18), intermediate risk (18–30), or high risk (≥ 31) for recurrence, among the tamoxifen-treated patients, cancers with a high-risk recurrence score had a significantly worse rate of distant recurrence and overall survival [21]. Inferior breast cancer survival with a high recurrence score was also confirmed in other series of endocrine-treated patients with node-negative and node-positive disease [23–25].

The predictive utility of the 21-gene recurrence score for endocrine therapy has also been evaluated. A comparison of the placebo- and tamoxifen-treated patients from the NSABP B-14 trial demonstrated that the 21-gene recurrence score predicted benefit from tamoxifen in cancers with low- or intermediate-risk recurrence scores [26]. However, there was no benefit from the use of tamoxifen over placebo in cancers with high-risk recurrence scores. To date, this intriguing data has not been prospectively confirmed, and thus the 21-gene recurrence score is not used to avoid endocrine therapy.

The 21-gene recurrence score is primarily used by oncologists to aid in decision-making regarding adjuvant chemotherapy in patients with node-negative and node-positive (with up to 3 positive lymph nodes), HR-positive/HER2-negative breast cancers. The predictive utility of the 21-gene recurrence score for adjuvant chemotherapy was initially tested using tumor samples from the NSABP B-20 study. This study initially compared adjuvant tamoxifen alone with tamoxifen plus chemotherapy in patients with node-negative, HR-positive tumors. The prospective-retrospective biomarker analysis showed that the patients with high-risk 21-gene recurrence scores benefited from the addition of chemotherapy, whereas those with low- or intermediate-risk did not have an improved freedom from distant recurrence with chemotherapy [27]. Similarly, an analysis from the prospective phase 3 Southwest Oncology Group (SWOG) 8814 trial comparing tamoxifen to tamoxifen with chemotherapy showed that for node-positive tumors, chemotherapy benefit was only seen in those with high 21-gene recurrence scores [24].

Prospective studies are now starting to report results regarding the predictive role of the 21-gene recurrence score. The TAILORx (Trial Assigning Individualized Options for Treatment) trial includes women with node-negative, HR-positive and HER2-negative tumors measuring 0.6 to 5 cm. All patients were treated with standard of care endocrine therapy for at least 5 years. Chemotherapy was determined based on the 21-gene recurrence score results on the primary tumor. The 21-gene recurrence score cutoffs were changed to low (0–10), intermediate (11–25), and high (≥ 26). Patients with scores of 26 or higher were treated with chemotherapy, and those with intermediate scores were randomly assigned to hemotherapy or no chemotherapy; results from this cohort are still pending. However, excellent breast cancer outcomes with endocrine therapy alone were reported from the 1626 (15.9% of total cohort) prospectively followed patients with low-recurrence score tumors. The 5-year invasive disease-free survival was 93.8%, with overall survival of 98% [28]. Given that 5 years is appropriate follow-up to see any chemotherapy benefit, this data supports the recommendation for no chemotherapy in this cohort of patients with very low 21-gene recurrence scores.

The RxPONDER (Rx for Positive Node, Endocrine Responsive Breast Cancer) trial is evaluating women with 1 to 3 node-positive, HR-positive, HER2-negative tumors. In this trial, patients with 21-gene recurrence scores of 0 to 25 were assigned to adjuvant chemotherapy or none. Those with scores of 26 or higher were assigned to chemotherapy. All patients received standard adjuvant endocrine therapy. This study has completed accrual and results are pending. Of note, TAILORx and RxPONDER did not investigate the potential lack of benefit of endocrine therapy in cancers with high recurrence scores. Furthermore, despite data suggesting that chemotherapy may not even benefit women with 4 or more nodes involved but who have a low recurrence score [24], due to the lack of prospective data in this cohort and the quite high risk for distant recurrence, chemotherapy continues to be the standard of care for these patients.

PAM50 (Breast Cancer Prognostic Gene Signature)

Using microarray and quantitative reverse transcriptase PCR (RT-PCR) on formalin-fixed paraffin-embedded (FFPE) tissues, the Breast Cancer Prognostic Gene Signature (PAM50) assay was initially developed to identify intrinsic breast cancer subtypes, including luminal A, luminal B, HER2-enriched, and basal-like [7,29]. Based on the prediction analysis of microarray (PAM) method, the assay measures the expression levels of 50 genes, provides a risk category (low, intermediate, and high), and generates a numerical risk of recurrence score (ROR). The intrinsic subtype and ROR have been shown to add significant prognostic value to the clinicopathological characteristics of tumors. Clinical validity of PAM50 was evaluated in postmenopausal women with HR-positive, early-stage breast cancer treated in the prospective ATAC and ABCSG-8 (Austrian Breast and Colorectal Cancer Study Group 8) trials [30,31]. In 1017 patients with ER-positive breast cancer treated with anastrozole or tamoxifen in the ATAC trial, ROR added significant prognostic information beyond the clinical treatment score (integrated prognostic information from nodal status, tumor size, histopathologic grade, age, and anastrozole or tamoxifen treatment) in all patients. Also, compared with the 21-gene recurrence score, ROR provided more prognostic information in ER-positive, node-negative disease and better differentiation of intermediate- and higher-risk groups. Fewer patients were categorized as intermediate risk by ROR and more as high risk, which could reduce the uncertainty in the estimate of clinical benefit from chemotherapy [30]. The clinical utility of PAM50 as a prognostic model was also validated in 1478 postmenopausal women with ER-positive early-stage breast cancer enrolled in the ABCSG-8 trial. In this study, ROR assigned 47% of patients with node-negative disease to the low-risk category. In this low-risk group, the 10-year metastasis risk was less than 3.5 %, indicating lack of benefit from additional chemotherapy [31]. A key limitation of the PAM50 is the lack of any prospective studies with this assay.

PAM50 has been designed to be carried out in any qualified pathology laboratory. Moreover, the ROR score provides additional prognostic information about risk of late recurrence, which will be discussed in the next section.

 

 

70-Gene Breast Cancer Recurrence Assay (MammaPrint)

MammaPrint is a 70-gene assay that was initially developed using an unsupervised, hierarchical clustering algorithm on whole-genome expression arrays with early-stage breast cancer. Among 295 consecutive patients who had MammaPrint testing, those classified with a good-prognosis tumor signature (n = 115) had an excellent 10-year survival rate (94.5%) compared to those with a poor-prognosis signature (54.5%), and the signature remained prognostic upon multivariate analysis [32]. Subsequently, a pooled analysis comparing outcomes by MammaPrint score in patients with node-negative or 1 to 3 node-positive breast cancers treated as per discretion of their medical team with either adjuvant chemotherapy plus endocrine therapy or endocrine therapy alone reported that only those patients with a high-risk score benefited from chemotherapy [33]. Recently, a prospective phase 3 study (MINDACT [Microarray In Node negative Disease may Avoid ChemoTherapy]) evaluating the utility of MammaPrint for adjuvant chemotherapy decision-making reported results [34]. In this study, 6693 women with early-stage breast cancer were assessed by clinical risk and genomic risk using MammaPrint. Those with low clinical and genomic risk did not receive chemotherapy, while those with high clinical and genomic risk all received chemotherapy. The primary goal of the study was to assess whether forgoing chemotherapy would be associated with a low rate of recurrence in those patients with a low-risk prognostic MammaPrint signature but high clinical risk. A total of 1550 patients (23.2%) were in the discordant group, and the majority of these patients had HR-positive disease (98.1%). Without chemotherapy, the rate of survival without distant metastasis at 5 years in this group was 94.7% (95% confidence interval [CI] 92.5% to 96.2%), which met the primary endpoint. Of note, initially, MammaPrint was only available for fresh tissue analysis, but recent advances in RNA processing now allow for this analysis on FFPE tissue [35].

Summary

These genomic and biomarker assays can identify different subsets of HR-positive breast cancers, including those patients who have tumors with an excellent prognosis with endocrine therapies alone. Thus, we now have the tools to help avoid the toxicities of chemotherapy in many women with early-stage breast cancer. A summary of the genomic tests available is shown in Table 1.

 

 

Case Continued

The patient undergoes 21-gene recurrence score testing, which shows a low recurrence score of 10, estimating the 10-year risk of distant recurrence to be approximately 7% with 5 years of tamoxifen. Chemo-therapy is not recommended. The patient completes adjuvant whole breast radiation therapy, and then, based on data supporting AIs over tamoxifen in postmenopausal women, she is started on anastrozole [36]. She initially experiences mild side effects from treatment, including fatigue, arthralgia, and vaginal dryness, but her symptoms are able to be managed. As she approaches 5 years of adjuvant endocrine therapy with anastrozole, she is struggling with rotator cuff injury and is anxious about recurrence, but has no evidence of recurrent cancer. Her bone density scan in the beginning of her fourth year of therapy shows a decrease in bone mineral density, with the lowest T score of –1.5 at the left femoral neck, consistent with osteopenia. She has been treated with calcium and vitamin D supplements.

  • How long should this patient continue treatment with anastrozole?

The risk for recurrence is highest during the first 5 years after diagnosis for all patients with early breast cancer [37]. Although HR-positive breast cancers have a better prognosis than HR-negative disease, the pattern of recurrence is different between the 2 groups, and it is estimated that approximately half of the recurrences among patients with HR-positive early breast cancer occur after the first 5 years from diagnosis. Annualized hazard of recurrence in HR-positive breast cancer has been shown to remain elevated and fairly stable beyond 10 years, even for those with low tumor burden and node-negative disease [38]. Prospective trials showed that for women with HR-positive early breast cancer, 5 years of adjuvant tamoxifen could substantially reduce recurrence rates and improve survival, and this became the standard of care [39]. AIs are considered the standard of care for adjuvant endocrine therapy in most postmenopausal women, as they result in a significantly lower recurrence rate compared with tamoxifen, either as initial adjuvant therapy or sequentially following 2 to 3 years of tamoxifen [40].

Due to the risk for later recurrences with HR-positive breast cancer, more patients and oncologists are considering extended endocrine therapy. This is based on results from the ATLAS (Adjuvant Tamoxifen: Longer Against Shorter) and aTTOM (Adjuvant Tamoxifen–To Offer More?) studies (Table 2), both of which showed that women with HR-positive breast cancer who continued tamoxifen for 10 years had a lower late recurrence rate and a lower breast cancer mortality rate compared with those who stopped at 5 years [41,42]. Furthermore, the NCIC MA.17 trial evaluated extended endocrine therapy in postmenopausal women with 5 years of letrozole following 5 years of tamoxifen. Letrozole was shown to improve both disease-free and distant disease–free survival. The overall survival benefit was limited to patients with node-positive disease [43].

However, extending AI therapy from 5 years to 10 years is not clearly beneficial. In the MA.17R trial, although longer AI therapy resulted in significantly better disease-free survival (95% versus 91%, hazard ratio 0.66; P = 0.01), this was primarily due to a lower incidence of contralateral breast cancer in those taking the AI compared with placebo. The distant recurrence risks were similar and low (4.4% versus 5.5%), and there was no overall survival difference [2]. Also, the NSABP B-42 study, which was presented at the 2016 San Antonio Breast Cancer Symposium, did not meet its predefined endpoint for benefit from extending adjuvant AI therapy with letrozole beyond 5 years [3]. Thus, the absolute benefit from extended endocrine therapy has been modest across these studies. Although endocrine therapy is considered relatively safe and well tolerated, side effects can be significant and even associated with morbidity. Ideally, extended endocrine therapy should be offered to the subset of patients who would benefit the most. Several genomic diagnostic assays, including the EndoPredict test, PAM50, and the Breast Cancer Index (BCI) tests, specifically assess the risk for late recurrence in HR-positive cancers.

Tests for Assessing Risk for Late Recurrence

PAM50

Studies suggest that the ROR score also has value in predicting late recurrences. Analysis of data in patients enrolled in the ABCSG-8 trial showed that ROR could identify patients with endocrine-sensitive disease who are at low risk for late relapse and could be spared from unwanted toxicities of extended endocrine therapies. In 1246 ABCSG-8 patients between years 5 and 15, the PAM50 ROR demonstrated an absolute risk of distant recurrence of 2.4% in the low-risk group, as compared with 17.5% in the high-risk group [44]. Also, a combined analysis of patients from both the ATAC and ABCSG-8 trials demonstrated the utility of ROR in identifying this subgroup of patients with low risk for late relapse [45].

EndoPredict

EndoPredict (EP) is another quantitative RT-PCR–based assay which uses FFPE tissues to calculate a risk score based on 8 cancer-related and 3 reference genes. The score is combined with clinicopathological factors including tumor size and nodal status to make a comprehensive risk score (EPclin). EPclin is used to dichotomize patients into EP low- and EP high-risk groups. EP has been validated in 2 cohorts of patients enrolled in separate randomized studies, ABCSG-6 and ABCSG-8. EP provided prognostic information beyond clinicopathological variables to predict distant recurrence in patients with HR-positive, HER2-negative early breast cancer [46]. More important, EP has been shown to predict early (years 0–5) versus late (> 5 years after diagnosis) recurrences and identify a low-risk subset of patients who would not be expected to benefit from further treatment beyond 5 years of endocrine therapy [47]. Recently, EP and EPclin were compared with the 21-gene (Oncotype DX) recurrence score in a patient population from the TransATAC study. Both EP and EPclin provided more prognostic information compared to the 21-gene recurrence score and identified early and late relapse events [48]. EndoPredict is the first multigene expression assay that could be routinely performed in decentral molecular pathological laboratories with a short turnaround time [49].

Breast Cancer Index

The BCI is a RT-PCR–based gene expression assay that consists of 2 gene expression biomarkers: molecular grade index (MGI) and HOXB13/IL17BR (H/I). The BCI was developed as a prognostic test to assess risk for breast cancer recurrence using a cohort of ER-positive patients (n = 588) treated with adjuvant tamoxifen versus observation from the prospective randomized Stockholm trial [50]. In this blinded retrospective study, H/I and MGI were measured and a continuous risk model (BCI) was developed in the tamoxifen-treated group. More than 50% of the patients in this group were classified as having a low risk of recurrence. The rate of distant recurrence or death in this low-risk group at 10 years was less than 3%. The performance of the BCI model was then tested in the untreated arm of the Stockholm trial. In the untreated arm, BCI classified 53%, 27%, and 20% of patients as low, intermediate, and high risk, respectively. The rate of distant metastasis at 10 years in these risk groups was 8.3% (95% CI 4.7% to 14.4%), 22.9% (95% CI 14.5% to 35.2%), and 28.5% (95% CI 17.9% to 43.6%), respectively, and the rate of breast cancer–specific mortality was 5.1% (95% CI 1.3% to 8.7%), 19.8% (95% CI 10.0% to 28.6%), and 28.8% (95% CI 15.3% to 40.2%) [50].

 

 

The prognostic and predictive values of the BCI have been validated in other large, randomized studies and in patients with both node-negative and node-positive disease [51,52]. The predictive value of the endocrine-response biomarker, the H/I ratio, has been demonstrated in randomized studies. In the MA.17 trial, a high H/I ratio was associated with increased risk for late recurrence in the absence of letrozole. However, extended endocrine therapy with letrozole in patients with high H/I ratios predicted benefit from therapy and decreased the probability of late disease recurrence [53]. BCI was also compared to IHC4 and the 21-gene recurrence score in the TransATAC study and was the only test to show prognostic significance for both early (0–5 years) and late (5–10 year) recurrence [54].

The impact of the BCI results on physicians’ recommendations for extended endocrine therapy was assessed by a prospective study. This study showed that the test result had a significant effect on both physician treatment recommendation and patient satisfaction. BCI testing resulted in a change in physician recommendations for extended endocrine therapy, with an overall decrease in recommendations for extended endocrine therapy from 74% to 54%. Knowledge of the test result also led to improved patient satisfaction and decreased anxiety [55].

Summary

Due to the risk for late recurrence, extended endocrine therapy is being recommended for many patients with HR-positive breast cancers. Multiple genomic assays are being developed to better understand an individual’s risk for late recurrence and the potential for benefit from extended endocrine therapies. However, none of the assays have been validated in prospective randomized studies. Further validation is needed prior to routine use of these assays.

Case Continued

A BCI test is done and the result shows 4.3% BCI low-risk category in years 5–10; low likelihood of benefit from extended endocrine therapy. After discussing the results of the BCI test in the context of no survival benefit from extending AIs beyond 5 years, both the patient and her oncologist feel comfortable with discontinuing endocrine therapy at the end of 5 years.

Conclusion

Reduction in breast cancer mortality is mainly the result of improved systemic treatments. With advances in breast cancer screening tools in recent years, the rate of cancer detection has increased. This has raised concerns regarding overdiagnosis. To prevent unwanted toxicities associated with overtreatment, better treatment decision tools are needed. Several genomic assays are currently available and widely used to provide prognostic and predictive information and aid in decisions regarding appropriate use of adjuvant chemotherapy in HR-positive/HER2-negative early-stage breast cancer. Ongoing studies are refining the cutoffs for these assays and expanding the applicability to node-positive breast cancers. Furthermore, with several studies now showing benefit from the use of extended endocrine therapy, some of these assays may be able to identify the subset of patients who are at increased risk for late recurrence and who might benefit from extended endocrine therapy. Advances in molecular testing has enabled clinicians to offer more personalized treatments to their patients, improve patient’s compliance, and decrease anxiety and conflict associated with management decisions. Although small numbers of patients with HER2-positive and triple negative breast cancers were also included in some of these studies, use of genomic assays in this subset of patients is very limited and currently not recommended.

 

Corresponding author: Kari Braun Wisinski, MD, 1111 Highland Avenue, 6033 Wisconsin Institute for Medical Research, Madison, WI 53705-2275, [email protected].

Financial disclosures: This work was supported by the NCI Cancer Center Support Grant P30 CA014520.

From the University of Arizona Cancer Center, Tucson, AZ (Dr. Ehsani), and University of Wisconsin Carbone Cancer Center and School of Medicine and Public Health, Madison, WI (Dr. Wisinski).

 

Abstract

  • Objectives: To describe common genomic tests being used clinically to assess prognosis and guide adjuvant chemotherapy and endocrine therapy decisions for early-stage breast cancer.
  • Methods: Case presentation and review of the literature.
  • Results: Hormone receptor–positive (HR-positive) breast cancers, which express the estrogen and/or progesterone receptor, account for the majority of breast cancers. Endocrine therapy can be highly effective for patients with these HR-positive tumors, and identification of HR-positive breast cancers that do not require the addition of chemotherapy is critical. Clinicopathological features of the breast cancer, including tumor size, nodal involvement, grading, and HR status, are insufficient in predicting the risk for recurrence or the need for chemotherapy. Furthermore, a portion of HR-positive breast cancers have an ongoing risk for late recurrence, and longer durations of endocrine therapy are being used to reduce this risk.
  • Conclusion: There is sufficient evidence for use of genomic testing in early-stage HR-positive breast cancer to aid in chemotherapy recommendations. Further confirmation of genomic assays for prediction of benefit from prolonged endocrine therapy is needed.

Key words: molecular testing; decision aids; HR-positive cancer; recurrence risk; adjuvant chemotherapy; endocrine therapy.

 

 

Despite the increase in incidence of breast cancer, breast cancer mortality has decreased over the past several decades. This is likely due to both early detection and advances in systemic therapy. However, with more widespread use of screening mammography, there are increasing concerns regarding potential overdiagnosis of cancer [1]. One key challenge is that breast cancer is a heterogeneous disease. Thus, improved tools for determining breast cancer biology can help physicians individualize treatments, with low-risk cancers approached with less aggressive treatments, thus preventing unnecessary toxicities, and higher-risk cancers treated appropriately.

Traditionally, adjuvant chemotherapy was recommended based on tumor features such as stage (tumor size, regional nodal involvement), grade, expression of hormone receptors (estrogen receptor [ER] and progesterone receptor [PR]) and human epidermal growth factor receptor-2 (HER2), and patient features (age, menopausal status). However, this approach is not accurate enough to guide individualized treatment recommendations, which are based on the risk for recurrence and the reduction in this risk that can be achieved with various systemic treatments. In particular, there are individuals with low-risk HR-positive, HER2-negative breast cancers who could be spared the toxicities of cytotoxic chemotherapies without compromising the prognosis.

Beyond chemotherapy, endocrine therapies also have risks, especially when given for extended durations. Recently, extended endocrine therapy has been shown to prevent late recurrences of HR-positive breast cancers. In the MA.17R study, extended endocrine therapy with letrozole for a total of 10 years (beyond 5 years of an aromatase inhibitor [AI]) decreased the risk for breast cancer recurrence or the occurrence of contralateral breast cancer by 34% [2]. However, the overall survival was similar between the 2 groups and the results were not confirmed in other studies [3–5]. Identifying the subgroup of patients who benefit from this extended AI therapy is important in the era of personalized medicine. Several tumor genomic assays have been developed to provide additional prognostic and predictive information with the goal of individualizing adjuvant therapies for breast cancer. Although assays are also being evaluated in HER2-positive and triple negative breast cancer, this review will focus on HR-positive, HER2-negative breast cancer.

Case Study

Initial Presentation

A 54-year-old postmenopausal woman with no significant past medical history presents with an abnormal screening mammogram, which shows a focal asymmetry in the 10 o’clock position at middle depth of the left breast. Further work-up with a diagnostic mammogram and ultrasound of the left breast shows a suspicious hypoechoic solid mass with irregular margins measuring 17 mm. The patient undergoes an ultrasound-guided core needle biopsy of the suspicious mass, the results of which are consistent with an invasive ductal carcinoma, Nottingham grade 2, ER strongly positive (95%), PR weakly positive (5%), HER2 negative, and Ki-67 of 15%. She undergoes a left partial mastectomy and sentinel lymph node biopsy, with final pathology demonstrating a single focus of invasive ductal carcinoma, measuring 2.2 cm in greatest dimension with no evidence of lymphovascular invasion. Margins are clear and 2 sentinel lymph nodes are negative for metastatic disease (final pathologic stage IIA, pT2 pN0 cM0). She is referred to medical oncology to discuss adjuvant systemic therapy.

  • Can additional testing be used to determine prognosis and guide systemic therapy rec-ommendations for early-stage HR-positive/HER2-negative breast cancer?

After a diagnosis of early-stage breast cancer, the key clinical question faced by the patient and medical oncologist is: what is the individual’s risk for a metastatic breast cancer recurrence and thus the risk for death due to breast cancer? Once the risk for recurrence is established, systemic adjuvant chemotherapy, endocrine therapy, and/or HER2-directed therapy are considered based on the receptor status (ER/PR and HER2) to reduce this risk. Hormone receptor (HR)–positive, HER2-negative breast cancer is the most common type of breast cancer. Although adjuvant endocrine therapy has significantly reduced the risk for recurrence and improved survival for HR-positive breast cancer [6], the role of adjuvant chemotherapy for this subset of breast cancer remains unclear. Prior to genomic testing, the recommendation for adjuvant chemotherapy for HR-positive/HER2-negative tumors was primarily based on patient age and tumor stage and grade. However, chemotherapy overtreatment remained a concern given the potential short- and long-term risks of chemotherapy. Further studies into HR-positive/HER2-negative tumors have shown that these tumors can be divided into 2 main subtypes, luminal A and luminal B [7]. These subtypes represent unique biology and differ in terms of prognosis and response to endocrine therapy and chemotherapy. Luminal A tumors are strongly endocrine responsive and have a good prognosis, while luminal B tumors are less endocrine responsive and are associated with a poorer prognosis; the addition of adjuvant chemotherapy is often considered for luminal B tumors [8]. Several tests, including tumor genomic assays, are now available to help with delineating the tumor subtype and aid in decision-making regarding adjuvant chemotherapy for HR-positive/HER2-negative breast cancers.

Tests for Guiding Adjuvant Chemotherapy Decisions

Ki-67 Assays, Including IHC4 and PEPI

Chronic proliferation is a hallmark of cancer cells [9]. Ki-67, a nuclear nonhistone protein whose expression varies in intensity throughout the cell cycle, has been used as a measurement of tumor cell proliferation [10]. Two large meta-analyses have demonstrated that high Ki-67 expression in breast tumors is independently associated with worse disease-free and overall survival rates [11,12]. Ki-67 expression has also been used to classify HR-positive tumors as luminal A or B. After classifying tumor subtypes based on intrinsic gene expression profiling, Cheang et al determined that a Ki-67 cut point of 13.25% differentiated luminal A and B tumors [13]. However, the ideal cut point for Ki-67 remains unclear, as the sensitivity and specificity in this study was 77% and 78%, respectively. Others have combined Ki-67 with standard ER, PR, and HER2 testing. This IHC4 score, which weighs each of these variables, was validated in postmenopausal patients from the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial who had ER-positive tumors and did not receive chemotherapy [14]. The prognostic information from the IHC4 was similar to that seen with the 21-gene recurrence score (Oncotype DX), which is discussed later in this article. The key challenge with Ki-67 testing currently is the lack of a validated test methodology, and intraobserver variability in interpreting the Ki-67 results [15]. Recent series have suggested that Ki-67 be considered as a continuous marker rather than a set cut point [16]. These issues continue to impact the clinical utility of Ki-67 for decision making for adjuvant chemotherapy.

 

 

Ki-67 and the preoperative endocrine prognostic index (PEPI) score have been explored in the neoadjuvant setting to separate postmenopausal women with endocrine-sensitive versus intrinsically resistant disease and identify patients at risk for recurrent disease [17]. The on-treatment levels of Ki-67 in response to endocrine therapy have been shown to be more prognostic than baseline values, and a decrease in Ki-67 as early as 2 weeks after initiation of neoadjuvant endocrine therapy is associated with endocrine-sensitive tumors and improved outcome. The PEPI score was developed through retrospective analysis of the P024 trial [18] to evaluate the relationship between post-neoadjuvant endocrine therapy tumor characteristics and risk for early relapse. This was subsequently validated in an independent data set from the IMPACT trial [19]. Patients with low pathological stage (0 or 1) and a favorable biomarker profile (PEPI score 0) at surgery had the best prognosis in the absence of chemotherapy. On the other hand, higher pathological stage at surgery and a poor biomarker profile with loss of ER positivity or persistently elevated Ki-67 (PEPI score of 3) identified de novo endocrine-resistant tumors which are at higher risk for early relapse [20]. The ongoing Alliance A011106 ALTERNATE trial (ALTernate approaches for clinical stage II or III Estrogen Receptor positive breast cancer NeoAdjuvant TrEatment in postmenopausal women, NCT01953588) is a phase 3 study to prospectively test this hypothesis.

21-Gene Recurrence Score (Oncotype DX Assay)

The 21-gene Oncotype DX assay is conducted on paraffin-embedded tumor tissue and measures the expression of 16 cancer-related genes and 5 reference genes using quantitative polymerase chain reaction. The genes included in this assay are mainly related to proliferation (including Ki-67), invasion, and HER2 or estrogen signaling [21]. Originally, the 21-gene recurrence score assay was analyzed as a prognostic biomarker tool in a prospective-retrospective biomarker substudy of the National Surgical Adjuvant Breast and Bowel Project (NSABP) B-14 clinical trial in which patients with node-negative, ER-positive tumors were randomly assigned to receive tamoxifen or placebo without chemotherapy [22]. Using the standard reported values of low risk (< 18), intermediate risk (18–30), or high risk (≥ 31) for recurrence, among the tamoxifen-treated patients, cancers with a high-risk recurrence score had a significantly worse rate of distant recurrence and overall survival [21]. Inferior breast cancer survival with a high recurrence score was also confirmed in other series of endocrine-treated patients with node-negative and node-positive disease [23–25].

The predictive utility of the 21-gene recurrence score for endocrine therapy has also been evaluated. A comparison of the placebo- and tamoxifen-treated patients from the NSABP B-14 trial demonstrated that the 21-gene recurrence score predicted benefit from tamoxifen in cancers with low- or intermediate-risk recurrence scores [26]. However, there was no benefit from the use of tamoxifen over placebo in cancers with high-risk recurrence scores. To date, this intriguing data has not been prospectively confirmed, and thus the 21-gene recurrence score is not used to avoid endocrine therapy.

The 21-gene recurrence score is primarily used by oncologists to aid in decision-making regarding adjuvant chemotherapy in patients with node-negative and node-positive (with up to 3 positive lymph nodes), HR-positive/HER2-negative breast cancers. The predictive utility of the 21-gene recurrence score for adjuvant chemotherapy was initially tested using tumor samples from the NSABP B-20 study. This study initially compared adjuvant tamoxifen alone with tamoxifen plus chemotherapy in patients with node-negative, HR-positive tumors. The prospective-retrospective biomarker analysis showed that the patients with high-risk 21-gene recurrence scores benefited from the addition of chemotherapy, whereas those with low- or intermediate-risk did not have an improved freedom from distant recurrence with chemotherapy [27]. Similarly, an analysis from the prospective phase 3 Southwest Oncology Group (SWOG) 8814 trial comparing tamoxifen to tamoxifen with chemotherapy showed that for node-positive tumors, chemotherapy benefit was only seen in those with high 21-gene recurrence scores [24].

Prospective studies are now starting to report results regarding the predictive role of the 21-gene recurrence score. The TAILORx (Trial Assigning Individualized Options for Treatment) trial includes women with node-negative, HR-positive and HER2-negative tumors measuring 0.6 to 5 cm. All patients were treated with standard of care endocrine therapy for at least 5 years. Chemotherapy was determined based on the 21-gene recurrence score results on the primary tumor. The 21-gene recurrence score cutoffs were changed to low (0–10), intermediate (11–25), and high (≥ 26). Patients with scores of 26 or higher were treated with chemotherapy, and those with intermediate scores were randomly assigned to hemotherapy or no chemotherapy; results from this cohort are still pending. However, excellent breast cancer outcomes with endocrine therapy alone were reported from the 1626 (15.9% of total cohort) prospectively followed patients with low-recurrence score tumors. The 5-year invasive disease-free survival was 93.8%, with overall survival of 98% [28]. Given that 5 years is appropriate follow-up to see any chemotherapy benefit, this data supports the recommendation for no chemotherapy in this cohort of patients with very low 21-gene recurrence scores.

The RxPONDER (Rx for Positive Node, Endocrine Responsive Breast Cancer) trial is evaluating women with 1 to 3 node-positive, HR-positive, HER2-negative tumors. In this trial, patients with 21-gene recurrence scores of 0 to 25 were assigned to adjuvant chemotherapy or none. Those with scores of 26 or higher were assigned to chemotherapy. All patients received standard adjuvant endocrine therapy. This study has completed accrual and results are pending. Of note, TAILORx and RxPONDER did not investigate the potential lack of benefit of endocrine therapy in cancers with high recurrence scores. Furthermore, despite data suggesting that chemotherapy may not even benefit women with 4 or more nodes involved but who have a low recurrence score [24], due to the lack of prospective data in this cohort and the quite high risk for distant recurrence, chemotherapy continues to be the standard of care for these patients.

PAM50 (Breast Cancer Prognostic Gene Signature)

Using microarray and quantitative reverse transcriptase PCR (RT-PCR) on formalin-fixed paraffin-embedded (FFPE) tissues, the Breast Cancer Prognostic Gene Signature (PAM50) assay was initially developed to identify intrinsic breast cancer subtypes, including luminal A, luminal B, HER2-enriched, and basal-like [7,29]. Based on the prediction analysis of microarray (PAM) method, the assay measures the expression levels of 50 genes, provides a risk category (low, intermediate, and high), and generates a numerical risk of recurrence score (ROR). The intrinsic subtype and ROR have been shown to add significant prognostic value to the clinicopathological characteristics of tumors. Clinical validity of PAM50 was evaluated in postmenopausal women with HR-positive, early-stage breast cancer treated in the prospective ATAC and ABCSG-8 (Austrian Breast and Colorectal Cancer Study Group 8) trials [30,31]. In 1017 patients with ER-positive breast cancer treated with anastrozole or tamoxifen in the ATAC trial, ROR added significant prognostic information beyond the clinical treatment score (integrated prognostic information from nodal status, tumor size, histopathologic grade, age, and anastrozole or tamoxifen treatment) in all patients. Also, compared with the 21-gene recurrence score, ROR provided more prognostic information in ER-positive, node-negative disease and better differentiation of intermediate- and higher-risk groups. Fewer patients were categorized as intermediate risk by ROR and more as high risk, which could reduce the uncertainty in the estimate of clinical benefit from chemotherapy [30]. The clinical utility of PAM50 as a prognostic model was also validated in 1478 postmenopausal women with ER-positive early-stage breast cancer enrolled in the ABCSG-8 trial. In this study, ROR assigned 47% of patients with node-negative disease to the low-risk category. In this low-risk group, the 10-year metastasis risk was less than 3.5 %, indicating lack of benefit from additional chemotherapy [31]. A key limitation of the PAM50 is the lack of any prospective studies with this assay.

PAM50 has been designed to be carried out in any qualified pathology laboratory. Moreover, the ROR score provides additional prognostic information about risk of late recurrence, which will be discussed in the next section.

 

 

70-Gene Breast Cancer Recurrence Assay (MammaPrint)

MammaPrint is a 70-gene assay that was initially developed using an unsupervised, hierarchical clustering algorithm on whole-genome expression arrays with early-stage breast cancer. Among 295 consecutive patients who had MammaPrint testing, those classified with a good-prognosis tumor signature (n = 115) had an excellent 10-year survival rate (94.5%) compared to those with a poor-prognosis signature (54.5%), and the signature remained prognostic upon multivariate analysis [32]. Subsequently, a pooled analysis comparing outcomes by MammaPrint score in patients with node-negative or 1 to 3 node-positive breast cancers treated as per discretion of their medical team with either adjuvant chemotherapy plus endocrine therapy or endocrine therapy alone reported that only those patients with a high-risk score benefited from chemotherapy [33]. Recently, a prospective phase 3 study (MINDACT [Microarray In Node negative Disease may Avoid ChemoTherapy]) evaluating the utility of MammaPrint for adjuvant chemotherapy decision-making reported results [34]. In this study, 6693 women with early-stage breast cancer were assessed by clinical risk and genomic risk using MammaPrint. Those with low clinical and genomic risk did not receive chemotherapy, while those with high clinical and genomic risk all received chemotherapy. The primary goal of the study was to assess whether forgoing chemotherapy would be associated with a low rate of recurrence in those patients with a low-risk prognostic MammaPrint signature but high clinical risk. A total of 1550 patients (23.2%) were in the discordant group, and the majority of these patients had HR-positive disease (98.1%). Without chemotherapy, the rate of survival without distant metastasis at 5 years in this group was 94.7% (95% confidence interval [CI] 92.5% to 96.2%), which met the primary endpoint. Of note, initially, MammaPrint was only available for fresh tissue analysis, but recent advances in RNA processing now allow for this analysis on FFPE tissue [35].

Summary

These genomic and biomarker assays can identify different subsets of HR-positive breast cancers, including those patients who have tumors with an excellent prognosis with endocrine therapies alone. Thus, we now have the tools to help avoid the toxicities of chemotherapy in many women with early-stage breast cancer. A summary of the genomic tests available is shown in Table 1.

 

 

Case Continued

The patient undergoes 21-gene recurrence score testing, which shows a low recurrence score of 10, estimating the 10-year risk of distant recurrence to be approximately 7% with 5 years of tamoxifen. Chemo-therapy is not recommended. The patient completes adjuvant whole breast radiation therapy, and then, based on data supporting AIs over tamoxifen in postmenopausal women, she is started on anastrozole [36]. She initially experiences mild side effects from treatment, including fatigue, arthralgia, and vaginal dryness, but her symptoms are able to be managed. As she approaches 5 years of adjuvant endocrine therapy with anastrozole, she is struggling with rotator cuff injury and is anxious about recurrence, but has no evidence of recurrent cancer. Her bone density scan in the beginning of her fourth year of therapy shows a decrease in bone mineral density, with the lowest T score of –1.5 at the left femoral neck, consistent with osteopenia. She has been treated with calcium and vitamin D supplements.

  • How long should this patient continue treatment with anastrozole?

The risk for recurrence is highest during the first 5 years after diagnosis for all patients with early breast cancer [37]. Although HR-positive breast cancers have a better prognosis than HR-negative disease, the pattern of recurrence is different between the 2 groups, and it is estimated that approximately half of the recurrences among patients with HR-positive early breast cancer occur after the first 5 years from diagnosis. Annualized hazard of recurrence in HR-positive breast cancer has been shown to remain elevated and fairly stable beyond 10 years, even for those with low tumor burden and node-negative disease [38]. Prospective trials showed that for women with HR-positive early breast cancer, 5 years of adjuvant tamoxifen could substantially reduce recurrence rates and improve survival, and this became the standard of care [39]. AIs are considered the standard of care for adjuvant endocrine therapy in most postmenopausal women, as they result in a significantly lower recurrence rate compared with tamoxifen, either as initial adjuvant therapy or sequentially following 2 to 3 years of tamoxifen [40].

Due to the risk for later recurrences with HR-positive breast cancer, more patients and oncologists are considering extended endocrine therapy. This is based on results from the ATLAS (Adjuvant Tamoxifen: Longer Against Shorter) and aTTOM (Adjuvant Tamoxifen–To Offer More?) studies (Table 2), both of which showed that women with HR-positive breast cancer who continued tamoxifen for 10 years had a lower late recurrence rate and a lower breast cancer mortality rate compared with those who stopped at 5 years [41,42]. Furthermore, the NCIC MA.17 trial evaluated extended endocrine therapy in postmenopausal women with 5 years of letrozole following 5 years of tamoxifen. Letrozole was shown to improve both disease-free and distant disease–free survival. The overall survival benefit was limited to patients with node-positive disease [43].

However, extending AI therapy from 5 years to 10 years is not clearly beneficial. In the MA.17R trial, although longer AI therapy resulted in significantly better disease-free survival (95% versus 91%, hazard ratio 0.66; P = 0.01), this was primarily due to a lower incidence of contralateral breast cancer in those taking the AI compared with placebo. The distant recurrence risks were similar and low (4.4% versus 5.5%), and there was no overall survival difference [2]. Also, the NSABP B-42 study, which was presented at the 2016 San Antonio Breast Cancer Symposium, did not meet its predefined endpoint for benefit from extending adjuvant AI therapy with letrozole beyond 5 years [3]. Thus, the absolute benefit from extended endocrine therapy has been modest across these studies. Although endocrine therapy is considered relatively safe and well tolerated, side effects can be significant and even associated with morbidity. Ideally, extended endocrine therapy should be offered to the subset of patients who would benefit the most. Several genomic diagnostic assays, including the EndoPredict test, PAM50, and the Breast Cancer Index (BCI) tests, specifically assess the risk for late recurrence in HR-positive cancers.

Tests for Assessing Risk for Late Recurrence

PAM50

Studies suggest that the ROR score also has value in predicting late recurrences. Analysis of data in patients enrolled in the ABCSG-8 trial showed that ROR could identify patients with endocrine-sensitive disease who are at low risk for late relapse and could be spared from unwanted toxicities of extended endocrine therapies. In 1246 ABCSG-8 patients between years 5 and 15, the PAM50 ROR demonstrated an absolute risk of distant recurrence of 2.4% in the low-risk group, as compared with 17.5% in the high-risk group [44]. Also, a combined analysis of patients from both the ATAC and ABCSG-8 trials demonstrated the utility of ROR in identifying this subgroup of patients with low risk for late relapse [45].

EndoPredict

EndoPredict (EP) is another quantitative RT-PCR–based assay which uses FFPE tissues to calculate a risk score based on 8 cancer-related and 3 reference genes. The score is combined with clinicopathological factors including tumor size and nodal status to make a comprehensive risk score (EPclin). EPclin is used to dichotomize patients into EP low- and EP high-risk groups. EP has been validated in 2 cohorts of patients enrolled in separate randomized studies, ABCSG-6 and ABCSG-8. EP provided prognostic information beyond clinicopathological variables to predict distant recurrence in patients with HR-positive, HER2-negative early breast cancer [46]. More important, EP has been shown to predict early (years 0–5) versus late (> 5 years after diagnosis) recurrences and identify a low-risk subset of patients who would not be expected to benefit from further treatment beyond 5 years of endocrine therapy [47]. Recently, EP and EPclin were compared with the 21-gene (Oncotype DX) recurrence score in a patient population from the TransATAC study. Both EP and EPclin provided more prognostic information compared to the 21-gene recurrence score and identified early and late relapse events [48]. EndoPredict is the first multigene expression assay that could be routinely performed in decentral molecular pathological laboratories with a short turnaround time [49].

Breast Cancer Index

The BCI is a RT-PCR–based gene expression assay that consists of 2 gene expression biomarkers: molecular grade index (MGI) and HOXB13/IL17BR (H/I). The BCI was developed as a prognostic test to assess risk for breast cancer recurrence using a cohort of ER-positive patients (n = 588) treated with adjuvant tamoxifen versus observation from the prospective randomized Stockholm trial [50]. In this blinded retrospective study, H/I and MGI were measured and a continuous risk model (BCI) was developed in the tamoxifen-treated group. More than 50% of the patients in this group were classified as having a low risk of recurrence. The rate of distant recurrence or death in this low-risk group at 10 years was less than 3%. The performance of the BCI model was then tested in the untreated arm of the Stockholm trial. In the untreated arm, BCI classified 53%, 27%, and 20% of patients as low, intermediate, and high risk, respectively. The rate of distant metastasis at 10 years in these risk groups was 8.3% (95% CI 4.7% to 14.4%), 22.9% (95% CI 14.5% to 35.2%), and 28.5% (95% CI 17.9% to 43.6%), respectively, and the rate of breast cancer–specific mortality was 5.1% (95% CI 1.3% to 8.7%), 19.8% (95% CI 10.0% to 28.6%), and 28.8% (95% CI 15.3% to 40.2%) [50].

 

 

The prognostic and predictive values of the BCI have been validated in other large, randomized studies and in patients with both node-negative and node-positive disease [51,52]. The predictive value of the endocrine-response biomarker, the H/I ratio, has been demonstrated in randomized studies. In the MA.17 trial, a high H/I ratio was associated with increased risk for late recurrence in the absence of letrozole. However, extended endocrine therapy with letrozole in patients with high H/I ratios predicted benefit from therapy and decreased the probability of late disease recurrence [53]. BCI was also compared to IHC4 and the 21-gene recurrence score in the TransATAC study and was the only test to show prognostic significance for both early (0–5 years) and late (5–10 year) recurrence [54].

The impact of the BCI results on physicians’ recommendations for extended endocrine therapy was assessed by a prospective study. This study showed that the test result had a significant effect on both physician treatment recommendation and patient satisfaction. BCI testing resulted in a change in physician recommendations for extended endocrine therapy, with an overall decrease in recommendations for extended endocrine therapy from 74% to 54%. Knowledge of the test result also led to improved patient satisfaction and decreased anxiety [55].

Summary

Due to the risk for late recurrence, extended endocrine therapy is being recommended for many patients with HR-positive breast cancers. Multiple genomic assays are being developed to better understand an individual’s risk for late recurrence and the potential for benefit from extended endocrine therapies. However, none of the assays have been validated in prospective randomized studies. Further validation is needed prior to routine use of these assays.

Case Continued

A BCI test is done and the result shows 4.3% BCI low-risk category in years 5–10; low likelihood of benefit from extended endocrine therapy. After discussing the results of the BCI test in the context of no survival benefit from extending AIs beyond 5 years, both the patient and her oncologist feel comfortable with discontinuing endocrine therapy at the end of 5 years.

Conclusion

Reduction in breast cancer mortality is mainly the result of improved systemic treatments. With advances in breast cancer screening tools in recent years, the rate of cancer detection has increased. This has raised concerns regarding overdiagnosis. To prevent unwanted toxicities associated with overtreatment, better treatment decision tools are needed. Several genomic assays are currently available and widely used to provide prognostic and predictive information and aid in decisions regarding appropriate use of adjuvant chemotherapy in HR-positive/HER2-negative early-stage breast cancer. Ongoing studies are refining the cutoffs for these assays and expanding the applicability to node-positive breast cancers. Furthermore, with several studies now showing benefit from the use of extended endocrine therapy, some of these assays may be able to identify the subset of patients who are at increased risk for late recurrence and who might benefit from extended endocrine therapy. Advances in molecular testing has enabled clinicians to offer more personalized treatments to their patients, improve patient’s compliance, and decrease anxiety and conflict associated with management decisions. Although small numbers of patients with HER2-positive and triple negative breast cancers were also included in some of these studies, use of genomic assays in this subset of patients is very limited and currently not recommended.

 

Corresponding author: Kari Braun Wisinski, MD, 1111 Highland Avenue, 6033 Wisconsin Institute for Medical Research, Madison, WI 53705-2275, [email protected].

Financial disclosures: This work was supported by the NCI Cancer Center Support Grant P30 CA014520.

References

1. Welch HG, Prorok PC, O'Malley AJ, Kramer BS. Breast-cancer tumor size, overdiagnosis, and mammography screening effectiveness. N Engl J Med 2016;375:1438–47.

2. Goss PE, Ingle JN, Pritchard KI, et al. Extending aromatase-inhibitor adjuvant therapy to 10 years. N Engl J Med 2016;375:209–19.

3. Mamounas E, Bandos H, Lembersky B. A randomized, double-blinded, placebo-controlled clinical trial of extended adjuvant endocrine therapy with letrozole in postmenopausal women with hormone-receptor-positive breast cancer who have completed previous adjuvant treatment with an aromatase inhibitor. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-05.

4. Tjan-Heijnen VC, Van Hellemond IE, Peer PG, et al: First results from the multicenter phase III DATA study comparing 3 versus 6 years of anastrozole after 2-3 years of tamoxifen in postmenopausal women with hormone receptor-positive early breast cancer. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-03.

5. Blok EJ, Van de Velde CJH, Meershoek-Klein Kranenbarg EM, et al: Optimal duration of extended letrozole treatment after 5 years of adjuvant endocrine therapy. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-04.

6. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 2005;365:1687–717.

7. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature 2000;406:747–52.

8. Coates AS, Winer EP, Goldhirsch A, et al. Tailoring therapies--improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol 2015;26:1533–46.

9. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.

10. Urruticoechea A, Smith IE, Dowsett M. Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 2005;23:7212–20.

11. de Azambuja E, Cardoso F, de Castro G Jr, et al. Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer 2007;96:1504–13.

12. Petrelli F, Viale G, Cabiddu M, Barni S. Prognostic value of different cut-off levels of Ki-67 in breast cancer: a systematic review and meta-analysis of 64,196 patients. Breast Cancer Res Treat 2015;153:477–91.

13. Cheang MC, Chia SK, Voduc D, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 2009;101:736–50.

14. Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and com-parison with the Genomic Health recurrence score in early breast cancer. J Clin Oncol 2011;29:4273–8.

15. Pathmanathan N, Balleine RL. Ki67 and proliferation in breast cancer. J Clin Pathol 2013;66:512–6.

16. Denkert C, Budczies J, von Minckwitz G, et al. Strategies for developing Ki67 as a useful biomarker in breast cancer. Breast 2015; 24 Suppl 2:S67–72.

17. Ma CX, Bose R, Ellis MJ. Prognostic and predictive biomarkers of endocrine responsiveness for estrogen receptor positive breast cancer. Adv Exp Med Biol 2016;882:125–54.

18. Eiermann W, Paepke S, Appfelstaedt J, et al. Preoperative treatment of postmenopausal breast cancer patients with letrozole: a randomized double-blind multicenter study. Ann Oncol 2001;12:1527–32.

19. Smith IE, Dowsett M, Ebbs SR, et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anas-trozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin Oncol 2005;23:5108–16.

20. Ellis MJ, Tao Y, Luo J, et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst 2008;100:1380–8.

21. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 2004;351:2817–26.

22. Fisher B, Jeong JH, Bryant J, et al. Treatment of lymph-node-negative, oestrogen-receptor-positive breast cancer: long-term findings from National Surgical Adjuvant Breast and Bowel Project randomised clinical trials. Lancet 2004;364:858–68.

23. Habel LA, Shak S, Jacobs MK, et al. A population-based study of tumor gene expression and risk of breast cancer death among lymph node-negative patients. Breast Cancer Res 2006;8:R25.

24. Albain KS, Barlow WE, Shak S, et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol 2010;11:55–65.

25. Dowsett M, Cuzick J, Wale C, et al. Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study. J Clin Oncol 2010;28:1829–34.

26. Paik S, Shak S, Tang G, et al. Expression of the 21 genes in the recurrence score assay and tamoxifen clinical benefit in the NSABP study B-14 of node negative, estrogen receptor positive breast cancer. J Clin Oncol 2005;23: suppl:510.

27. Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol2006;24:3726–34.

28. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med 2015;373:2005–14.

29. Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 2009;27:1160–7.

30. Dowsett M, Sestak I, Lopez-Knowles E, et al. Comparison of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant recurrence after endocrine therapy. J Clin Oncol 2013;31:2783–90.

31. Gnant M, Filipits M, Greil R, et al. Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 post-menopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann Oncol 2014;25:339–45.

32. van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002;347:1999–2009.

33. Knauer M, Mook S, Rutgers EJ, et al. The predictive value of the 70-gene signature for adjuvant chemotherapy in early breast cancer. Breast Cancer Res Treat 2010;120:655–61.

34. Cardoso F, van't Veer LJ, Bogaerts J, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med 2016;375:717–29.

35. Sapino A, Roepman P, Linn SC, et al. MammaPrint molecular diagnostics on formalin-fixed, paraffin-embedded tissue. J Mol Diagn 2014;16:190–7.

36. Burstein HJ, Griggs JJ, Prestrud AA, Temin S. American society of clinical oncology clinical practice guideline update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Oncol Pract 2010;6:243–6.

37. Saphner T, Tormey DC, Gray R. Annual hazard rates of recurrence for breast cancer after primary therapy. J Clin Oncol 1996;14:2738–46.

38. Colleoni M, Sun Z, Price KN, et al. Annual hazard rates of recurrence for breast cancer during 24 years of follow-up: results from the International Breast Cancer Study Group Trials I to V. J Clin Oncol 2016;34:927–35.

39. Davies C, Godwin J, Gray R, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 2011;378:771–84.

40. Dowsett M, Forbes JF, Bradley R, et al. Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomised trials. Lancet 2015;386:1341–52.

41. Davies C, Pan H, Godwin J, et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013;381:805–16.

42. Gray R, Rea D, Handley K, et al. aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013;31 (suppl):5.

43. Goss PE, Ingle JN, Martino S, et al. Randomized trial of letrozole following tamoxifen as extended adjuvant therapy in receptor-positive breast cancer: updated findings from NCIC CTG MA.17. J Natl Can-cer Inst 2005;97:1262–71.

44. Filipits M, Nielsen TO, Rudas M, et al. The PAM50 risk-of-recurrence score predicts risk for late distant recurrence after endocrine therapy in postmenopausal women with endocrine-responsive early breast cancer. Clin Cancer Res 2014;20:1298–305.

45. Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant recurrence after 5 years of endocrine treatment: a combined analysis of patients from the Austrian breast and colorectal cancer study group 8 and arimidex, tamoxifen alone or in combination randomized trials using the PAM50 risk of recurrence score. J Clin Oncol 2015;33:916–22.

46. Filipits M, Rudas M, Jakesz R, et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res 2011;17:6012–20.

47. Dubsky P, Brase JC, Jakesz R, et al. The EndoPredict score provides prognostic information on late distant metastases in ER+/HER2- breast cancer patients. Br J Cancer 2013;109:2959–64.

48. Buus R, Sestak I, Kronenwett R, et al. Comparison of EndoPredict and EPclin with Oncotype DX Recurrence Score for prediction of risk of distant recurrence after endocrine therapy. J Natl Cancer Inst 2016;108:djw149.

49. Muller BM, Keil E, Lehmann A, et al. The EndoPredict gene-expression assay in clinical practice - performance and impact on clinical decisions. PLoS One 2013;8:e68252.

50. Jerevall PL, Ma XJ, Li H, et al. Prognostic utility of HOXB13:IL17BR and molecular grade index in early-stage breast cancer patients from the Stockholm trial. Br J Cancer 2011;104:1762–9.

51. Sgroi DC, Chapman JA, Badovinac-Crnjevic T, et al. Assessment of the prognostic and predictive utility of the Breast Cancer Index (BCI): an NCIC CTG MA.14 study. Breast Cancer Res 2016;18:1.

52. Zhang Y, Schnabel CA, Schroeder BE, et al. Breast cancer index identifies early-stage estrogen receptor-positive breast cancer patients at risk for early- and late-distant recurrence. Clin Cancer Res 2013;19:4196–205.

53. Sgroi DC, Carney E, Zarrella E, et al. Prediction of late disease recurrence and extended adjuvant letrozole benefit by the HOXB13/IL17BR biomarker. J Natl Cancer Inst 2013;105:1036–42.

54. Sgroi DC, Sestak I, Cuzick J, et al. Prediction of late distant recurrence in patients with oestrogen-receptor-positive breast cancer: a prospective comparison of the breast-cancer index (BCI) assay, 21-gene recurrence score, and IHC4 in the TransATAC study population. Lancet Oncol 2013;14:1067–76.

55. Sanft T, Aktas B, Schroeder B, et al. Prospective assessment of the decision-making impact of the Breast Cancer Index in recommending extended adjuvant endocrine therapy for patients with early-stage ER-positive breast cancer. Breast Cancer Res Treat 2015;154:533–41.

56. Nielsen TO, Parker JS, Leung S, et al. A comparison of PAM50 Insrinsic Subtyping with Immunohistochemistry and Clinical Prognostic Factors in Tamoxifen-Treated Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res 2010;16:5222–32.

57. Mamounas EP, Jeong JH, Wickerham DL, et al. Benefit from exemestane as extended adjuvant therapy after 5 years of adjuvant tamoxifen: intention-to-treat analysis of the National Surgical Adjuvant Breast And Bowel Project B-33 trial. J Clin Oncol 2008;26:1965–71.

References

1. Welch HG, Prorok PC, O'Malley AJ, Kramer BS. Breast-cancer tumor size, overdiagnosis, and mammography screening effectiveness. N Engl J Med 2016;375:1438–47.

2. Goss PE, Ingle JN, Pritchard KI, et al. Extending aromatase-inhibitor adjuvant therapy to 10 years. N Engl J Med 2016;375:209–19.

3. Mamounas E, Bandos H, Lembersky B. A randomized, double-blinded, placebo-controlled clinical trial of extended adjuvant endocrine therapy with letrozole in postmenopausal women with hormone-receptor-positive breast cancer who have completed previous adjuvant treatment with an aromatase inhibitor. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-05.

4. Tjan-Heijnen VC, Van Hellemond IE, Peer PG, et al: First results from the multicenter phase III DATA study comparing 3 versus 6 years of anastrozole after 2-3 years of tamoxifen in postmenopausal women with hormone receptor-positive early breast cancer. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-03.

5. Blok EJ, Van de Velde CJH, Meershoek-Klein Kranenbarg EM, et al: Optimal duration of extended letrozole treatment after 5 years of adjuvant endocrine therapy. In: Proceedings from the San Antonio Breast Cancer Symposium; December 6–10, 2016; San Antonio, TX. Abstract S1-04.

6. Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials. Early Breast Cancer Trialists' Collaborative Group. Lancet 2005;365:1687–717.

7. Perou CM, Sorlie T, Eisen MB, et al. Molecular portraits of human breast tumours. Nature 2000;406:747–52.

8. Coates AS, Winer EP, Goldhirsch A, et al. Tailoring therapies--improving the management of early breast cancer: St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2015. Ann Oncol 2015;26:1533–46.

9. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57–70.

10. Urruticoechea A, Smith IE, Dowsett M. Proliferation marker Ki-67 in early breast cancer. J Clin Oncol 2005;23:7212–20.

11. de Azambuja E, Cardoso F, de Castro G Jr, et al. Ki-67 as prognostic marker in early breast cancer: a meta-analysis of published studies involving 12,155 patients. Br J Cancer 2007;96:1504–13.

12. Petrelli F, Viale G, Cabiddu M, Barni S. Prognostic value of different cut-off levels of Ki-67 in breast cancer: a systematic review and meta-analysis of 64,196 patients. Breast Cancer Res Treat 2015;153:477–91.

13. Cheang MC, Chia SK, Voduc D, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst 2009;101:736–50.

14. Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined estrogen receptor, progesterone receptor, Ki-67, and human epidermal growth factor receptor 2 immunohistochemical score and com-parison with the Genomic Health recurrence score in early breast cancer. J Clin Oncol 2011;29:4273–8.

15. Pathmanathan N, Balleine RL. Ki67 and proliferation in breast cancer. J Clin Pathol 2013;66:512–6.

16. Denkert C, Budczies J, von Minckwitz G, et al. Strategies for developing Ki67 as a useful biomarker in breast cancer. Breast 2015; 24 Suppl 2:S67–72.

17. Ma CX, Bose R, Ellis MJ. Prognostic and predictive biomarkers of endocrine responsiveness for estrogen receptor positive breast cancer. Adv Exp Med Biol 2016;882:125–54.

18. Eiermann W, Paepke S, Appfelstaedt J, et al. Preoperative treatment of postmenopausal breast cancer patients with letrozole: a randomized double-blind multicenter study. Ann Oncol 2001;12:1527–32.

19. Smith IE, Dowsett M, Ebbs SR, et al. Neoadjuvant treatment of postmenopausal breast cancer with anastrozole, tamoxifen, or both in combination: the Immediate Preoperative Anas-trozole, Tamoxifen, or Combined with Tamoxifen (IMPACT) multicenter double-blind randomized trial. J Clin Oncol 2005;23:5108–16.

20. Ellis MJ, Tao Y, Luo J, et al. Outcome prediction for estrogen receptor-positive breast cancer based on postneoadjuvant endocrine therapy tumor characteristics. J Natl Cancer Inst 2008;100:1380–8.

21. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med 2004;351:2817–26.

22. Fisher B, Jeong JH, Bryant J, et al. Treatment of lymph-node-negative, oestrogen-receptor-positive breast cancer: long-term findings from National Surgical Adjuvant Breast and Bowel Project randomised clinical trials. Lancet 2004;364:858–68.

23. Habel LA, Shak S, Jacobs MK, et al. A population-based study of tumor gene expression and risk of breast cancer death among lymph node-negative patients. Breast Cancer Res 2006;8:R25.

24. Albain KS, Barlow WE, Shak S, et al. Prognostic and predictive value of the 21-gene recurrence score assay in postmenopausal women with node-positive, oestrogen-receptor-positive breast cancer on chemotherapy: a retrospective analysis of a randomised trial. Lancet Oncol 2010;11:55–65.

25. Dowsett M, Cuzick J, Wale C, et al. Prediction of risk of distant recurrence using the 21-gene recurrence score in node-negative and node-positive postmenopausal patients with breast cancer treated with anastrozole or tamoxifen: a TransATAC study. J Clin Oncol 2010;28:1829–34.

26. Paik S, Shak S, Tang G, et al. Expression of the 21 genes in the recurrence score assay and tamoxifen clinical benefit in the NSABP study B-14 of node negative, estrogen receptor positive breast cancer. J Clin Oncol 2005;23: suppl:510.

27. Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol2006;24:3726–34.

28. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-gene expression assay in breast cancer. N Engl J Med 2015;373:2005–14.

29. Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast cancer based on intrinsic subtypes. J Clin Oncol 2009;27:1160–7.

30. Dowsett M, Sestak I, Lopez-Knowles E, et al. Comparison of PAM50 risk of recurrence score with oncotype DX and IHC4 for predicting risk of distant recurrence after endocrine therapy. J Clin Oncol 2013;31:2783–90.

31. Gnant M, Filipits M, Greil R, et al. Predicting distant recurrence in receptor-positive breast cancer patients with limited clinicopathological risk: using the PAM50 Risk of Recurrence score in 1478 post-menopausal patients of the ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann Oncol 2014;25:339–45.

32. van de Vijver MJ, He YD, van't Veer LJ, et al. A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med 2002;347:1999–2009.

33. Knauer M, Mook S, Rutgers EJ, et al. The predictive value of the 70-gene signature for adjuvant chemotherapy in early breast cancer. Breast Cancer Res Treat 2010;120:655–61.

34. Cardoso F, van't Veer LJ, Bogaerts J, et al. 70-gene signature as an aid to treatment decisions in early-stage breast cancer. N Engl J Med 2016;375:717–29.

35. Sapino A, Roepman P, Linn SC, et al. MammaPrint molecular diagnostics on formalin-fixed, paraffin-embedded tissue. J Mol Diagn 2014;16:190–7.

36. Burstein HJ, Griggs JJ, Prestrud AA, Temin S. American society of clinical oncology clinical practice guideline update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Oncol Pract 2010;6:243–6.

37. Saphner T, Tormey DC, Gray R. Annual hazard rates of recurrence for breast cancer after primary therapy. J Clin Oncol 1996;14:2738–46.

38. Colleoni M, Sun Z, Price KN, et al. Annual hazard rates of recurrence for breast cancer during 24 years of follow-up: results from the International Breast Cancer Study Group Trials I to V. J Clin Oncol 2016;34:927–35.

39. Davies C, Godwin J, Gray R, et al. Relevance of breast cancer hormone receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 2011;378:771–84.

40. Dowsett M, Forbes JF, Bradley R, et al. Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomised trials. Lancet 2015;386:1341–52.

41. Davies C, Pan H, Godwin J, et al. Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years after diagnosis of oestrogen receptor-positive breast cancer: ATLAS, a randomised trial. Lancet 2013;381:805–16.

42. Gray R, Rea D, Handley K, et al. aTTom: Long-term effects of continuing adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953 women with early breast cancer. J Clin Oncol 2013;31 (suppl):5.

43. Goss PE, Ingle JN, Martino S, et al. Randomized trial of letrozole following tamoxifen as extended adjuvant therapy in receptor-positive breast cancer: updated findings from NCIC CTG MA.17. J Natl Can-cer Inst 2005;97:1262–71.

44. Filipits M, Nielsen TO, Rudas M, et al. The PAM50 risk-of-recurrence score predicts risk for late distant recurrence after endocrine therapy in postmenopausal women with endocrine-responsive early breast cancer. Clin Cancer Res 2014;20:1298–305.

45. Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant recurrence after 5 years of endocrine treatment: a combined analysis of patients from the Austrian breast and colorectal cancer study group 8 and arimidex, tamoxifen alone or in combination randomized trials using the PAM50 risk of recurrence score. J Clin Oncol 2015;33:916–22.

46. Filipits M, Rudas M, Jakesz R, et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res 2011;17:6012–20.

47. Dubsky P, Brase JC, Jakesz R, et al. The EndoPredict score provides prognostic information on late distant metastases in ER+/HER2- breast cancer patients. Br J Cancer 2013;109:2959–64.

48. Buus R, Sestak I, Kronenwett R, et al. Comparison of EndoPredict and EPclin with Oncotype DX Recurrence Score for prediction of risk of distant recurrence after endocrine therapy. J Natl Cancer Inst 2016;108:djw149.

49. Muller BM, Keil E, Lehmann A, et al. The EndoPredict gene-expression assay in clinical practice - performance and impact on clinical decisions. PLoS One 2013;8:e68252.

50. Jerevall PL, Ma XJ, Li H, et al. Prognostic utility of HOXB13:IL17BR and molecular grade index in early-stage breast cancer patients from the Stockholm trial. Br J Cancer 2011;104:1762–9.

51. Sgroi DC, Chapman JA, Badovinac-Crnjevic T, et al. Assessment of the prognostic and predictive utility of the Breast Cancer Index (BCI): an NCIC CTG MA.14 study. Breast Cancer Res 2016;18:1.

52. Zhang Y, Schnabel CA, Schroeder BE, et al. Breast cancer index identifies early-stage estrogen receptor-positive breast cancer patients at risk for early- and late-distant recurrence. Clin Cancer Res 2013;19:4196–205.

53. Sgroi DC, Carney E, Zarrella E, et al. Prediction of late disease recurrence and extended adjuvant letrozole benefit by the HOXB13/IL17BR biomarker. J Natl Cancer Inst 2013;105:1036–42.

54. Sgroi DC, Sestak I, Cuzick J, et al. Prediction of late distant recurrence in patients with oestrogen-receptor-positive breast cancer: a prospective comparison of the breast-cancer index (BCI) assay, 21-gene recurrence score, and IHC4 in the TransATAC study population. Lancet Oncol 2013;14:1067–76.

55. Sanft T, Aktas B, Schroeder B, et al. Prospective assessment of the decision-making impact of the Breast Cancer Index in recommending extended adjuvant endocrine therapy for patients with early-stage ER-positive breast cancer. Breast Cancer Res Treat 2015;154:533–41.

56. Nielsen TO, Parker JS, Leung S, et al. A comparison of PAM50 Insrinsic Subtyping with Immunohistochemistry and Clinical Prognostic Factors in Tamoxifen-Treated Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res 2010;16:5222–32.

57. Mamounas EP, Jeong JH, Wickerham DL, et al. Benefit from exemestane as extended adjuvant therapy after 5 years of adjuvant tamoxifen: intention-to-treat analysis of the National Surgical Adjuvant Breast And Bowel Project B-33 trial. J Clin Oncol 2008;26:1965–71.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Publications
Topics
Article Type
Display Headline
Genomic Testing in the Management of Early-Stage Breast Cancer
Display Headline
Genomic Testing in the Management of Early-Stage Breast Cancer
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

Clinical Assessment and Management of Cancer-Related Fatigue

Article Type
Changed
Mon, 04/23/2018 - 14:45
Display Headline
Clinical Assessment and Management of Cancer-Related Fatigue

From the University of Texas MD Anderson Cancer Center, Houston, TX.

 

Abstract

  • Objective: To review the evidence on interventions for managing cancer-related fatigue (CRF) and provide evidence-based guidance on approaches to its management.
  • Methods: Nonsystematic review of the literature.
  • Results: Several theories have been proposed to explain the biology of CRF, but there is no single clear mechanism that can be targeted for therapy. The approach to patients begins with screening for fatigue and assessing its intensity, followed by a thorough history and examination to determine whether any reversible medical conditions are contributing to fatigue. Management of underlying medical comorbidities may help some patients. For patients whose fatigue persists, pharmacologic and nonpharmacologic treatment options are available. Pharmacologic options include psychostimulants, such as methylphenidate and modafinil, and corticosteroids. Nonpharmacologic approaches include exercise, cognitive behavior therapy, yoga, acupuncture, and tai chi.
  • Conclusion: We recommend an individualized approach, often with a combination of the available options. Patients need to be evaluated periodically to assess their fatigue, and since cancer-related fatigue affects survivors, long-term follow-up is needed.

Key words: fatigue; cancer; pro-inflammatory cytokines; nonpharmacologic; psychostimulants.

 

Fatigue is a common distressing effect of cancer [1].It impairs the quality of life of patients undergoing active cancer treatment and of post-treatment survivors. The National Comprehensive Cancer Network (NCCN) defines cancer-related fatigue (CRF) as “a distressing, persistent, subjective sense of physical, emotional and/or cognitive tiredness related to cancer or cancer treatment that is not proportional to recent activity and interferes with usual functioning [2].” Differences between CRF and fatigue reported by individuals without cancer are that CRF is more severe and is not relieved by rest. The prevalence of CRF in cancer patients and survivors is highly variable, ranging between 25% and 99% [2,3]. This variability may be secondary to methods used for screening fatigue and characteristics of the patient groups. In this article, we discuss recognition of CRF and approaches to its management.

 

Pathophysiology

The specific pathophysiologic mechanism underlying CRF is unknown, making targeted treatment a challenge. The multidimensional and subjective nature of CRF has limited the development of research methodologies to explain this condition. However, research has been done in both human and animal models, and several theories have been proposed to explain the pathophysiology of CRF. While pro-inflammatory cytokines remain the central factor playing a significant role at multiple levels in CRF, there may be a complex interplay of more than 1 mechanism contributing to fatigue in an individual patient.

Central Nervous System Disturbances

The basal ganglia are known to influence motivation. Lack of motivation and drive may cause failure to complete physical and mental tasks, even with preserved cognitive ability and motor function. In a study of melanoma patients receiving interferon, increased activity of the basal ganglia and the cerebellum resulted in higher fatigue scores [4]. Higher levels of cytokines may alter blood flow to the cerebellum and lead to the perception of fatigue. In a study of 12 patients and matched controls, when patients were asked to perform sustained elbow flexion until they perceived exhaustion, CRF patients perceived physical exhaustion sooner than controls. In CRF patients in this study, muscle fatigue measured by electromyogram was less than that in healthy individuals at the time of exhaustion, suggesting the role of the central nervous system in CRF [5]. However, there is not enough evidence at this time to support central nervous system disturbance as the main contributing factor to fatigue in cancer patients.

Circadian Rhythm Dysregulation

Circadian rhythm is regulated by the suprachiasmatic nucleus in the hypothalamus through cortisol and melatonin. Sleep disturbances occur with disruption of the circadian rhythm. Tumor-related peptides such as epidermal growth factor or alterations in serotonin and cortisol can influence the suprachiasmatic nucleus and the complex signaling pathways [2]. Positive feedback loops that are activated by cortisol under the influence of cytokines may lead to continuous cytokine production and altered circadian rhythm. Bower et al showed that changes in the cortisol curve influence fatigue in breast cancer survivors [6]. These patients had a late evening peak in cortisol levels, compared with an early morning peak in individuals without cancer.

Inhibition of Hypothalamic–Pituitary–Adrenal Axis

The hypothalamic–pituitary–adrenal (HPA) axis regulates the release of the stress hormone cortisol. One of several hypotheses advanced to explain the effect of serotonin and the HPA axis on CRF suggests that lower serotonin levels cause decreased activation of 5-hydroxytrytophan 1-a (5-HT1-a) receptors in the hypothalamus, leading to decreased activity of the HPA axis [6]. The inhibition of the HPA axis may occur with higher levels of serotonin as well [7]. The 5-HT1-a receptors are also triggered by cytokines. However, the correction of serotonin levels by antidepressants was not shown to improve fatigue [8]. Inhibition of the HPA axis can also lead to lower testosterone, progesterone, or estrogen levels, which may indirectly contribute to fatigue [2].

Skeletal Muscle Effect

Chemotherapy- and tumor-related cachexia have a direct effect on the metabolism of skeletal muscles. This effect may lead to impaired adenosine triphosphate (ATP) generation during muscle contraction [9]. ATP infusion improved muscle strength in one trial, but this was not confirmed in another trial [10,11]. Muscle contraction studies showed no differences in the contractile properties of muscles in fatigued patients who failed earlier in motor tasks and healthy controls [12]. This finding suggests that there could be a failure of skeletal muscle activation by the central nervous system or inhibition of skeletal muscle activity. Cytokines and other neurotransmitters activate vagal efferent nerve fibers, which may lead to reflex inhibition in skeletal muscles [13,14].

Pro-inflammatory Cytokines

Tumors or treatment of them may cause tissue injury, which triggers immune cells to release cytokines, signaling the brain to manifest the symptom fatigue. Inflammatory pathways are influenced by psychological, behavioral, and biological factors, which play a role as risk factors in CRF. Interleukin 6 (IL-6), interleukin-1 receptor antagonist, interleukin-1, and tumor necrosis factor (TNF) have been shown to be elevated in fatigued patients being treated for leukemia and non-Hodgkin lymphoma [15]. IL-6 was also associated with increased fatigue in breast cancer survivors [16]. Similar findings were reported in patients undergoing stem cell transplantation and high-dose chemotherapy [17]. Elevated levels of IL-6 and C-reactive protein were also linked to fatigue in terminally ill cancer patients [18,19]. Furthermore, TNF-α signaling was associated with post-chemotherapy fatigue in breast cancer patients [20]. Leukocytes in breast cancer survivors with fatigue also have increased gene expression of pro-inflammatory cytokines, emphasizing the role of cytokines and inflammation in the pathogenesis of CRF [21].

Other Hypotheses

Several other hypotheses for CRF pathogenesis have been proposed. Activation of latent viruses such as Epstein-Barr virus, lack of social support [22], genetic alterations in immune pathway [23], epigenetic changes [24], accumulation of neurotoxic metabolites and depletion of serotonin by indoleamine 2,3-dioxygenase pathway activation [25], elevated vascular endothelial growth factor levels [26], and hypoxia-related organ dysfunction due to anemia or hemoglobin dysfunction [13] all have been postulated to cause CRF.

Approach to Evaluation and Treatment

The evaluation and treatment of CRF involve 4 steps (Figure). First, patients are screened for fatigue, and in the second step those who have fatigue undergo primary evaluation to assess for potential precipitating causes. The third step is implementation of pharmacologic and non-pharmacologic interventions aimed at alleviating or mitigating fatigue. The fourth step involves re-evaluating patients at periodic intervals to recognize and manage changes in fatigue levels. A multidisciplinary approach involving nursing, physical therapy, social work, and nutrition is critical in managing fatigue in these patients. Education and counselling of patients and involvement of the family are essential for effective management.

Screening

Because patients and health care professionals may be unaware of the treatment options available for CRF, patients may not report fatigue levels to their clinicians, and clinicians may not understand the impact of fatigue on their patients’ quality of life. This leads to underrecognition of the problem. The NCCN recommends screening every cancer patient and post-treatment survivor for fatigue [2]. Patients should be screened at their first visit and then at periodic intervals during and after cancer treatment.

Many scales are available to screen patients for CRF in clinical practice and clinical trials [27]. A single item that asks patients to rate their fatigue on a scale from 0 to 10—in which 0 indicates no fatigue, 1 to 3 indicates mild fatigue, 4 to 6 indicates moderate fatigue, 7 to 9 indicates severe fatigue, and 10 indicates the worst fatigue imaginable—is commonly used to screen for CRF [2]. This scale was adapted from the MD Anderson Symptom Inventory scale and is based on a large nationwide study of cancer patients and survivors [28]. The statistically derived cutoff points in this study are consistent with other scales such as the Brief Fatigue Inventory (BFI) and support the cutoff points (4–6 for moderate and ≥ 7 for severe fatigue) used in various fatigue management guidelines. Furthermore, studies of fatigue in cancer patients have revealed a marked decrease in physical function at levels of 7 or higher, suggesting 7 as an optimal cutoff to identify severe fatigue [29,30]. The Visual Analog Scale is another simple-to-use tool that helps in understanding variations in fatigue throughout the course of the day [31]. The 9-item BFI is often used in clinical trials [29]. It measures the severity of fatigue over the previous 24 hours and has been validated in non-English speaking patients [32].

 

 

CRF affects not only the somatic domain, but also the cognitive, behavioral, and affective domains; therefore, multidimensional scales have been developed for screening. One such tool is the Multidimensional Fatigue Inventory, which measures general, physical, mental, and emotional fatigue domains as well as activity and compares them with those of individuals without cancer [33,34]. The Functional Assessment of Cancer Therapy for Fatigue (FACT-F) is a 13-item questionnaire that has been used to measure CRF in clinical trials as well as in patients receiving various treatments [35].

Although many scales are available, the validity of self-reporting on simple fatigue-rating scales is equal to or better than most complex, lengthy scales [36]. Therefore, unidimensional tools such as the numeric rating scale of 0–10 are commonly used in clinical practice. Mild fatigue (0–3) requires periodic re-evaluation, and moderate and severe fatigue need further evaluation and management [37].

Primary Evaluation

This phase involves a focused history and physical examination and assessment of concurrent symptoms and contributing factors.

 

History and Physical Examination

A detailed history of the patient’s malignancy and type of previous and current treatment may help reveal the cause of fatigue. New-onset fatigue or increase in fatigue may be related to the progression of disease in patients with active malignancy or recurrence of cancer in survivors. These patients may require appropriate testing to assess the underlying disease pattern. A detailed review of systems may help identify some of the contributing factors, which are discussed below. A detailed history regarding medications, including over-the-counter drugs, complementary agents, and past and prior cancer therapies, is helpful as medications can contribute to fatigue. For example, opioids may cause drowsiness and fatigue, which could be improved by dose adjustments. A focused history of fatigue should be obtained in all patients with moderate to severe CRF, which includes the onset, pattern, duration, associated or alleviating factors, and interference with functioning, including activities of daily living [37]. Physical examination should focus on identifying signs of organ dysfunction and features of substance or alcohol abuse which may cause poor sleep and fatigue.

 

Assessment of Contributing Factors

The management of fatigue should be multifactorial, with a comprehensive assessment and treatment plan to address all modifiable fatigue etiologies. The Table lists potential contributing factors to fatigue that should be considered when evaluating patients for CRF; several common conditions are discussed below.

Anemia. Anemia has been correlated with fatigue and quality of life. In a study of 4382 cancer patients receiving chemotherapy, quality-of-life measures using FACT-Anemia scores improved with increased hemoglobin levels [38]. Cancer patients may have anemia due to marrow-suppressing effects of chemotherapy and may also have iron deficiency anemia due to blood loss or autoimmune hemolytic anemia. Therefore, a detailed work-up is required to identify the etiology of anemia. Patients with CRF whose anemia is related to chemotherapy or anemia of chronic disease may benefit from red blood cell transfusion or erythropoiesis-stimulating agents (ESAs). ESAs have been studied extensively; however, their use is controversial because of concerns about thromboembolic side effects leading to adverse outcomes [39]. Also, ESA therapy is not recommended in patients with hematologic malignancies. ESA use should be restricted to patients with chemotherapy-related anemia with hemoglobin below 10 mg/dL and should be discontinued in 6 to 8 weeks if patients do not respond [40]. Other patients may benefit from blood transfusions, which were shown to help in patients with hemoglobin levels between 7.5 and 8.5 g/dL [41].

Sleep disturbance. Poor sleep is common in fatigued cancer survivors [42]. Pro-inflammatory cytokines can disrupt the sleep–wake cycle, causing changes in the HPA axis and neuroendocrine system, which in turn may lead to increasing fatigue. Heckler et al showed that improvement in nighttime sleep leads to improvement of fatigue [43]. Cognitive behavioral therapy and sleep hygiene are important in caring for patients with CRF and poor sleep [44]. Taking a warm bath and/or drinking a glass of milk before bedtime, avoiding caffeinated drinks, and avoiding frequent napping in the day might help. Some patients may require medications such as benzodiazepines or non-benzodiazepine hypnotics (eg, zolpidem) to promote sleep [45]. Melatonin agonists are approved for insomnia in the United states, but not in Europe [46].

Malnutrition. Patients with advanced-stage cancer and with cancers affecting the gastrointestinal tract frequently develop mechanical bowel obstructions, especially at the end of their life, which cause malnutrition. Chemotherapy-related nausea and vomiting may also cause poor oral intake and malnutrition, causing fatigue from muscle weakness. Dehydration and electrolyte imbalances frequently occur as a result of poor oral intake, which might worsen fatigue. In our experience, modifying dietary intake with appropriate caloric exchanges with the help of a nutrition expert has lessened fatigue in some patients. However, terminally ill patients are advised to eat based on their comfort.

Medical comorbidities. Congestive heart failure from anthracycline chemotherapy, hypothyroidism after radiation therapy for head and neck cancers, renal failure, or hepatic failure from chemotherapy may indirectly lead to fatigue. Chemotherapy, opioids, and steroids may cause hypogonadism, which can contribute to fatigue in men [47].

 

 

Assessment of Concurrent Symptoms

Depression is common in cancer patients and coexists with pain, insomnia, fatigue, and anxiety as a symptom cluster [48]. A symptom cluster is defined as 2 or more concurrent and interrelated symptoms occurring together; treating of one of these symptoms without addressing other symptoms is not effective [49]. Therefore, screening for and management of depression, anxiety, and insomnia play an important role in the management of CRF.

Physical symptoms due to the tumor or to therapy—such as pain, dyspnea, nausea, and decreased physical activity—may also contribute to fatigue both directly and indirectly. Patients with lung cancer may have hypoxemia, which can contribute to dyspnea with activity and a perception of fatigue. Optimal management of pain and other physical symptoms in patients with cancer may significantly alleviate fatigue [50].

Management

Management of CRF is individualized based on the patient’s clinical status: active cancer treatment, survivor, or end of life. Education and counselling of patients and their caregivers play an important role in CRF. NCCN guidelines recommend focusing on pain control, distress management, energy conservation, physical activity, nutrition, and sleep hygiene.

 

Nonpharmacologic Interventions

Energy conservation. Energy conservation strategies, in which patients are advised to set priorities and realistic expectations, are highly recommended. Some energy-conserving strategies are to pace oneself, delegate and schedule activities at times of peak energy, postpone nonessential activities, attend to 1 activity at a time, structure daily routines, and maintain a diary to identify their peak energy period and structure activities around that time [51,52]. When patients approach the end of life, increasing fatigue may limit their activity level, and they may depend on caregivers for assistance with activities of daily living, monitoring treatment-related adverse effects, and taking medications, especially elderly patients [53].

Cognitive behavioral therapy. Cognitive behavioral therapy (CBT) has been shown to improve CRF during active treatment, and the benefits persist for a minimum of 2 years after therapy [54]. CBT interventions that optimize sleep quality may improve fatigue [55]. More studies are needed to understand whether referral to a psychologist for formal CBT is required. Randomized clinical trials (RCTs) showed patient fatigue education, learned self-care, coping techniques, and balancing rest and activity benefit patients with CRF [56].

Exercise. Physical activity is highly encouraged in patients with CRF. Exercise increases muscle protein synthesis, improves cytokine response, and decreases the rate of sarcopenia in healthy populations [57]. Studies have shown that exercise helps CRF at all phases of the cancer journey, including radiation therapy, chemotherapy, and survivorship [58]. Some patients may feel less motivated to exercise and may not believe that exercise is possible or could potentially help them. Counselling is needed for such patients.

Older cancer survivors have a decline in their functional capacity and reduced muscle mass. Exercise can improve cardiorespiratory fitness, muscle strength, and body composition [57]. Exercise not only helps at the cellular level but also has psychosocial benefits from improved self-esteem. Therefore, exercise may be recommended not only for younger patients, but also in the older population, who may have comorbidities and less motivation than younger patients.

In a meta-analysis of 56 randomized controlled trials involving 4068 participants, aerobic exercise was found to have beneficial effects on CRF for patients during and after chemotherapy, specifically for patients with solid tumors [59]. In another meta-analysis of breast and prostate cancer survivors, a combination of aerobic exercise with resistance training (3–6 metabolic equivalents, 60%–80% range of motion) was shown to reduce CRF more than aerobic exercise alone [60]. This effect was also shown in an RCT of 160 patients with stage 0 to III breast cancer undergoing radiation therapy [61]. The control group in this study had a group-based non-exercise intervention/relaxation; therefore, the study showed that the effect of resistance training extends beyond the psychosocial benefits of group-based interventions. The intervention comprised 8 progressive machine-based resistance exercises (3 sets, 8–12 repetitions at 60%–80% of 1 repetition maximum) for 60 minutes twice weekly for 12 weeks. However, fatigue assessment questionnaire scores showed benefits in the physical fatigue but not the affective and cognitive components.

The American Society of Clinical Oncology’s guidelines for cancer survivors with fatigue recommends 150 minutes of moderate aerobic exercise (eg, fast walking, cycling, or swimming) per week, with 2 or 3 sessions of strength training per week [62]. An individualized approach to exercise is recommended, as patients’ ability to perform certain types of exercises may be limited by thrombocytopenia, neutropenia, or lytic bone metastasis. Routine use of pre-exercise cardiovascular testing is not recommended but may be considered in high-risk populations, especially patients with risk factors for coronary heart disease and diabetes [63]. Patients withcomorbidities, substantial deconditioning, functional and anatomic defects, or recent major surgery may benefit from referral to physical therapy [37]. Patients near end of life may also benefit from an exercise program, as demonstrated in several studies that showed benefit in CRF and quality of life [64,65]. We recommend that physicians use their best clinical judgement in suggesting the type and intensity of exercise program, as it may not be feasible in some patients.

Mind-body interventions. Mindfulness-based stress reduction (MBSR) has shown promise in breast cancer survivors, who reported immediate improvements in fatigue severity that continued up to 6 weeks after cessation of the training [66]. Prior studies had similar findings, suggesting that MBSR modestly decreases fatigue and sleep disturbances and has a greater effect on the degree to which symptoms interfere with many facets of life [67].

Yoga. A study of a yoga intervention showed a benefit in older cancer survivors [68]. In breast cancer patients undergoing chemotherapy, yoga was shown to benefit not only physical fatigue, but also cognitive fatigue [69]. DVD-based yoga had benefits similar to strengthening exercises in a study of 34 early-stage breast cancer survivors with CRF [70]. More studies are needed in men and patients and survivors of other cancers, as most studies of yoga were conducted in women with breast cancer.

Tai chi/qigong. Like yoga, tai chi and qigong are practices of meditative movement. These practices use postures or movements with a focus on breath and a meditative state to bring about deep states of relaxation. Qigong is a series of simple, repeated practices including body posture/movement, breath practice, and meditation performed in synchrony. Tai chi easy (TCE) is a simplified set of common, repetitive tai chi movements. In a trial, qigong/TCE was compared with sham qigong, which had physical movements but no breathing or meditative practice. Breast cancer survivors in the qigong/TCE group had improved fatigue scores, and the effect persisted for 3 months [71]. Additional research is needed in this area.

Acupuncture. An RCT in breast cancer patients with CRF showed an improvement in the mean general fatigue score (per the Multidimensional Fatigue Inventory) in patients who received acupuncture versus those who did not (−3.11 [95% confidence interval −3.97 to −2.25]; P < 0.001) at 6 weeks. Improvements were seen in both the mental and physical aspects of fatigue [72]. However, Deng et al noted that true acupuncture was no more effective than sham acupuncture for reducing post-chemotherapy chronic fatigue [73]. Presently, there is not sufficient evidence to evaluate the benefits of acupuncture in CRF.

Other modalities. Massage therapy, music therapy, hypnosis, therapeutic touch, biofield therapies, relaxation, and reiki are other therapies for which few studies have been done, with mixed results, and additional research is needed [74]. Currently, there are not sufficient data to recommend any of these modalities.

Pharmacologic Interventions

Psychostimulants. Methylphenidate and modafinil are psychostimulants or wakefulness-promoting agents. Pilot studies showed benefit from methylphenidate and modafinil in CRF [75–77], but RCTs have yielded mixed results. Therefore, in patients with severe fatigue during cancer therapy, the initial management strategy involves evaluation and treatment of medical conditions such as anemia and a trial of non-pharmacological strategies as discussed above. If symptoms persist, then a therapeutic trial of a psychostimulant may be considered per NCCN guidelines for patients undergoing active cancer treatment [37].

Methylphenidate directly stimulates adrenergic receptors and indirectly releases dopamine and norepinephrine from presynaptic terminals, which may explain why the drug benefits patients receiving opioid-induced sedation. It is a commonly studied psychostimulant, though its mechanism of action in CRF is unclear. RCTs of methylphenidate have resulted in a wide range of findings due to the heterogeneity of study populations and variations in the dosage of methylphenidate. A meta-analysis of 7 studies indicates that methylphenidate benefitted the subgroup of patients with CRF [78]. Likewise, in an analysis of 5 RCTs, Minton et al showed a benefit of psychostimulants in fatigue compared with placebo [79]. However, another study of methylphenidate in patients with CRF showed a benefit only in patients with severe fatigue or advanced disease [80]. Methylphenidate was found to benefit cancer patients receiving opioid-induced sedation, as methylphenidate promotes wakefulness, though fatigue was not studied specifically [81]. In a trial with 30 hospice patients in which the methylphenidate dose was titrated based on response and adverse effects, Kerr at al found that the drug improved fatigue in a dose-dependent manner [82]. However, a study in patients with CRF at the University of Texas MD Anderson Cancer Center found no significant difference in BFI scores between patients receiving methylphenidate and those receiving placebo at the end of 2 weeks of treatment [83]. Also, other RCTs in patients undergoing adjuvant chemotherapy for breast cancer [84] and patients receiving radiation therapy for brain tumors [85] failed to demonstrate the efficacy of methylphenidate in CRF. It should be used cautiously after ruling out other causes of fatigue. The drug is overall well tolerated and side effects include headache and nausea.

Modafinil is a non-amphetamine psychostimulant that has been approved for the treatment of narcolepsy. In a trial studying the effect of modafinil on patients receiving docetaxel-based chemotherapy for metastatic breast or prostate cancer, there was a modest but not statistically significant improvement in fatigue scores on the MD Anderson Symptom Inventory compared with placebo. Nausea and vomiting were higher in the modafinil arm than in the placebo arm [86]. Similarly, modafinil was not superior to placebo for CRF in 208 patients with non-squamous cell lung cancer not undergoing chemotherapy or radiation [87]. A placebo effect was also noted in patients with multiple myeloma [88] and patients with primary brain tumors [89]. In a phase 3, multicenter, randomized, placebo-controlled, double-blind clinical trial of modafinil for CRF in 867 patients undergoing chemotherapy, there was a reduction in fatigue only for patients with severe baseline fatigue, with no significant effect on mild to moderate fatigue [90]. In another recent study, modafinil was shown to reduce depressive symptoms only in patients with severe fatigue (BFI item 3 score ≥ 7) [91]. This finding is consistent with previous studies showing benefit in patients with high baseline fatigue, but additional RCTs are needed to provide clarity. NCCN guidelines do not recommend the use of modafinil to treat CRF [37].

Other pharmacologic interventions. Corticosteroids are often used for symptom control in cancer patients. These drugs have anti-inflammatory effects through their modulation of pro-inflammatory cytokines [92]. In a RCT evaluating the efficacy of corticosteroids, patients receiving dexamethasone (4 mg twice daily) experienced significant improvement in their FACT-F scores compared with patients receiving placebo [93]. A similar benefit in fatigue was demonstrated in another study of methylprednisolone (32 mg daily) versus placebo [94]. Despite the benefits of steroids, their adverse effects, such as mood swings, gastritis, hyperglycemia, and immune suppression, limit their long-term use. Therefore, the use of steroids should be restricted to terminally ill fatigued patients with other symptoms such as anorexia, brain metastasis, or pain related to bone metastasis [37].

Testosterone replacement has been shown to diminish fatigue in non-cancer patients. Many men with advanced cancer have hypogonadism leading to low serum testosterone, which may cause fatigue. In a small trial in which cancer patients with hypogonadism received intramuscular testosterone every 14 days or placebo, the group receiving testosterone showed improvement in FACT-F scores, but there was no significant difference in FACT-F scores between the 2 groups [95].

Antidepressants have failed to demonstrate benefit in CRF without depression [8]. However, if a patient has both fatigue and depression, antidepressants may help [96]. A selective serotonin receptor inhibitor is recommended as a first-line antidepressant [97]. Patients with cancer are often receiving multiple medications, and medication interactions should be considered to prevent adverse events such as serotonin syndrome.

 

 

Complementary and Alternative Supplements

Studies using vitamin supplementation have been inconclusive in patients with CRF [74]. The use of other dietary supplements has yielded mixed results, and coenzyme Q has shown no benefit for patients with CRF [98].

The benefit of ginseng was studied in a RCT involving 364 patients with CRF. There was an improvement in Multidimensional Fatigue Symptom Inventory-short form (MFSI-SF) scores at 8 weeks in patients receiving 2000 mg of Wisconsin ginseng compared with patients receiving placebo [99]. Patients on active treatment had greater improvement as compared to the post-treatment group in this trial. In another study of high-dose panax ginseng (ginseng root) at 800 mg daily for 29 days, patients had improvement of CRF as well as overall quality of life, appetite, and sleep at night. It was also well tolerated with few adverse effects [100]. Interaction with warfarin, calcium channel blockers, antiplatelet agents, thrombolytic agents, imatinib, and other agents may occur; therefore, ginseng must be used with careful monitoring in selected patients. There is not enough evidence at this time to support the routine use of ginseng in CRF.

The seed extract of the guarana plant (Paullinia cupana) traditionally has been used as a stimulant. An improvement in fatigue scores was seen with the use of oral guarana (100 mg daily) at the end of 21 days in breast cancer patients receiving chemotherapy [101]. Further studies are needed for these results to be generalized and to understand the adverse effects and interaction profile of guarana.

 

Re-evaluation

Patients who have completed cancer treatment must be monitored for fatigue over the long term, as fatigue may exist beyond the period of active treatment. Many studies have shown fatigue in breast cancer survivors, and fatigue has been demonstrated in survivors of colorectal, lung, and prostate cancers as well as myeloproliferative neoplasms [28]. Therefore, it is important to screen patients for fatigue during follow-up visits. There are currently no studies evaluating the long-term treatment of fatigue. In our experience, the timing of follow-up depends on the level of fatigue and interventions prescribed. Once fatigue is stabilized to a level with which the patient is able to cope, the time interval for follow up may be lengthened. Annual visits may suffice in patients with mild fatigue. Follow-up of patients with moderate to severe fatigue depends on the level of fatigue, the ability to cope, choice of treatment, and presence of contributing factors.

Conclusion

CRF is a complex condition that places a significant burden on patients and caregivers, resulting in emotional distress, poor functioning, and suffering. Fatigue can occur before, during, and long after cancer treatment. The approach to CRF begins with screening for and educating patients and their caregivers about the symptoms. Many screening scales are available that may be used to follow patients’ progress over time. The evaluation and management of contributing conditions may help improve fatigue. If the fatigue persists, an individualized approach with a combination of nonpharmacologic and pharmacologic approaches should be considered. More research is needed to understand brain signaling pathways, cytokine changes, and genomic changes in cancer patients with fatigue. Though many hypotheses have been proposed, to date there is no biological marker to assess this condition. Biomarker research needs to be advanced to help to identify patients at risk for fatigue. As cytokines have a major role in CRF, targeted therapy to block cytokine pathways may also be explored in the future.

 

Acknowledgment: Bryan Tutt provided editorial assistance.

Corresponding author: Carmelita P. Escalante, MD, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX 77030, [email protected].

Financial disclosures: None.

References

1. Scherber RM, Kosiorek HE, Senyak Z, et al. Comprehensively understanding fatigue in patients with myeloproliferative neoplasms. Cancer 2016;122:477–85.

2. Neefjes EC, van der Vorst MJ, Blauwhoff-Buskermolen S, Verheul HM. Aiming for a better understanding and management of cancer-related fatigue. Oncologist 2013;18:1135–43.

3. Radbruch L, Strasser F, Elsner F, et al. Fatigue in palliative care patients -- an EAPC approach. Palliat Med 2008;22:13–32.

4. Capuron L, Pagnoni G, Demetrashvili MF, et al. Basal ganglia hypermetabolism and symptoms of fatigue during interferon-alpha therapy. Neuropsychopharmacology 2007;32:2384–92.

5. Kisiel-Sajewicz K, Siemionow V, Seyidova-Khoshknabi D, et al. Myoelectrical manifestation of fatigue less prominent in patients with cancer related fatigue. PLoS One 2013;8:e83636.

6. Bower JE, Ganz PA, Aziz N. Altered cortisol response to psychologic stress in breast cancer survivors with persistent fatigue. Psychosom Med 2005;67:277–80.

7. Barsevick A, Frost M, Zwinderman A, et al. I’m so tired: biological and genetic mechanisms of cancer-related fatigue. Qual Life Res 2010;19:1419–27.

8. Morrow GR, Hickok JT, Roscoe JA, et al. Differential effects of paroxetine on fatigue and depression: a randomized, double-blind trial from the University of Rochester Cancer Center Community Clinical Oncology Program. J Clin Oncol 2003;21:4635–41.

9. Fontes-Oliveira CC, Busquets S, Toledo M, et al. Mitochondrial and sarcoplasmic reticulum abnormalities in cancer cachexia: altered energetic efficiency? Biochim Biophys Acta 2013;1830:2770–8.

10. Agteresch HJ, Dagnelie PC, van der Gaast A, et al. Randomized clinical trial of adenosine 5’-triphosphate in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 2000;92:321–8.

11. Beijer S, Hupperets PS, van den Borne BE, et al. Randomized clinical trial on the effects of adenosine 5’-triphosphate infusions on quality of life, functional status, and fatigue in preterminal cancer patients. J Pain Symptom Manage 2010;40:520–30.

12. Kisiel-Sajewicz K, Davis MP, Siemionow V, et al. Lack of muscle contractile property changes at the time of perceived physical exhaustion suggests central mechanisms contributing to early motor task failure in patients with cancer-related fatigue. J Pain Symptom Manage 2012;44:351–61.

13. Ryan JL, Carroll JK, Ryan EP, et al. Mechanisms of cancer-related fatigue. Oncologist 2007;12 Suppl 1:22–34.

14. Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer 2008;8:887–99.

15. Wang XS, Giralt SA, Mendoza TR, et al. Clinical factors associated with cancer-related fatigue in patients being treated for leukemia and non-Hodgkin’s lymphoma. J Clin Oncol 2002;20:1319–28.

16. Collado-Hidalgo A, Bower JE, Ganz PA, et al. Inflammatory biomarkers for persistent fatigue in breast cancer survivors. Clin Cancer Res 2006;12:2759–66.

17. Wang XS, Shi Q, Williams LA, et al. Serum interleukin-6 predicts the development of multiple symptoms at nadir of allogeneic hematopoietic stem cell transplantation. Cancer 2008;113:2102–9.

18. Inagaki M, Isono M, Okuyama T, et al. Plasma interleukin-6 and fatigue in terminally ill cancer patients. J Pain Symptom Manage 2008;35:153–61.

19. Laird BJ, McMillan DC, Fayers P, et al. The systemic inflammatory response and its relationship to pain and other symptoms in advanced cancer. Oncologist 2013;18:1050–5.

20. Bower JE, Ganz PA, Irwin MR, et al. Inflammation and behavioral symptoms after breast cancer treatment: do fatigue, depression, and sleep disturbance share a common underlying mechanism? J Clin Oncol 2011;29:3517–22.

21. Whistler T, Taylor R, Craddock RC, et al. Gene expression correlates of unexplained fatigue. Pharmacogenomics 2006;7:395–405.

22. Fagundes CP, Bennett JM, Alfano CM, et al. Social support and socioeconomic status interact to predict Epstein-Barr virus latency in women awaiting diagnosis or newly diagnosed with breast cancer. Health Psychol 2012;31:11–19.

23. Landmark-Hoyvik H, Reinertsen KV, Loge JH, et al. Alterations of gene expression in blood cells associated with chronic fatigue in breast cancer survivors. Pharmacogenomics J 2009;9:333–40.

24. Smith AK, Conneely KN, Pace TW, et al. Epigenetic changes associated with inflammation in breast cancer patients treated with chemotherapy. Brain Behav Immun 2014;38:227–36.

25. Kim S, Miller BJ, Stefanek ME, Miller AH. Inflammation-induced activation of the indoleamine 2,3-dioxygenase pathway: Relevance to cancer-related fatigue. Cancer 2015;121:2129–36.

26. Mills PJ, Parker B, Dimsdale JE, et al. The relationship between fatigue and quality of life and inflammation during anthracycline-based chemotherapy in breast cancer. Biol Psychol 2005;69:85–96.

27. Jean-Pierre P, Figueroa-Moseley CD, Kohli S, et al. Assessment of cancer-related fatigue: implications for clinical diagnosis and treatment. Oncologist 2007;12 Suppl 1:11–21.

28. Wang XS, Zhao F, Fisch MJ, et al. Prevalence and characteristics of moderate to severe fatigue: a multicenter study in cancer patients and survivors. Cancer 2014;120:425–32.

29. Mendoza TR, Wang XS, Cleeland CS, et al. The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory. Cancer 1999;85:1186–96.

30. Mendoza ME, Capafons A, Gralow JR, et al. Randomized controlled trial of the Valencia model of waking hypnosis plus CBT for pain, fatigue, and sleep management in patients with cancer and cancer survivors. Psychooncology 2016 Jul 28.

31. Glaus A. Assessment of fatigue in cancer and non-cancer patients and in healthy individuals. Support Care Cancer 1993;1:305–15.

32. Seyidova-Khoshknabi D, Davis MP, Walsh D. A systematic review of cancer-related fatigue measurement questionnaires. Am J Hosp Palliat Care 2011;28:119–29.

33. Holzner B, Kemmler G, Greil R, et al. The impact of hemoglobin levels on fatigue and quality of life in cancer patients. Ann Oncol 2002;13:965–73.

34. Stein KD, Jacobsen PB, Blanchard CM, Thors C. Further validation of the multidimensional fatigue symptom inventory-short form. J Pain Symptom Manage 2004;27:14–23.

35. Hwang SS, Chang VT, Rue M, Kasimis B. Multidimensional independent predictors of cancer-related fatigue. J Pain Symptom Manage 2003;26:604–14.

36. Peterspm DR. Scope and generality of verbally defined personality factors. Psychol Rev 1965;72:48–59.

37. Berger AM, Abernethy AP, Atkinson A, et al. NCCN Clinical Practice Guidelines Cancer-related fatigue. J Natl Compr Canc Netw 2010;8:904–31.

38. Crawford J, Cella D, Cleeland CS, et al. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002;95:888–95.

39. Tonia T, Mettler A, Robert N, et al. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst Rev 2012;12:CD003407.

40. Rizzo JD, Brouwers M, Hurley P, et al. American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood 2010;116:4045–59.

41. Preston NJ, Hurlow A, Brine J, Bennett MI. Blood transfusions for anaemia in patients with advanced cancer. Cochrane Database Syst Rev 2012(2):CD009007.

42. Minton O, Stone PC. A comparison of cognitive function, sleep and activity levels in disease-free breast cancer patients with or without cancer-related fatigue syndrome. BMJ Support Palliat Care 2012;2:231–8.

43. Heckler CE, Garland SN, Peoples AR, et al. Cognitive behavioral therapy for insomnia, but not armodafinil, improves fatigue in cancer survivors with insomnia: a randomized placebo-controlled trial. Support Care Cancer 2016;24:2059–66.

44. Howell D, Oliver TK, Keller-Olaman S, et al. Sleep disturbance in adults with cancer: a systematic review of evidence for best practices in assessment and management for clinical practice. Ann Oncol 2014;25:791–800.

45. Wilt TJ, MacDonald R, Brasure M, et al. Pharmacologic treatment of insomnia disorder: an evidence report for a clinical practice guideline by the American College of Physicians. Ann Intern Med 2016;165:103–12.

46. Kuriyama A, Honda M, Hayashino Y. Ramelteon for the treatment of insomnia in adults: a systematic review and meta-analysis. Sleep Med 2014;15:385–92.

47. Strasser F, Palmer JL, Schover LR, et al. The impact of hypogonadism and autonomic dysfunction on fatigue, emotional function, and sexual desire in male patients with advanced cancer: a pilot study. Cancer 2006;107:2949–57.

48. Agasi-Idenburg SC, Thong MS, Punt CJ, et al. Comparison of symptom clusters associated with fatigue in older and younger survivors of colorectal cancer. Support Care Cancer 2017;25:625–32.

49. Miaskowski C, Aouizerat BE. Is there a biological basis for the clustering of symptoms? Semin Oncol Nurs 2007;23:99–105.

50. de Raaf PJ, de Klerk C, Timman R, et al. Systematic monitoring and treatment of physical symptoms to alleviate fatigue in patients with advanced cancer: a randomized controlled trial. J Clin Oncol 2013;31:716–23.

51. Barsevick AM, Whitmer K, Sweeney C, Nail LM. A pilot study examining energy conservation for cancer treatment-related fatigue. Cancer Nurs 2002;25:333–41.

52. Barsevick AM, Dudley W, Beck S, et a;. A randomized clinical trial of energy conservation for patients with cancer-related fatigue. Cancer 2004;100:1302–10.

53. Luciani A, Jacobsen PB, Extermann M, et al. Fatigue and functional dependence in older cancer patients. Am J Clin Oncol 2008;31:424–30.

54. Abrahams HJ, Gielissen MF, Goedendorp MM, et al. A randomized controlled trial of web-based cognitive behavioral therapy for severely fatigued breast cancer survivors (CHANGE-study): study protocol. BMC Cancer 2015;15:765.

55. Quesnel C, Savard J, Simard S, et al. Efficacy of cognitive-behavioral therapy for insomnia in women treated for nonmetastatic breast cancer. J Consult Clin Psychol 2003;71:189–200.

56. Goedendorp MM, Gielissen MF, Verhagen CA, Bleijenberg G. Psychosocial interventions for reducing fatigue during cancer treatment in adults. Cochrane Database Syst Rev 2009(1):CD006953.

57. Greiwe JS, Cheng B, Rubin DC, et al. Resistance exercise decreases skeletal muscle tumor necrosis factor alpha in frail elderly humans. FASEB J 2001;15:475–82.

58. Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev 2016;(9):CD005001.

59. Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database Syst Rev 2012;(11):CD006145.

60. Brown JC, Huedo-Medina TB, Pescatello LS, et al. Efficacy of exercise interventions in modulating cancer-related fatigue among adult cancer survivors: a meta-analysis. Cancer Epidemiol Biomarkers Prev 2011;20:123–33.

61. Steindorf K, Schmidt ME, Klassen O, et al. Randomized, controlled trial of resistance training in breast cancer patients receiving adjuvant radiotherapy: results on cancer-related fatigue and quality of life. Ann Oncol 2014;25:2237–43.

62. Bower JE, Bak K, Berger A, et al. Screening, assessment, and management of fatigue in adult survivors of cancer: an American Society of Clinical oncology clinical practice guideline adaptation. J Clin Oncol 2014;32:1840–50.

63. Kenjale AA, Hornsby WE, Crowgey T, et al. Pre-exercise participation cardiovascular screening in a heterogeneous cohort of adult cancer patients. Oncologist 2014;19:999–1005.

64. Oldervoll LM, Loge JH, Paltiel H, et al. The effect of a physical exercise program in palliative care: A phase II study. J Pain Symptom Manage 2006;31:421–30.

65. Porock D, Kristjanson LJ, Tinnelly K, et al. An exercise intervention for advanced cancer patients experiencing fatigue: a pilot study. J Palliat Care 2000;16:30–6.

66. Lengacher CA, Kip KE, Reich RR, et al. A cost-effective mindfulness stress reduction program: a randomized control trial for breast cancer survivors. Nursing Econ 2015;33:210–8, 32.

67. Lengacher CA, Reich RR, Post-White J, et al. Mindfulness based stress reduction in post-treatment breast cancer patients: an examination of symptoms and symptom clusters. J Behav Med 2012;35:86–94.

68. Sprod LK, Fernandez ID, Janelsins MC, et al. Effects of yoga on cancer-related fatigue and global side-effect burden in older cancer survivors. J Geriatr Oncol 2015;6:8–14.

69. Wang G, Wang S, Jiang P, Zeng C. [Effect of Yoga on cancer related fatigue in breast cancer patients with chemotherapy]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2014;39:1077–82.

70. Stan DL, Croghan KA, Croghan IT, et al. Randomized pilot trial of yoga versus strengthening exercises in breast cancer survivors with cancer-related fatigue. Support Care Cancer 2016;24:4005–15.

71. Larkey LK, Roe DJ, Weihs KL, et al. Randomized controlled trial of Qigong/Tai Chi Easy on cancer-related fatigue in breast cancer survivors. Ann Behav Med 2015;49:165–76.

72. Molassiotis A, Bardy J, Finnegan-John J, et al. Acupuncture for cancer-related fatigue in patients with breast cancer: a pragmatic randomized controlled trial. J Clin Oncol 2012;30:4470–6.

73. Deng G, Chan Y, Sjoberg D, et al. Acupuncture for the treatment of post-chemotherapy chronic fatigue: a randomized, blinded, sham-controlled trial. Support Care Cancer 2013;21:1735–41.

74. Finnegan-John J, Molassiotis A, Richardson A, Ream E. A systematic review of complementary and alternative medicine interventions for the management of cancer-related fatigue. Integr Cancer Ther 2013;12:276–90.

75. Schwartz AL, Thompson JA, Masood N. Interferon-induced fatigue in patients with melanoma: a pilot study of exercise and methylphenidate. Oncol Nurs Forum 2002;29:E85–90.

76. Spathis A, Dhillan R, Booden D, et al. Modafinil for the treatment of fatigue in lung cancer: a pilot study. Palliat Med 2009;23:325–31.

77. Blackhall L, Petroni G, Shu J, et al. A pilot study evaluating the safety and efficacy of modafinal for cancer-related fatigue. J Palliat Med 2009;12:433–9.

78. Qu D, Zhang Z, Yu X, et al. Psychotropic drugs for the management of cancer-related fatigue: a systematic review and meta-analysis. Eur J Cancer Care (Engl) 2015;25:970–9.

79. Minton O, Richardson A, Sharpe M, et al. Drug therapy for the management of cancer-related fatigue. Cochrane Database Syst Rev 2010(7):CD006704.

80. Moraska AR, Sood A, Dakhil SR, et al. Phase III, randomized, double-blind, placebo-controlled study of long-acting methylphenidate for cancer-related fatigue: North Central Cancer Treatment Group NCCTG-N05C7 trial. J Clin Oncol 2010;28:3673–9.

81. Bruera E, Driver L, Barnes EA, et al. Patient-controlled methylphenidate for the management of fatigue in patients with advanced cancer: a preliminary report. J Clin Oncol 2003;21:4439–43.

82. Kerr CW, Drake J, Milch RA, et al. Effects of methylphenidate on fatigue and depression: a randomized, double-blind, placebo-controlled trial. J Pain Symptom Manage 2012;43:68–77.

83. Escalante CP, Meyers C, Reuben JM, et al. A randomized, double-blind, 2-period, placebo-controlled crossover trial of a sustained-release methylphenidate in the treatment of fatigue in cancer patients. Cancer J 2014;20:8–14.

84. Mar Fan HG, Clemons M, Xu W, et al. A randomised, placebo-controlled, double-blind trial of the effects of d-methylphenidate on fatigue and cognitive dysfunction in women undergoing adjuvant chemotherapy for breast cancer. Support Care Cancer 2008;16:577–83.

85. Butler JM Jr, Case LD, Atkins J, et al. A phase III, double-blind, placebo-controlled prospective randomized clinical trial of d-threo-methylphenidate HCl in brain tumor patients receiving radiation therapy. Int J Radiat Oncol Biol Phys 2007;69:1496–501.

86. Hovey E, de Souza P, Marx G, et al. Phase III, randomized, double-blind, placebo-controlled study of modafinil for fatigue in patients treated with docetaxel-based chemotherapy. Support Care Cancer 2014;22:1233–42.

87. Spathis A, Fife K, Blackhall F, et al. Modafinil for the treatment of fatigue in lung cancer: results of a placebo-controlled, double-blind, randomized trial. J Clin Oncol 2014;32:1882–8.

88. Berenson JR, Yellin O, Shamasunder HK, et al. A phase 3 trial of armodafinil for the treatment of cancer-related fatigue for patients with multiple myeloma. Support Care Cancer 2015;23:1503–12.

89. Boele FW, Douw L, de Groot M, et al. The effect of modafinil on fatigue, cognitive functioning, and mood in primary brain tumor patients: a multicenter randomized controlled trial. Neuro Oncol 2013;15:1420–8.

90. Jean-Pierre P, Morrow GR, Roscoe JA, et al. A phase 3 randomized, placebo-controlled, double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631 patients receiving chemotherapy: a University of Rochester Cancer Center Community Clinical Oncology Program Research base study. Cancer 2010;116:3513–20.

91. Conley CC, Kamen CS, Heckler CE, et al. Modafinil moderates the relationship between cancer-related fatigue and depression in 541 patients receiving chemotherapy. J Clin Psychopharmacol 2016;36:82–5.

92. Brattsand R, Linden M. Cytokine modulation by glucocorticoids: mechanisms and actions in cellular studies. Aliment Pharmacol Ther 1996;10 Suppl 2:81–90.

93. Yennurajalingam S, Frisbee-Hume S, Palmer JL, et al. Reduction of cancer-related fatigue with dexamethasone: a double-blind, randomized, placebo-controlled trial in patients with advanced cancer. J Clin Oncol 2013;31:3076–82.

94. Bruera E, Roca E, Cedaro L, et al. Action of oral methylprednisolone in terminal cancer patients: a prospective randomized double-blind study. Cancer Treat Rep 1985;69:751–4.

95. Pulivarthi K, Dev R, Garcia J, et al. Testosterone replacement for fatigue in male hypogonadic patients with advanced cancer: A preliminary double-blind placebo-controlled trial. J Clin Oncol 2012;30 (suppl). Abstract e19643.

96. Palesh OG, Mustian KM, Peppone LJ, et al. Impact of paroxetine on sleep problems in 426 cancer patients receiving chemotherapy: a trial from the University of Rochester Cancer Center Community Clinical Oncology Program. Sleep Med 2012;13:1184–90.

97. Thekdi SM, Trinidad A, Roth A. Psychopharmacology in Cancer. Curr Psychiatry Rep 2014;17:529.

98. Lesser GJ. Case D, Stark N, et al. A randomized, double-blind, placebo-controlled study of oral coenzyme Q10 to relieve self-reported treatment-related fatigue in newly diagnosed patients with breast cancer. J Support Oncol 2013;11:31–42.

99. Barton DL, Liu H, Dakhil SR, et al. Wisconsin Ginseng (Panax quinquefolius) to improve cancer-related fatigue: a randomized, double-blind trial, N07C2. J Natl Cancer Inst 2013;105:1230–8.

100. Yennurajalingam S, Reddy A, Tannir NM, et al. High-dose Asian ginseng (panax ginseng) for cancer-related fatigue: a preliminary report. Integr Cancer Ther 2015;14:419–27.

101. Howell D, Keller-Olaman S, Oliver TK, et al. A pan-Canadian practice guideline and algorithm: screening, assessment, and supportive care of adults with cancer-related fatigue. Curr Oncol 2013;20:e233–46.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Sections

From the University of Texas MD Anderson Cancer Center, Houston, TX.

 

Abstract

  • Objective: To review the evidence on interventions for managing cancer-related fatigue (CRF) and provide evidence-based guidance on approaches to its management.
  • Methods: Nonsystematic review of the literature.
  • Results: Several theories have been proposed to explain the biology of CRF, but there is no single clear mechanism that can be targeted for therapy. The approach to patients begins with screening for fatigue and assessing its intensity, followed by a thorough history and examination to determine whether any reversible medical conditions are contributing to fatigue. Management of underlying medical comorbidities may help some patients. For patients whose fatigue persists, pharmacologic and nonpharmacologic treatment options are available. Pharmacologic options include psychostimulants, such as methylphenidate and modafinil, and corticosteroids. Nonpharmacologic approaches include exercise, cognitive behavior therapy, yoga, acupuncture, and tai chi.
  • Conclusion: We recommend an individualized approach, often with a combination of the available options. Patients need to be evaluated periodically to assess their fatigue, and since cancer-related fatigue affects survivors, long-term follow-up is needed.

Key words: fatigue; cancer; pro-inflammatory cytokines; nonpharmacologic; psychostimulants.

 

Fatigue is a common distressing effect of cancer [1].It impairs the quality of life of patients undergoing active cancer treatment and of post-treatment survivors. The National Comprehensive Cancer Network (NCCN) defines cancer-related fatigue (CRF) as “a distressing, persistent, subjective sense of physical, emotional and/or cognitive tiredness related to cancer or cancer treatment that is not proportional to recent activity and interferes with usual functioning [2].” Differences between CRF and fatigue reported by individuals without cancer are that CRF is more severe and is not relieved by rest. The prevalence of CRF in cancer patients and survivors is highly variable, ranging between 25% and 99% [2,3]. This variability may be secondary to methods used for screening fatigue and characteristics of the patient groups. In this article, we discuss recognition of CRF and approaches to its management.

 

Pathophysiology

The specific pathophysiologic mechanism underlying CRF is unknown, making targeted treatment a challenge. The multidimensional and subjective nature of CRF has limited the development of research methodologies to explain this condition. However, research has been done in both human and animal models, and several theories have been proposed to explain the pathophysiology of CRF. While pro-inflammatory cytokines remain the central factor playing a significant role at multiple levels in CRF, there may be a complex interplay of more than 1 mechanism contributing to fatigue in an individual patient.

Central Nervous System Disturbances

The basal ganglia are known to influence motivation. Lack of motivation and drive may cause failure to complete physical and mental tasks, even with preserved cognitive ability and motor function. In a study of melanoma patients receiving interferon, increased activity of the basal ganglia and the cerebellum resulted in higher fatigue scores [4]. Higher levels of cytokines may alter blood flow to the cerebellum and lead to the perception of fatigue. In a study of 12 patients and matched controls, when patients were asked to perform sustained elbow flexion until they perceived exhaustion, CRF patients perceived physical exhaustion sooner than controls. In CRF patients in this study, muscle fatigue measured by electromyogram was less than that in healthy individuals at the time of exhaustion, suggesting the role of the central nervous system in CRF [5]. However, there is not enough evidence at this time to support central nervous system disturbance as the main contributing factor to fatigue in cancer patients.

Circadian Rhythm Dysregulation

Circadian rhythm is regulated by the suprachiasmatic nucleus in the hypothalamus through cortisol and melatonin. Sleep disturbances occur with disruption of the circadian rhythm. Tumor-related peptides such as epidermal growth factor or alterations in serotonin and cortisol can influence the suprachiasmatic nucleus and the complex signaling pathways [2]. Positive feedback loops that are activated by cortisol under the influence of cytokines may lead to continuous cytokine production and altered circadian rhythm. Bower et al showed that changes in the cortisol curve influence fatigue in breast cancer survivors [6]. These patients had a late evening peak in cortisol levels, compared with an early morning peak in individuals without cancer.

Inhibition of Hypothalamic–Pituitary–Adrenal Axis

The hypothalamic–pituitary–adrenal (HPA) axis regulates the release of the stress hormone cortisol. One of several hypotheses advanced to explain the effect of serotonin and the HPA axis on CRF suggests that lower serotonin levels cause decreased activation of 5-hydroxytrytophan 1-a (5-HT1-a) receptors in the hypothalamus, leading to decreased activity of the HPA axis [6]. The inhibition of the HPA axis may occur with higher levels of serotonin as well [7]. The 5-HT1-a receptors are also triggered by cytokines. However, the correction of serotonin levels by antidepressants was not shown to improve fatigue [8]. Inhibition of the HPA axis can also lead to lower testosterone, progesterone, or estrogen levels, which may indirectly contribute to fatigue [2].

Skeletal Muscle Effect

Chemotherapy- and tumor-related cachexia have a direct effect on the metabolism of skeletal muscles. This effect may lead to impaired adenosine triphosphate (ATP) generation during muscle contraction [9]. ATP infusion improved muscle strength in one trial, but this was not confirmed in another trial [10,11]. Muscle contraction studies showed no differences in the contractile properties of muscles in fatigued patients who failed earlier in motor tasks and healthy controls [12]. This finding suggests that there could be a failure of skeletal muscle activation by the central nervous system or inhibition of skeletal muscle activity. Cytokines and other neurotransmitters activate vagal efferent nerve fibers, which may lead to reflex inhibition in skeletal muscles [13,14].

Pro-inflammatory Cytokines

Tumors or treatment of them may cause tissue injury, which triggers immune cells to release cytokines, signaling the brain to manifest the symptom fatigue. Inflammatory pathways are influenced by psychological, behavioral, and biological factors, which play a role as risk factors in CRF. Interleukin 6 (IL-6), interleukin-1 receptor antagonist, interleukin-1, and tumor necrosis factor (TNF) have been shown to be elevated in fatigued patients being treated for leukemia and non-Hodgkin lymphoma [15]. IL-6 was also associated with increased fatigue in breast cancer survivors [16]. Similar findings were reported in patients undergoing stem cell transplantation and high-dose chemotherapy [17]. Elevated levels of IL-6 and C-reactive protein were also linked to fatigue in terminally ill cancer patients [18,19]. Furthermore, TNF-α signaling was associated with post-chemotherapy fatigue in breast cancer patients [20]. Leukocytes in breast cancer survivors with fatigue also have increased gene expression of pro-inflammatory cytokines, emphasizing the role of cytokines and inflammation in the pathogenesis of CRF [21].

Other Hypotheses

Several other hypotheses for CRF pathogenesis have been proposed. Activation of latent viruses such as Epstein-Barr virus, lack of social support [22], genetic alterations in immune pathway [23], epigenetic changes [24], accumulation of neurotoxic metabolites and depletion of serotonin by indoleamine 2,3-dioxygenase pathway activation [25], elevated vascular endothelial growth factor levels [26], and hypoxia-related organ dysfunction due to anemia or hemoglobin dysfunction [13] all have been postulated to cause CRF.

Approach to Evaluation and Treatment

The evaluation and treatment of CRF involve 4 steps (Figure). First, patients are screened for fatigue, and in the second step those who have fatigue undergo primary evaluation to assess for potential precipitating causes. The third step is implementation of pharmacologic and non-pharmacologic interventions aimed at alleviating or mitigating fatigue. The fourth step involves re-evaluating patients at periodic intervals to recognize and manage changes in fatigue levels. A multidisciplinary approach involving nursing, physical therapy, social work, and nutrition is critical in managing fatigue in these patients. Education and counselling of patients and involvement of the family are essential for effective management.

Screening

Because patients and health care professionals may be unaware of the treatment options available for CRF, patients may not report fatigue levels to their clinicians, and clinicians may not understand the impact of fatigue on their patients’ quality of life. This leads to underrecognition of the problem. The NCCN recommends screening every cancer patient and post-treatment survivor for fatigue [2]. Patients should be screened at their first visit and then at periodic intervals during and after cancer treatment.

Many scales are available to screen patients for CRF in clinical practice and clinical trials [27]. A single item that asks patients to rate their fatigue on a scale from 0 to 10—in which 0 indicates no fatigue, 1 to 3 indicates mild fatigue, 4 to 6 indicates moderate fatigue, 7 to 9 indicates severe fatigue, and 10 indicates the worst fatigue imaginable—is commonly used to screen for CRF [2]. This scale was adapted from the MD Anderson Symptom Inventory scale and is based on a large nationwide study of cancer patients and survivors [28]. The statistically derived cutoff points in this study are consistent with other scales such as the Brief Fatigue Inventory (BFI) and support the cutoff points (4–6 for moderate and ≥ 7 for severe fatigue) used in various fatigue management guidelines. Furthermore, studies of fatigue in cancer patients have revealed a marked decrease in physical function at levels of 7 or higher, suggesting 7 as an optimal cutoff to identify severe fatigue [29,30]. The Visual Analog Scale is another simple-to-use tool that helps in understanding variations in fatigue throughout the course of the day [31]. The 9-item BFI is often used in clinical trials [29]. It measures the severity of fatigue over the previous 24 hours and has been validated in non-English speaking patients [32].

 

 

CRF affects not only the somatic domain, but also the cognitive, behavioral, and affective domains; therefore, multidimensional scales have been developed for screening. One such tool is the Multidimensional Fatigue Inventory, which measures general, physical, mental, and emotional fatigue domains as well as activity and compares them with those of individuals without cancer [33,34]. The Functional Assessment of Cancer Therapy for Fatigue (FACT-F) is a 13-item questionnaire that has been used to measure CRF in clinical trials as well as in patients receiving various treatments [35].

Although many scales are available, the validity of self-reporting on simple fatigue-rating scales is equal to or better than most complex, lengthy scales [36]. Therefore, unidimensional tools such as the numeric rating scale of 0–10 are commonly used in clinical practice. Mild fatigue (0–3) requires periodic re-evaluation, and moderate and severe fatigue need further evaluation and management [37].

Primary Evaluation

This phase involves a focused history and physical examination and assessment of concurrent symptoms and contributing factors.

 

History and Physical Examination

A detailed history of the patient’s malignancy and type of previous and current treatment may help reveal the cause of fatigue. New-onset fatigue or increase in fatigue may be related to the progression of disease in patients with active malignancy or recurrence of cancer in survivors. These patients may require appropriate testing to assess the underlying disease pattern. A detailed review of systems may help identify some of the contributing factors, which are discussed below. A detailed history regarding medications, including over-the-counter drugs, complementary agents, and past and prior cancer therapies, is helpful as medications can contribute to fatigue. For example, opioids may cause drowsiness and fatigue, which could be improved by dose adjustments. A focused history of fatigue should be obtained in all patients with moderate to severe CRF, which includes the onset, pattern, duration, associated or alleviating factors, and interference with functioning, including activities of daily living [37]. Physical examination should focus on identifying signs of organ dysfunction and features of substance or alcohol abuse which may cause poor sleep and fatigue.

 

Assessment of Contributing Factors

The management of fatigue should be multifactorial, with a comprehensive assessment and treatment plan to address all modifiable fatigue etiologies. The Table lists potential contributing factors to fatigue that should be considered when evaluating patients for CRF; several common conditions are discussed below.

Anemia. Anemia has been correlated with fatigue and quality of life. In a study of 4382 cancer patients receiving chemotherapy, quality-of-life measures using FACT-Anemia scores improved with increased hemoglobin levels [38]. Cancer patients may have anemia due to marrow-suppressing effects of chemotherapy and may also have iron deficiency anemia due to blood loss or autoimmune hemolytic anemia. Therefore, a detailed work-up is required to identify the etiology of anemia. Patients with CRF whose anemia is related to chemotherapy or anemia of chronic disease may benefit from red blood cell transfusion or erythropoiesis-stimulating agents (ESAs). ESAs have been studied extensively; however, their use is controversial because of concerns about thromboembolic side effects leading to adverse outcomes [39]. Also, ESA therapy is not recommended in patients with hematologic malignancies. ESA use should be restricted to patients with chemotherapy-related anemia with hemoglobin below 10 mg/dL and should be discontinued in 6 to 8 weeks if patients do not respond [40]. Other patients may benefit from blood transfusions, which were shown to help in patients with hemoglobin levels between 7.5 and 8.5 g/dL [41].

Sleep disturbance. Poor sleep is common in fatigued cancer survivors [42]. Pro-inflammatory cytokines can disrupt the sleep–wake cycle, causing changes in the HPA axis and neuroendocrine system, which in turn may lead to increasing fatigue. Heckler et al showed that improvement in nighttime sleep leads to improvement of fatigue [43]. Cognitive behavioral therapy and sleep hygiene are important in caring for patients with CRF and poor sleep [44]. Taking a warm bath and/or drinking a glass of milk before bedtime, avoiding caffeinated drinks, and avoiding frequent napping in the day might help. Some patients may require medications such as benzodiazepines or non-benzodiazepine hypnotics (eg, zolpidem) to promote sleep [45]. Melatonin agonists are approved for insomnia in the United states, but not in Europe [46].

Malnutrition. Patients with advanced-stage cancer and with cancers affecting the gastrointestinal tract frequently develop mechanical bowel obstructions, especially at the end of their life, which cause malnutrition. Chemotherapy-related nausea and vomiting may also cause poor oral intake and malnutrition, causing fatigue from muscle weakness. Dehydration and electrolyte imbalances frequently occur as a result of poor oral intake, which might worsen fatigue. In our experience, modifying dietary intake with appropriate caloric exchanges with the help of a nutrition expert has lessened fatigue in some patients. However, terminally ill patients are advised to eat based on their comfort.

Medical comorbidities. Congestive heart failure from anthracycline chemotherapy, hypothyroidism after radiation therapy for head and neck cancers, renal failure, or hepatic failure from chemotherapy may indirectly lead to fatigue. Chemotherapy, opioids, and steroids may cause hypogonadism, which can contribute to fatigue in men [47].

 

 

Assessment of Concurrent Symptoms

Depression is common in cancer patients and coexists with pain, insomnia, fatigue, and anxiety as a symptom cluster [48]. A symptom cluster is defined as 2 or more concurrent and interrelated symptoms occurring together; treating of one of these symptoms without addressing other symptoms is not effective [49]. Therefore, screening for and management of depression, anxiety, and insomnia play an important role in the management of CRF.

Physical symptoms due to the tumor or to therapy—such as pain, dyspnea, nausea, and decreased physical activity—may also contribute to fatigue both directly and indirectly. Patients with lung cancer may have hypoxemia, which can contribute to dyspnea with activity and a perception of fatigue. Optimal management of pain and other physical symptoms in patients with cancer may significantly alleviate fatigue [50].

Management

Management of CRF is individualized based on the patient’s clinical status: active cancer treatment, survivor, or end of life. Education and counselling of patients and their caregivers play an important role in CRF. NCCN guidelines recommend focusing on pain control, distress management, energy conservation, physical activity, nutrition, and sleep hygiene.

 

Nonpharmacologic Interventions

Energy conservation. Energy conservation strategies, in which patients are advised to set priorities and realistic expectations, are highly recommended. Some energy-conserving strategies are to pace oneself, delegate and schedule activities at times of peak energy, postpone nonessential activities, attend to 1 activity at a time, structure daily routines, and maintain a diary to identify their peak energy period and structure activities around that time [51,52]. When patients approach the end of life, increasing fatigue may limit their activity level, and they may depend on caregivers for assistance with activities of daily living, monitoring treatment-related adverse effects, and taking medications, especially elderly patients [53].

Cognitive behavioral therapy. Cognitive behavioral therapy (CBT) has been shown to improve CRF during active treatment, and the benefits persist for a minimum of 2 years after therapy [54]. CBT interventions that optimize sleep quality may improve fatigue [55]. More studies are needed to understand whether referral to a psychologist for formal CBT is required. Randomized clinical trials (RCTs) showed patient fatigue education, learned self-care, coping techniques, and balancing rest and activity benefit patients with CRF [56].

Exercise. Physical activity is highly encouraged in patients with CRF. Exercise increases muscle protein synthesis, improves cytokine response, and decreases the rate of sarcopenia in healthy populations [57]. Studies have shown that exercise helps CRF at all phases of the cancer journey, including radiation therapy, chemotherapy, and survivorship [58]. Some patients may feel less motivated to exercise and may not believe that exercise is possible or could potentially help them. Counselling is needed for such patients.

Older cancer survivors have a decline in their functional capacity and reduced muscle mass. Exercise can improve cardiorespiratory fitness, muscle strength, and body composition [57]. Exercise not only helps at the cellular level but also has psychosocial benefits from improved self-esteem. Therefore, exercise may be recommended not only for younger patients, but also in the older population, who may have comorbidities and less motivation than younger patients.

In a meta-analysis of 56 randomized controlled trials involving 4068 participants, aerobic exercise was found to have beneficial effects on CRF for patients during and after chemotherapy, specifically for patients with solid tumors [59]. In another meta-analysis of breast and prostate cancer survivors, a combination of aerobic exercise with resistance training (3–6 metabolic equivalents, 60%–80% range of motion) was shown to reduce CRF more than aerobic exercise alone [60]. This effect was also shown in an RCT of 160 patients with stage 0 to III breast cancer undergoing radiation therapy [61]. The control group in this study had a group-based non-exercise intervention/relaxation; therefore, the study showed that the effect of resistance training extends beyond the psychosocial benefits of group-based interventions. The intervention comprised 8 progressive machine-based resistance exercises (3 sets, 8–12 repetitions at 60%–80% of 1 repetition maximum) for 60 minutes twice weekly for 12 weeks. However, fatigue assessment questionnaire scores showed benefits in the physical fatigue but not the affective and cognitive components.

The American Society of Clinical Oncology’s guidelines for cancer survivors with fatigue recommends 150 minutes of moderate aerobic exercise (eg, fast walking, cycling, or swimming) per week, with 2 or 3 sessions of strength training per week [62]. An individualized approach to exercise is recommended, as patients’ ability to perform certain types of exercises may be limited by thrombocytopenia, neutropenia, or lytic bone metastasis. Routine use of pre-exercise cardiovascular testing is not recommended but may be considered in high-risk populations, especially patients with risk factors for coronary heart disease and diabetes [63]. Patients withcomorbidities, substantial deconditioning, functional and anatomic defects, or recent major surgery may benefit from referral to physical therapy [37]. Patients near end of life may also benefit from an exercise program, as demonstrated in several studies that showed benefit in CRF and quality of life [64,65]. We recommend that physicians use their best clinical judgement in suggesting the type and intensity of exercise program, as it may not be feasible in some patients.

Mind-body interventions. Mindfulness-based stress reduction (MBSR) has shown promise in breast cancer survivors, who reported immediate improvements in fatigue severity that continued up to 6 weeks after cessation of the training [66]. Prior studies had similar findings, suggesting that MBSR modestly decreases fatigue and sleep disturbances and has a greater effect on the degree to which symptoms interfere with many facets of life [67].

Yoga. A study of a yoga intervention showed a benefit in older cancer survivors [68]. In breast cancer patients undergoing chemotherapy, yoga was shown to benefit not only physical fatigue, but also cognitive fatigue [69]. DVD-based yoga had benefits similar to strengthening exercises in a study of 34 early-stage breast cancer survivors with CRF [70]. More studies are needed in men and patients and survivors of other cancers, as most studies of yoga were conducted in women with breast cancer.

Tai chi/qigong. Like yoga, tai chi and qigong are practices of meditative movement. These practices use postures or movements with a focus on breath and a meditative state to bring about deep states of relaxation. Qigong is a series of simple, repeated practices including body posture/movement, breath practice, and meditation performed in synchrony. Tai chi easy (TCE) is a simplified set of common, repetitive tai chi movements. In a trial, qigong/TCE was compared with sham qigong, which had physical movements but no breathing or meditative practice. Breast cancer survivors in the qigong/TCE group had improved fatigue scores, and the effect persisted for 3 months [71]. Additional research is needed in this area.

Acupuncture. An RCT in breast cancer patients with CRF showed an improvement in the mean general fatigue score (per the Multidimensional Fatigue Inventory) in patients who received acupuncture versus those who did not (−3.11 [95% confidence interval −3.97 to −2.25]; P < 0.001) at 6 weeks. Improvements were seen in both the mental and physical aspects of fatigue [72]. However, Deng et al noted that true acupuncture was no more effective than sham acupuncture for reducing post-chemotherapy chronic fatigue [73]. Presently, there is not sufficient evidence to evaluate the benefits of acupuncture in CRF.

Other modalities. Massage therapy, music therapy, hypnosis, therapeutic touch, biofield therapies, relaxation, and reiki are other therapies for which few studies have been done, with mixed results, and additional research is needed [74]. Currently, there are not sufficient data to recommend any of these modalities.

Pharmacologic Interventions

Psychostimulants. Methylphenidate and modafinil are psychostimulants or wakefulness-promoting agents. Pilot studies showed benefit from methylphenidate and modafinil in CRF [75–77], but RCTs have yielded mixed results. Therefore, in patients with severe fatigue during cancer therapy, the initial management strategy involves evaluation and treatment of medical conditions such as anemia and a trial of non-pharmacological strategies as discussed above. If symptoms persist, then a therapeutic trial of a psychostimulant may be considered per NCCN guidelines for patients undergoing active cancer treatment [37].

Methylphenidate directly stimulates adrenergic receptors and indirectly releases dopamine and norepinephrine from presynaptic terminals, which may explain why the drug benefits patients receiving opioid-induced sedation. It is a commonly studied psychostimulant, though its mechanism of action in CRF is unclear. RCTs of methylphenidate have resulted in a wide range of findings due to the heterogeneity of study populations and variations in the dosage of methylphenidate. A meta-analysis of 7 studies indicates that methylphenidate benefitted the subgroup of patients with CRF [78]. Likewise, in an analysis of 5 RCTs, Minton et al showed a benefit of psychostimulants in fatigue compared with placebo [79]. However, another study of methylphenidate in patients with CRF showed a benefit only in patients with severe fatigue or advanced disease [80]. Methylphenidate was found to benefit cancer patients receiving opioid-induced sedation, as methylphenidate promotes wakefulness, though fatigue was not studied specifically [81]. In a trial with 30 hospice patients in which the methylphenidate dose was titrated based on response and adverse effects, Kerr at al found that the drug improved fatigue in a dose-dependent manner [82]. However, a study in patients with CRF at the University of Texas MD Anderson Cancer Center found no significant difference in BFI scores between patients receiving methylphenidate and those receiving placebo at the end of 2 weeks of treatment [83]. Also, other RCTs in patients undergoing adjuvant chemotherapy for breast cancer [84] and patients receiving radiation therapy for brain tumors [85] failed to demonstrate the efficacy of methylphenidate in CRF. It should be used cautiously after ruling out other causes of fatigue. The drug is overall well tolerated and side effects include headache and nausea.

Modafinil is a non-amphetamine psychostimulant that has been approved for the treatment of narcolepsy. In a trial studying the effect of modafinil on patients receiving docetaxel-based chemotherapy for metastatic breast or prostate cancer, there was a modest but not statistically significant improvement in fatigue scores on the MD Anderson Symptom Inventory compared with placebo. Nausea and vomiting were higher in the modafinil arm than in the placebo arm [86]. Similarly, modafinil was not superior to placebo for CRF in 208 patients with non-squamous cell lung cancer not undergoing chemotherapy or radiation [87]. A placebo effect was also noted in patients with multiple myeloma [88] and patients with primary brain tumors [89]. In a phase 3, multicenter, randomized, placebo-controlled, double-blind clinical trial of modafinil for CRF in 867 patients undergoing chemotherapy, there was a reduction in fatigue only for patients with severe baseline fatigue, with no significant effect on mild to moderate fatigue [90]. In another recent study, modafinil was shown to reduce depressive symptoms only in patients with severe fatigue (BFI item 3 score ≥ 7) [91]. This finding is consistent with previous studies showing benefit in patients with high baseline fatigue, but additional RCTs are needed to provide clarity. NCCN guidelines do not recommend the use of modafinil to treat CRF [37].

Other pharmacologic interventions. Corticosteroids are often used for symptom control in cancer patients. These drugs have anti-inflammatory effects through their modulation of pro-inflammatory cytokines [92]. In a RCT evaluating the efficacy of corticosteroids, patients receiving dexamethasone (4 mg twice daily) experienced significant improvement in their FACT-F scores compared with patients receiving placebo [93]. A similar benefit in fatigue was demonstrated in another study of methylprednisolone (32 mg daily) versus placebo [94]. Despite the benefits of steroids, their adverse effects, such as mood swings, gastritis, hyperglycemia, and immune suppression, limit their long-term use. Therefore, the use of steroids should be restricted to terminally ill fatigued patients with other symptoms such as anorexia, brain metastasis, or pain related to bone metastasis [37].

Testosterone replacement has been shown to diminish fatigue in non-cancer patients. Many men with advanced cancer have hypogonadism leading to low serum testosterone, which may cause fatigue. In a small trial in which cancer patients with hypogonadism received intramuscular testosterone every 14 days or placebo, the group receiving testosterone showed improvement in FACT-F scores, but there was no significant difference in FACT-F scores between the 2 groups [95].

Antidepressants have failed to demonstrate benefit in CRF without depression [8]. However, if a patient has both fatigue and depression, antidepressants may help [96]. A selective serotonin receptor inhibitor is recommended as a first-line antidepressant [97]. Patients with cancer are often receiving multiple medications, and medication interactions should be considered to prevent adverse events such as serotonin syndrome.

 

 

Complementary and Alternative Supplements

Studies using vitamin supplementation have been inconclusive in patients with CRF [74]. The use of other dietary supplements has yielded mixed results, and coenzyme Q has shown no benefit for patients with CRF [98].

The benefit of ginseng was studied in a RCT involving 364 patients with CRF. There was an improvement in Multidimensional Fatigue Symptom Inventory-short form (MFSI-SF) scores at 8 weeks in patients receiving 2000 mg of Wisconsin ginseng compared with patients receiving placebo [99]. Patients on active treatment had greater improvement as compared to the post-treatment group in this trial. In another study of high-dose panax ginseng (ginseng root) at 800 mg daily for 29 days, patients had improvement of CRF as well as overall quality of life, appetite, and sleep at night. It was also well tolerated with few adverse effects [100]. Interaction with warfarin, calcium channel blockers, antiplatelet agents, thrombolytic agents, imatinib, and other agents may occur; therefore, ginseng must be used with careful monitoring in selected patients. There is not enough evidence at this time to support the routine use of ginseng in CRF.

The seed extract of the guarana plant (Paullinia cupana) traditionally has been used as a stimulant. An improvement in fatigue scores was seen with the use of oral guarana (100 mg daily) at the end of 21 days in breast cancer patients receiving chemotherapy [101]. Further studies are needed for these results to be generalized and to understand the adverse effects and interaction profile of guarana.

 

Re-evaluation

Patients who have completed cancer treatment must be monitored for fatigue over the long term, as fatigue may exist beyond the period of active treatment. Many studies have shown fatigue in breast cancer survivors, and fatigue has been demonstrated in survivors of colorectal, lung, and prostate cancers as well as myeloproliferative neoplasms [28]. Therefore, it is important to screen patients for fatigue during follow-up visits. There are currently no studies evaluating the long-term treatment of fatigue. In our experience, the timing of follow-up depends on the level of fatigue and interventions prescribed. Once fatigue is stabilized to a level with which the patient is able to cope, the time interval for follow up may be lengthened. Annual visits may suffice in patients with mild fatigue. Follow-up of patients with moderate to severe fatigue depends on the level of fatigue, the ability to cope, choice of treatment, and presence of contributing factors.

Conclusion

CRF is a complex condition that places a significant burden on patients and caregivers, resulting in emotional distress, poor functioning, and suffering. Fatigue can occur before, during, and long after cancer treatment. The approach to CRF begins with screening for and educating patients and their caregivers about the symptoms. Many screening scales are available that may be used to follow patients’ progress over time. The evaluation and management of contributing conditions may help improve fatigue. If the fatigue persists, an individualized approach with a combination of nonpharmacologic and pharmacologic approaches should be considered. More research is needed to understand brain signaling pathways, cytokine changes, and genomic changes in cancer patients with fatigue. Though many hypotheses have been proposed, to date there is no biological marker to assess this condition. Biomarker research needs to be advanced to help to identify patients at risk for fatigue. As cytokines have a major role in CRF, targeted therapy to block cytokine pathways may also be explored in the future.

 

Acknowledgment: Bryan Tutt provided editorial assistance.

Corresponding author: Carmelita P. Escalante, MD, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX 77030, [email protected].

Financial disclosures: None.

From the University of Texas MD Anderson Cancer Center, Houston, TX.

 

Abstract

  • Objective: To review the evidence on interventions for managing cancer-related fatigue (CRF) and provide evidence-based guidance on approaches to its management.
  • Methods: Nonsystematic review of the literature.
  • Results: Several theories have been proposed to explain the biology of CRF, but there is no single clear mechanism that can be targeted for therapy. The approach to patients begins with screening for fatigue and assessing its intensity, followed by a thorough history and examination to determine whether any reversible medical conditions are contributing to fatigue. Management of underlying medical comorbidities may help some patients. For patients whose fatigue persists, pharmacologic and nonpharmacologic treatment options are available. Pharmacologic options include psychostimulants, such as methylphenidate and modafinil, and corticosteroids. Nonpharmacologic approaches include exercise, cognitive behavior therapy, yoga, acupuncture, and tai chi.
  • Conclusion: We recommend an individualized approach, often with a combination of the available options. Patients need to be evaluated periodically to assess their fatigue, and since cancer-related fatigue affects survivors, long-term follow-up is needed.

Key words: fatigue; cancer; pro-inflammatory cytokines; nonpharmacologic; psychostimulants.

 

Fatigue is a common distressing effect of cancer [1].It impairs the quality of life of patients undergoing active cancer treatment and of post-treatment survivors. The National Comprehensive Cancer Network (NCCN) defines cancer-related fatigue (CRF) as “a distressing, persistent, subjective sense of physical, emotional and/or cognitive tiredness related to cancer or cancer treatment that is not proportional to recent activity and interferes with usual functioning [2].” Differences between CRF and fatigue reported by individuals without cancer are that CRF is more severe and is not relieved by rest. The prevalence of CRF in cancer patients and survivors is highly variable, ranging between 25% and 99% [2,3]. This variability may be secondary to methods used for screening fatigue and characteristics of the patient groups. In this article, we discuss recognition of CRF and approaches to its management.

 

Pathophysiology

The specific pathophysiologic mechanism underlying CRF is unknown, making targeted treatment a challenge. The multidimensional and subjective nature of CRF has limited the development of research methodologies to explain this condition. However, research has been done in both human and animal models, and several theories have been proposed to explain the pathophysiology of CRF. While pro-inflammatory cytokines remain the central factor playing a significant role at multiple levels in CRF, there may be a complex interplay of more than 1 mechanism contributing to fatigue in an individual patient.

Central Nervous System Disturbances

The basal ganglia are known to influence motivation. Lack of motivation and drive may cause failure to complete physical and mental tasks, even with preserved cognitive ability and motor function. In a study of melanoma patients receiving interferon, increased activity of the basal ganglia and the cerebellum resulted in higher fatigue scores [4]. Higher levels of cytokines may alter blood flow to the cerebellum and lead to the perception of fatigue. In a study of 12 patients and matched controls, when patients were asked to perform sustained elbow flexion until they perceived exhaustion, CRF patients perceived physical exhaustion sooner than controls. In CRF patients in this study, muscle fatigue measured by electromyogram was less than that in healthy individuals at the time of exhaustion, suggesting the role of the central nervous system in CRF [5]. However, there is not enough evidence at this time to support central nervous system disturbance as the main contributing factor to fatigue in cancer patients.

Circadian Rhythm Dysregulation

Circadian rhythm is regulated by the suprachiasmatic nucleus in the hypothalamus through cortisol and melatonin. Sleep disturbances occur with disruption of the circadian rhythm. Tumor-related peptides such as epidermal growth factor or alterations in serotonin and cortisol can influence the suprachiasmatic nucleus and the complex signaling pathways [2]. Positive feedback loops that are activated by cortisol under the influence of cytokines may lead to continuous cytokine production and altered circadian rhythm. Bower et al showed that changes in the cortisol curve influence fatigue in breast cancer survivors [6]. These patients had a late evening peak in cortisol levels, compared with an early morning peak in individuals without cancer.

Inhibition of Hypothalamic–Pituitary–Adrenal Axis

The hypothalamic–pituitary–adrenal (HPA) axis regulates the release of the stress hormone cortisol. One of several hypotheses advanced to explain the effect of serotonin and the HPA axis on CRF suggests that lower serotonin levels cause decreased activation of 5-hydroxytrytophan 1-a (5-HT1-a) receptors in the hypothalamus, leading to decreased activity of the HPA axis [6]. The inhibition of the HPA axis may occur with higher levels of serotonin as well [7]. The 5-HT1-a receptors are also triggered by cytokines. However, the correction of serotonin levels by antidepressants was not shown to improve fatigue [8]. Inhibition of the HPA axis can also lead to lower testosterone, progesterone, or estrogen levels, which may indirectly contribute to fatigue [2].

Skeletal Muscle Effect

Chemotherapy- and tumor-related cachexia have a direct effect on the metabolism of skeletal muscles. This effect may lead to impaired adenosine triphosphate (ATP) generation during muscle contraction [9]. ATP infusion improved muscle strength in one trial, but this was not confirmed in another trial [10,11]. Muscle contraction studies showed no differences in the contractile properties of muscles in fatigued patients who failed earlier in motor tasks and healthy controls [12]. This finding suggests that there could be a failure of skeletal muscle activation by the central nervous system or inhibition of skeletal muscle activity. Cytokines and other neurotransmitters activate vagal efferent nerve fibers, which may lead to reflex inhibition in skeletal muscles [13,14].

Pro-inflammatory Cytokines

Tumors or treatment of them may cause tissue injury, which triggers immune cells to release cytokines, signaling the brain to manifest the symptom fatigue. Inflammatory pathways are influenced by psychological, behavioral, and biological factors, which play a role as risk factors in CRF. Interleukin 6 (IL-6), interleukin-1 receptor antagonist, interleukin-1, and tumor necrosis factor (TNF) have been shown to be elevated in fatigued patients being treated for leukemia and non-Hodgkin lymphoma [15]. IL-6 was also associated with increased fatigue in breast cancer survivors [16]. Similar findings were reported in patients undergoing stem cell transplantation and high-dose chemotherapy [17]. Elevated levels of IL-6 and C-reactive protein were also linked to fatigue in terminally ill cancer patients [18,19]. Furthermore, TNF-α signaling was associated with post-chemotherapy fatigue in breast cancer patients [20]. Leukocytes in breast cancer survivors with fatigue also have increased gene expression of pro-inflammatory cytokines, emphasizing the role of cytokines and inflammation in the pathogenesis of CRF [21].

Other Hypotheses

Several other hypotheses for CRF pathogenesis have been proposed. Activation of latent viruses such as Epstein-Barr virus, lack of social support [22], genetic alterations in immune pathway [23], epigenetic changes [24], accumulation of neurotoxic metabolites and depletion of serotonin by indoleamine 2,3-dioxygenase pathway activation [25], elevated vascular endothelial growth factor levels [26], and hypoxia-related organ dysfunction due to anemia or hemoglobin dysfunction [13] all have been postulated to cause CRF.

Approach to Evaluation and Treatment

The evaluation and treatment of CRF involve 4 steps (Figure). First, patients are screened for fatigue, and in the second step those who have fatigue undergo primary evaluation to assess for potential precipitating causes. The third step is implementation of pharmacologic and non-pharmacologic interventions aimed at alleviating or mitigating fatigue. The fourth step involves re-evaluating patients at periodic intervals to recognize and manage changes in fatigue levels. A multidisciplinary approach involving nursing, physical therapy, social work, and nutrition is critical in managing fatigue in these patients. Education and counselling of patients and involvement of the family are essential for effective management.

Screening

Because patients and health care professionals may be unaware of the treatment options available for CRF, patients may not report fatigue levels to their clinicians, and clinicians may not understand the impact of fatigue on their patients’ quality of life. This leads to underrecognition of the problem. The NCCN recommends screening every cancer patient and post-treatment survivor for fatigue [2]. Patients should be screened at their first visit and then at periodic intervals during and after cancer treatment.

Many scales are available to screen patients for CRF in clinical practice and clinical trials [27]. A single item that asks patients to rate their fatigue on a scale from 0 to 10—in which 0 indicates no fatigue, 1 to 3 indicates mild fatigue, 4 to 6 indicates moderate fatigue, 7 to 9 indicates severe fatigue, and 10 indicates the worst fatigue imaginable—is commonly used to screen for CRF [2]. This scale was adapted from the MD Anderson Symptom Inventory scale and is based on a large nationwide study of cancer patients and survivors [28]. The statistically derived cutoff points in this study are consistent with other scales such as the Brief Fatigue Inventory (BFI) and support the cutoff points (4–6 for moderate and ≥ 7 for severe fatigue) used in various fatigue management guidelines. Furthermore, studies of fatigue in cancer patients have revealed a marked decrease in physical function at levels of 7 or higher, suggesting 7 as an optimal cutoff to identify severe fatigue [29,30]. The Visual Analog Scale is another simple-to-use tool that helps in understanding variations in fatigue throughout the course of the day [31]. The 9-item BFI is often used in clinical trials [29]. It measures the severity of fatigue over the previous 24 hours and has been validated in non-English speaking patients [32].

 

 

CRF affects not only the somatic domain, but also the cognitive, behavioral, and affective domains; therefore, multidimensional scales have been developed for screening. One such tool is the Multidimensional Fatigue Inventory, which measures general, physical, mental, and emotional fatigue domains as well as activity and compares them with those of individuals without cancer [33,34]. The Functional Assessment of Cancer Therapy for Fatigue (FACT-F) is a 13-item questionnaire that has been used to measure CRF in clinical trials as well as in patients receiving various treatments [35].

Although many scales are available, the validity of self-reporting on simple fatigue-rating scales is equal to or better than most complex, lengthy scales [36]. Therefore, unidimensional tools such as the numeric rating scale of 0–10 are commonly used in clinical practice. Mild fatigue (0–3) requires periodic re-evaluation, and moderate and severe fatigue need further evaluation and management [37].

Primary Evaluation

This phase involves a focused history and physical examination and assessment of concurrent symptoms and contributing factors.

 

History and Physical Examination

A detailed history of the patient’s malignancy and type of previous and current treatment may help reveal the cause of fatigue. New-onset fatigue or increase in fatigue may be related to the progression of disease in patients with active malignancy or recurrence of cancer in survivors. These patients may require appropriate testing to assess the underlying disease pattern. A detailed review of systems may help identify some of the contributing factors, which are discussed below. A detailed history regarding medications, including over-the-counter drugs, complementary agents, and past and prior cancer therapies, is helpful as medications can contribute to fatigue. For example, opioids may cause drowsiness and fatigue, which could be improved by dose adjustments. A focused history of fatigue should be obtained in all patients with moderate to severe CRF, which includes the onset, pattern, duration, associated or alleviating factors, and interference with functioning, including activities of daily living [37]. Physical examination should focus on identifying signs of organ dysfunction and features of substance or alcohol abuse which may cause poor sleep and fatigue.

 

Assessment of Contributing Factors

The management of fatigue should be multifactorial, with a comprehensive assessment and treatment plan to address all modifiable fatigue etiologies. The Table lists potential contributing factors to fatigue that should be considered when evaluating patients for CRF; several common conditions are discussed below.

Anemia. Anemia has been correlated with fatigue and quality of life. In a study of 4382 cancer patients receiving chemotherapy, quality-of-life measures using FACT-Anemia scores improved with increased hemoglobin levels [38]. Cancer patients may have anemia due to marrow-suppressing effects of chemotherapy and may also have iron deficiency anemia due to blood loss or autoimmune hemolytic anemia. Therefore, a detailed work-up is required to identify the etiology of anemia. Patients with CRF whose anemia is related to chemotherapy or anemia of chronic disease may benefit from red blood cell transfusion or erythropoiesis-stimulating agents (ESAs). ESAs have been studied extensively; however, their use is controversial because of concerns about thromboembolic side effects leading to adverse outcomes [39]. Also, ESA therapy is not recommended in patients with hematologic malignancies. ESA use should be restricted to patients with chemotherapy-related anemia with hemoglobin below 10 mg/dL and should be discontinued in 6 to 8 weeks if patients do not respond [40]. Other patients may benefit from blood transfusions, which were shown to help in patients with hemoglobin levels between 7.5 and 8.5 g/dL [41].

Sleep disturbance. Poor sleep is common in fatigued cancer survivors [42]. Pro-inflammatory cytokines can disrupt the sleep–wake cycle, causing changes in the HPA axis and neuroendocrine system, which in turn may lead to increasing fatigue. Heckler et al showed that improvement in nighttime sleep leads to improvement of fatigue [43]. Cognitive behavioral therapy and sleep hygiene are important in caring for patients with CRF and poor sleep [44]. Taking a warm bath and/or drinking a glass of milk before bedtime, avoiding caffeinated drinks, and avoiding frequent napping in the day might help. Some patients may require medications such as benzodiazepines or non-benzodiazepine hypnotics (eg, zolpidem) to promote sleep [45]. Melatonin agonists are approved for insomnia in the United states, but not in Europe [46].

Malnutrition. Patients with advanced-stage cancer and with cancers affecting the gastrointestinal tract frequently develop mechanical bowel obstructions, especially at the end of their life, which cause malnutrition. Chemotherapy-related nausea and vomiting may also cause poor oral intake and malnutrition, causing fatigue from muscle weakness. Dehydration and electrolyte imbalances frequently occur as a result of poor oral intake, which might worsen fatigue. In our experience, modifying dietary intake with appropriate caloric exchanges with the help of a nutrition expert has lessened fatigue in some patients. However, terminally ill patients are advised to eat based on their comfort.

Medical comorbidities. Congestive heart failure from anthracycline chemotherapy, hypothyroidism after radiation therapy for head and neck cancers, renal failure, or hepatic failure from chemotherapy may indirectly lead to fatigue. Chemotherapy, opioids, and steroids may cause hypogonadism, which can contribute to fatigue in men [47].

 

 

Assessment of Concurrent Symptoms

Depression is common in cancer patients and coexists with pain, insomnia, fatigue, and anxiety as a symptom cluster [48]. A symptom cluster is defined as 2 or more concurrent and interrelated symptoms occurring together; treating of one of these symptoms without addressing other symptoms is not effective [49]. Therefore, screening for and management of depression, anxiety, and insomnia play an important role in the management of CRF.

Physical symptoms due to the tumor or to therapy—such as pain, dyspnea, nausea, and decreased physical activity—may also contribute to fatigue both directly and indirectly. Patients with lung cancer may have hypoxemia, which can contribute to dyspnea with activity and a perception of fatigue. Optimal management of pain and other physical symptoms in patients with cancer may significantly alleviate fatigue [50].

Management

Management of CRF is individualized based on the patient’s clinical status: active cancer treatment, survivor, or end of life. Education and counselling of patients and their caregivers play an important role in CRF. NCCN guidelines recommend focusing on pain control, distress management, energy conservation, physical activity, nutrition, and sleep hygiene.

 

Nonpharmacologic Interventions

Energy conservation. Energy conservation strategies, in which patients are advised to set priorities and realistic expectations, are highly recommended. Some energy-conserving strategies are to pace oneself, delegate and schedule activities at times of peak energy, postpone nonessential activities, attend to 1 activity at a time, structure daily routines, and maintain a diary to identify their peak energy period and structure activities around that time [51,52]. When patients approach the end of life, increasing fatigue may limit their activity level, and they may depend on caregivers for assistance with activities of daily living, monitoring treatment-related adverse effects, and taking medications, especially elderly patients [53].

Cognitive behavioral therapy. Cognitive behavioral therapy (CBT) has been shown to improve CRF during active treatment, and the benefits persist for a minimum of 2 years after therapy [54]. CBT interventions that optimize sleep quality may improve fatigue [55]. More studies are needed to understand whether referral to a psychologist for formal CBT is required. Randomized clinical trials (RCTs) showed patient fatigue education, learned self-care, coping techniques, and balancing rest and activity benefit patients with CRF [56].

Exercise. Physical activity is highly encouraged in patients with CRF. Exercise increases muscle protein synthesis, improves cytokine response, and decreases the rate of sarcopenia in healthy populations [57]. Studies have shown that exercise helps CRF at all phases of the cancer journey, including radiation therapy, chemotherapy, and survivorship [58]. Some patients may feel less motivated to exercise and may not believe that exercise is possible or could potentially help them. Counselling is needed for such patients.

Older cancer survivors have a decline in their functional capacity and reduced muscle mass. Exercise can improve cardiorespiratory fitness, muscle strength, and body composition [57]. Exercise not only helps at the cellular level but also has psychosocial benefits from improved self-esteem. Therefore, exercise may be recommended not only for younger patients, but also in the older population, who may have comorbidities and less motivation than younger patients.

In a meta-analysis of 56 randomized controlled trials involving 4068 participants, aerobic exercise was found to have beneficial effects on CRF for patients during and after chemotherapy, specifically for patients with solid tumors [59]. In another meta-analysis of breast and prostate cancer survivors, a combination of aerobic exercise with resistance training (3–6 metabolic equivalents, 60%–80% range of motion) was shown to reduce CRF more than aerobic exercise alone [60]. This effect was also shown in an RCT of 160 patients with stage 0 to III breast cancer undergoing radiation therapy [61]. The control group in this study had a group-based non-exercise intervention/relaxation; therefore, the study showed that the effect of resistance training extends beyond the psychosocial benefits of group-based interventions. The intervention comprised 8 progressive machine-based resistance exercises (3 sets, 8–12 repetitions at 60%–80% of 1 repetition maximum) for 60 minutes twice weekly for 12 weeks. However, fatigue assessment questionnaire scores showed benefits in the physical fatigue but not the affective and cognitive components.

The American Society of Clinical Oncology’s guidelines for cancer survivors with fatigue recommends 150 minutes of moderate aerobic exercise (eg, fast walking, cycling, or swimming) per week, with 2 or 3 sessions of strength training per week [62]. An individualized approach to exercise is recommended, as patients’ ability to perform certain types of exercises may be limited by thrombocytopenia, neutropenia, or lytic bone metastasis. Routine use of pre-exercise cardiovascular testing is not recommended but may be considered in high-risk populations, especially patients with risk factors for coronary heart disease and diabetes [63]. Patients withcomorbidities, substantial deconditioning, functional and anatomic defects, or recent major surgery may benefit from referral to physical therapy [37]. Patients near end of life may also benefit from an exercise program, as demonstrated in several studies that showed benefit in CRF and quality of life [64,65]. We recommend that physicians use their best clinical judgement in suggesting the type and intensity of exercise program, as it may not be feasible in some patients.

Mind-body interventions. Mindfulness-based stress reduction (MBSR) has shown promise in breast cancer survivors, who reported immediate improvements in fatigue severity that continued up to 6 weeks after cessation of the training [66]. Prior studies had similar findings, suggesting that MBSR modestly decreases fatigue and sleep disturbances and has a greater effect on the degree to which symptoms interfere with many facets of life [67].

Yoga. A study of a yoga intervention showed a benefit in older cancer survivors [68]. In breast cancer patients undergoing chemotherapy, yoga was shown to benefit not only physical fatigue, but also cognitive fatigue [69]. DVD-based yoga had benefits similar to strengthening exercises in a study of 34 early-stage breast cancer survivors with CRF [70]. More studies are needed in men and patients and survivors of other cancers, as most studies of yoga were conducted in women with breast cancer.

Tai chi/qigong. Like yoga, tai chi and qigong are practices of meditative movement. These practices use postures or movements with a focus on breath and a meditative state to bring about deep states of relaxation. Qigong is a series of simple, repeated practices including body posture/movement, breath practice, and meditation performed in synchrony. Tai chi easy (TCE) is a simplified set of common, repetitive tai chi movements. In a trial, qigong/TCE was compared with sham qigong, which had physical movements but no breathing or meditative practice. Breast cancer survivors in the qigong/TCE group had improved fatigue scores, and the effect persisted for 3 months [71]. Additional research is needed in this area.

Acupuncture. An RCT in breast cancer patients with CRF showed an improvement in the mean general fatigue score (per the Multidimensional Fatigue Inventory) in patients who received acupuncture versus those who did not (−3.11 [95% confidence interval −3.97 to −2.25]; P < 0.001) at 6 weeks. Improvements were seen in both the mental and physical aspects of fatigue [72]. However, Deng et al noted that true acupuncture was no more effective than sham acupuncture for reducing post-chemotherapy chronic fatigue [73]. Presently, there is not sufficient evidence to evaluate the benefits of acupuncture in CRF.

Other modalities. Massage therapy, music therapy, hypnosis, therapeutic touch, biofield therapies, relaxation, and reiki are other therapies for which few studies have been done, with mixed results, and additional research is needed [74]. Currently, there are not sufficient data to recommend any of these modalities.

Pharmacologic Interventions

Psychostimulants. Methylphenidate and modafinil are psychostimulants or wakefulness-promoting agents. Pilot studies showed benefit from methylphenidate and modafinil in CRF [75–77], but RCTs have yielded mixed results. Therefore, in patients with severe fatigue during cancer therapy, the initial management strategy involves evaluation and treatment of medical conditions such as anemia and a trial of non-pharmacological strategies as discussed above. If symptoms persist, then a therapeutic trial of a psychostimulant may be considered per NCCN guidelines for patients undergoing active cancer treatment [37].

Methylphenidate directly stimulates adrenergic receptors and indirectly releases dopamine and norepinephrine from presynaptic terminals, which may explain why the drug benefits patients receiving opioid-induced sedation. It is a commonly studied psychostimulant, though its mechanism of action in CRF is unclear. RCTs of methylphenidate have resulted in a wide range of findings due to the heterogeneity of study populations and variations in the dosage of methylphenidate. A meta-analysis of 7 studies indicates that methylphenidate benefitted the subgroup of patients with CRF [78]. Likewise, in an analysis of 5 RCTs, Minton et al showed a benefit of psychostimulants in fatigue compared with placebo [79]. However, another study of methylphenidate in patients with CRF showed a benefit only in patients with severe fatigue or advanced disease [80]. Methylphenidate was found to benefit cancer patients receiving opioid-induced sedation, as methylphenidate promotes wakefulness, though fatigue was not studied specifically [81]. In a trial with 30 hospice patients in which the methylphenidate dose was titrated based on response and adverse effects, Kerr at al found that the drug improved fatigue in a dose-dependent manner [82]. However, a study in patients with CRF at the University of Texas MD Anderson Cancer Center found no significant difference in BFI scores between patients receiving methylphenidate and those receiving placebo at the end of 2 weeks of treatment [83]. Also, other RCTs in patients undergoing adjuvant chemotherapy for breast cancer [84] and patients receiving radiation therapy for brain tumors [85] failed to demonstrate the efficacy of methylphenidate in CRF. It should be used cautiously after ruling out other causes of fatigue. The drug is overall well tolerated and side effects include headache and nausea.

Modafinil is a non-amphetamine psychostimulant that has been approved for the treatment of narcolepsy. In a trial studying the effect of modafinil on patients receiving docetaxel-based chemotherapy for metastatic breast or prostate cancer, there was a modest but not statistically significant improvement in fatigue scores on the MD Anderson Symptom Inventory compared with placebo. Nausea and vomiting were higher in the modafinil arm than in the placebo arm [86]. Similarly, modafinil was not superior to placebo for CRF in 208 patients with non-squamous cell lung cancer not undergoing chemotherapy or radiation [87]. A placebo effect was also noted in patients with multiple myeloma [88] and patients with primary brain tumors [89]. In a phase 3, multicenter, randomized, placebo-controlled, double-blind clinical trial of modafinil for CRF in 867 patients undergoing chemotherapy, there was a reduction in fatigue only for patients with severe baseline fatigue, with no significant effect on mild to moderate fatigue [90]. In another recent study, modafinil was shown to reduce depressive symptoms only in patients with severe fatigue (BFI item 3 score ≥ 7) [91]. This finding is consistent with previous studies showing benefit in patients with high baseline fatigue, but additional RCTs are needed to provide clarity. NCCN guidelines do not recommend the use of modafinil to treat CRF [37].

Other pharmacologic interventions. Corticosteroids are often used for symptom control in cancer patients. These drugs have anti-inflammatory effects through their modulation of pro-inflammatory cytokines [92]. In a RCT evaluating the efficacy of corticosteroids, patients receiving dexamethasone (4 mg twice daily) experienced significant improvement in their FACT-F scores compared with patients receiving placebo [93]. A similar benefit in fatigue was demonstrated in another study of methylprednisolone (32 mg daily) versus placebo [94]. Despite the benefits of steroids, their adverse effects, such as mood swings, gastritis, hyperglycemia, and immune suppression, limit their long-term use. Therefore, the use of steroids should be restricted to terminally ill fatigued patients with other symptoms such as anorexia, brain metastasis, or pain related to bone metastasis [37].

Testosterone replacement has been shown to diminish fatigue in non-cancer patients. Many men with advanced cancer have hypogonadism leading to low serum testosterone, which may cause fatigue. In a small trial in which cancer patients with hypogonadism received intramuscular testosterone every 14 days or placebo, the group receiving testosterone showed improvement in FACT-F scores, but there was no significant difference in FACT-F scores between the 2 groups [95].

Antidepressants have failed to demonstrate benefit in CRF without depression [8]. However, if a patient has both fatigue and depression, antidepressants may help [96]. A selective serotonin receptor inhibitor is recommended as a first-line antidepressant [97]. Patients with cancer are often receiving multiple medications, and medication interactions should be considered to prevent adverse events such as serotonin syndrome.

 

 

Complementary and Alternative Supplements

Studies using vitamin supplementation have been inconclusive in patients with CRF [74]. The use of other dietary supplements has yielded mixed results, and coenzyme Q has shown no benefit for patients with CRF [98].

The benefit of ginseng was studied in a RCT involving 364 patients with CRF. There was an improvement in Multidimensional Fatigue Symptom Inventory-short form (MFSI-SF) scores at 8 weeks in patients receiving 2000 mg of Wisconsin ginseng compared with patients receiving placebo [99]. Patients on active treatment had greater improvement as compared to the post-treatment group in this trial. In another study of high-dose panax ginseng (ginseng root) at 800 mg daily for 29 days, patients had improvement of CRF as well as overall quality of life, appetite, and sleep at night. It was also well tolerated with few adverse effects [100]. Interaction with warfarin, calcium channel blockers, antiplatelet agents, thrombolytic agents, imatinib, and other agents may occur; therefore, ginseng must be used with careful monitoring in selected patients. There is not enough evidence at this time to support the routine use of ginseng in CRF.

The seed extract of the guarana plant (Paullinia cupana) traditionally has been used as a stimulant. An improvement in fatigue scores was seen with the use of oral guarana (100 mg daily) at the end of 21 days in breast cancer patients receiving chemotherapy [101]. Further studies are needed for these results to be generalized and to understand the adverse effects and interaction profile of guarana.

 

Re-evaluation

Patients who have completed cancer treatment must be monitored for fatigue over the long term, as fatigue may exist beyond the period of active treatment. Many studies have shown fatigue in breast cancer survivors, and fatigue has been demonstrated in survivors of colorectal, lung, and prostate cancers as well as myeloproliferative neoplasms [28]. Therefore, it is important to screen patients for fatigue during follow-up visits. There are currently no studies evaluating the long-term treatment of fatigue. In our experience, the timing of follow-up depends on the level of fatigue and interventions prescribed. Once fatigue is stabilized to a level with which the patient is able to cope, the time interval for follow up may be lengthened. Annual visits may suffice in patients with mild fatigue. Follow-up of patients with moderate to severe fatigue depends on the level of fatigue, the ability to cope, choice of treatment, and presence of contributing factors.

Conclusion

CRF is a complex condition that places a significant burden on patients and caregivers, resulting in emotional distress, poor functioning, and suffering. Fatigue can occur before, during, and long after cancer treatment. The approach to CRF begins with screening for and educating patients and their caregivers about the symptoms. Many screening scales are available that may be used to follow patients’ progress over time. The evaluation and management of contributing conditions may help improve fatigue. If the fatigue persists, an individualized approach with a combination of nonpharmacologic and pharmacologic approaches should be considered. More research is needed to understand brain signaling pathways, cytokine changes, and genomic changes in cancer patients with fatigue. Though many hypotheses have been proposed, to date there is no biological marker to assess this condition. Biomarker research needs to be advanced to help to identify patients at risk for fatigue. As cytokines have a major role in CRF, targeted therapy to block cytokine pathways may also be explored in the future.

 

Acknowledgment: Bryan Tutt provided editorial assistance.

Corresponding author: Carmelita P. Escalante, MD, The University of Texas MD Anderson Cancer Center, 1400 Pressler St., Houston, TX 77030, [email protected].

Financial disclosures: None.

References

1. Scherber RM, Kosiorek HE, Senyak Z, et al. Comprehensively understanding fatigue in patients with myeloproliferative neoplasms. Cancer 2016;122:477–85.

2. Neefjes EC, van der Vorst MJ, Blauwhoff-Buskermolen S, Verheul HM. Aiming for a better understanding and management of cancer-related fatigue. Oncologist 2013;18:1135–43.

3. Radbruch L, Strasser F, Elsner F, et al. Fatigue in palliative care patients -- an EAPC approach. Palliat Med 2008;22:13–32.

4. Capuron L, Pagnoni G, Demetrashvili MF, et al. Basal ganglia hypermetabolism and symptoms of fatigue during interferon-alpha therapy. Neuropsychopharmacology 2007;32:2384–92.

5. Kisiel-Sajewicz K, Siemionow V, Seyidova-Khoshknabi D, et al. Myoelectrical manifestation of fatigue less prominent in patients with cancer related fatigue. PLoS One 2013;8:e83636.

6. Bower JE, Ganz PA, Aziz N. Altered cortisol response to psychologic stress in breast cancer survivors with persistent fatigue. Psychosom Med 2005;67:277–80.

7. Barsevick A, Frost M, Zwinderman A, et al. I’m so tired: biological and genetic mechanisms of cancer-related fatigue. Qual Life Res 2010;19:1419–27.

8. Morrow GR, Hickok JT, Roscoe JA, et al. Differential effects of paroxetine on fatigue and depression: a randomized, double-blind trial from the University of Rochester Cancer Center Community Clinical Oncology Program. J Clin Oncol 2003;21:4635–41.

9. Fontes-Oliveira CC, Busquets S, Toledo M, et al. Mitochondrial and sarcoplasmic reticulum abnormalities in cancer cachexia: altered energetic efficiency? Biochim Biophys Acta 2013;1830:2770–8.

10. Agteresch HJ, Dagnelie PC, van der Gaast A, et al. Randomized clinical trial of adenosine 5’-triphosphate in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 2000;92:321–8.

11. Beijer S, Hupperets PS, van den Borne BE, et al. Randomized clinical trial on the effects of adenosine 5’-triphosphate infusions on quality of life, functional status, and fatigue in preterminal cancer patients. J Pain Symptom Manage 2010;40:520–30.

12. Kisiel-Sajewicz K, Davis MP, Siemionow V, et al. Lack of muscle contractile property changes at the time of perceived physical exhaustion suggests central mechanisms contributing to early motor task failure in patients with cancer-related fatigue. J Pain Symptom Manage 2012;44:351–61.

13. Ryan JL, Carroll JK, Ryan EP, et al. Mechanisms of cancer-related fatigue. Oncologist 2007;12 Suppl 1:22–34.

14. Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer 2008;8:887–99.

15. Wang XS, Giralt SA, Mendoza TR, et al. Clinical factors associated with cancer-related fatigue in patients being treated for leukemia and non-Hodgkin’s lymphoma. J Clin Oncol 2002;20:1319–28.

16. Collado-Hidalgo A, Bower JE, Ganz PA, et al. Inflammatory biomarkers for persistent fatigue in breast cancer survivors. Clin Cancer Res 2006;12:2759–66.

17. Wang XS, Shi Q, Williams LA, et al. Serum interleukin-6 predicts the development of multiple symptoms at nadir of allogeneic hematopoietic stem cell transplantation. Cancer 2008;113:2102–9.

18. Inagaki M, Isono M, Okuyama T, et al. Plasma interleukin-6 and fatigue in terminally ill cancer patients. J Pain Symptom Manage 2008;35:153–61.

19. Laird BJ, McMillan DC, Fayers P, et al. The systemic inflammatory response and its relationship to pain and other symptoms in advanced cancer. Oncologist 2013;18:1050–5.

20. Bower JE, Ganz PA, Irwin MR, et al. Inflammation and behavioral symptoms after breast cancer treatment: do fatigue, depression, and sleep disturbance share a common underlying mechanism? J Clin Oncol 2011;29:3517–22.

21. Whistler T, Taylor R, Craddock RC, et al. Gene expression correlates of unexplained fatigue. Pharmacogenomics 2006;7:395–405.

22. Fagundes CP, Bennett JM, Alfano CM, et al. Social support and socioeconomic status interact to predict Epstein-Barr virus latency in women awaiting diagnosis or newly diagnosed with breast cancer. Health Psychol 2012;31:11–19.

23. Landmark-Hoyvik H, Reinertsen KV, Loge JH, et al. Alterations of gene expression in blood cells associated with chronic fatigue in breast cancer survivors. Pharmacogenomics J 2009;9:333–40.

24. Smith AK, Conneely KN, Pace TW, et al. Epigenetic changes associated with inflammation in breast cancer patients treated with chemotherapy. Brain Behav Immun 2014;38:227–36.

25. Kim S, Miller BJ, Stefanek ME, Miller AH. Inflammation-induced activation of the indoleamine 2,3-dioxygenase pathway: Relevance to cancer-related fatigue. Cancer 2015;121:2129–36.

26. Mills PJ, Parker B, Dimsdale JE, et al. The relationship between fatigue and quality of life and inflammation during anthracycline-based chemotherapy in breast cancer. Biol Psychol 2005;69:85–96.

27. Jean-Pierre P, Figueroa-Moseley CD, Kohli S, et al. Assessment of cancer-related fatigue: implications for clinical diagnosis and treatment. Oncologist 2007;12 Suppl 1:11–21.

28. Wang XS, Zhao F, Fisch MJ, et al. Prevalence and characteristics of moderate to severe fatigue: a multicenter study in cancer patients and survivors. Cancer 2014;120:425–32.

29. Mendoza TR, Wang XS, Cleeland CS, et al. The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory. Cancer 1999;85:1186–96.

30. Mendoza ME, Capafons A, Gralow JR, et al. Randomized controlled trial of the Valencia model of waking hypnosis plus CBT for pain, fatigue, and sleep management in patients with cancer and cancer survivors. Psychooncology 2016 Jul 28.

31. Glaus A. Assessment of fatigue in cancer and non-cancer patients and in healthy individuals. Support Care Cancer 1993;1:305–15.

32. Seyidova-Khoshknabi D, Davis MP, Walsh D. A systematic review of cancer-related fatigue measurement questionnaires. Am J Hosp Palliat Care 2011;28:119–29.

33. Holzner B, Kemmler G, Greil R, et al. The impact of hemoglobin levels on fatigue and quality of life in cancer patients. Ann Oncol 2002;13:965–73.

34. Stein KD, Jacobsen PB, Blanchard CM, Thors C. Further validation of the multidimensional fatigue symptom inventory-short form. J Pain Symptom Manage 2004;27:14–23.

35. Hwang SS, Chang VT, Rue M, Kasimis B. Multidimensional independent predictors of cancer-related fatigue. J Pain Symptom Manage 2003;26:604–14.

36. Peterspm DR. Scope and generality of verbally defined personality factors. Psychol Rev 1965;72:48–59.

37. Berger AM, Abernethy AP, Atkinson A, et al. NCCN Clinical Practice Guidelines Cancer-related fatigue. J Natl Compr Canc Netw 2010;8:904–31.

38. Crawford J, Cella D, Cleeland CS, et al. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002;95:888–95.

39. Tonia T, Mettler A, Robert N, et al. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst Rev 2012;12:CD003407.

40. Rizzo JD, Brouwers M, Hurley P, et al. American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood 2010;116:4045–59.

41. Preston NJ, Hurlow A, Brine J, Bennett MI. Blood transfusions for anaemia in patients with advanced cancer. Cochrane Database Syst Rev 2012(2):CD009007.

42. Minton O, Stone PC. A comparison of cognitive function, sleep and activity levels in disease-free breast cancer patients with or without cancer-related fatigue syndrome. BMJ Support Palliat Care 2012;2:231–8.

43. Heckler CE, Garland SN, Peoples AR, et al. Cognitive behavioral therapy for insomnia, but not armodafinil, improves fatigue in cancer survivors with insomnia: a randomized placebo-controlled trial. Support Care Cancer 2016;24:2059–66.

44. Howell D, Oliver TK, Keller-Olaman S, et al. Sleep disturbance in adults with cancer: a systematic review of evidence for best practices in assessment and management for clinical practice. Ann Oncol 2014;25:791–800.

45. Wilt TJ, MacDonald R, Brasure M, et al. Pharmacologic treatment of insomnia disorder: an evidence report for a clinical practice guideline by the American College of Physicians. Ann Intern Med 2016;165:103–12.

46. Kuriyama A, Honda M, Hayashino Y. Ramelteon for the treatment of insomnia in adults: a systematic review and meta-analysis. Sleep Med 2014;15:385–92.

47. Strasser F, Palmer JL, Schover LR, et al. The impact of hypogonadism and autonomic dysfunction on fatigue, emotional function, and sexual desire in male patients with advanced cancer: a pilot study. Cancer 2006;107:2949–57.

48. Agasi-Idenburg SC, Thong MS, Punt CJ, et al. Comparison of symptom clusters associated with fatigue in older and younger survivors of colorectal cancer. Support Care Cancer 2017;25:625–32.

49. Miaskowski C, Aouizerat BE. Is there a biological basis for the clustering of symptoms? Semin Oncol Nurs 2007;23:99–105.

50. de Raaf PJ, de Klerk C, Timman R, et al. Systematic monitoring and treatment of physical symptoms to alleviate fatigue in patients with advanced cancer: a randomized controlled trial. J Clin Oncol 2013;31:716–23.

51. Barsevick AM, Whitmer K, Sweeney C, Nail LM. A pilot study examining energy conservation for cancer treatment-related fatigue. Cancer Nurs 2002;25:333–41.

52. Barsevick AM, Dudley W, Beck S, et a;. A randomized clinical trial of energy conservation for patients with cancer-related fatigue. Cancer 2004;100:1302–10.

53. Luciani A, Jacobsen PB, Extermann M, et al. Fatigue and functional dependence in older cancer patients. Am J Clin Oncol 2008;31:424–30.

54. Abrahams HJ, Gielissen MF, Goedendorp MM, et al. A randomized controlled trial of web-based cognitive behavioral therapy for severely fatigued breast cancer survivors (CHANGE-study): study protocol. BMC Cancer 2015;15:765.

55. Quesnel C, Savard J, Simard S, et al. Efficacy of cognitive-behavioral therapy for insomnia in women treated for nonmetastatic breast cancer. J Consult Clin Psychol 2003;71:189–200.

56. Goedendorp MM, Gielissen MF, Verhagen CA, Bleijenberg G. Psychosocial interventions for reducing fatigue during cancer treatment in adults. Cochrane Database Syst Rev 2009(1):CD006953.

57. Greiwe JS, Cheng B, Rubin DC, et al. Resistance exercise decreases skeletal muscle tumor necrosis factor alpha in frail elderly humans. FASEB J 2001;15:475–82.

58. Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev 2016;(9):CD005001.

59. Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database Syst Rev 2012;(11):CD006145.

60. Brown JC, Huedo-Medina TB, Pescatello LS, et al. Efficacy of exercise interventions in modulating cancer-related fatigue among adult cancer survivors: a meta-analysis. Cancer Epidemiol Biomarkers Prev 2011;20:123–33.

61. Steindorf K, Schmidt ME, Klassen O, et al. Randomized, controlled trial of resistance training in breast cancer patients receiving adjuvant radiotherapy: results on cancer-related fatigue and quality of life. Ann Oncol 2014;25:2237–43.

62. Bower JE, Bak K, Berger A, et al. Screening, assessment, and management of fatigue in adult survivors of cancer: an American Society of Clinical oncology clinical practice guideline adaptation. J Clin Oncol 2014;32:1840–50.

63. Kenjale AA, Hornsby WE, Crowgey T, et al. Pre-exercise participation cardiovascular screening in a heterogeneous cohort of adult cancer patients. Oncologist 2014;19:999–1005.

64. Oldervoll LM, Loge JH, Paltiel H, et al. The effect of a physical exercise program in palliative care: A phase II study. J Pain Symptom Manage 2006;31:421–30.

65. Porock D, Kristjanson LJ, Tinnelly K, et al. An exercise intervention for advanced cancer patients experiencing fatigue: a pilot study. J Palliat Care 2000;16:30–6.

66. Lengacher CA, Kip KE, Reich RR, et al. A cost-effective mindfulness stress reduction program: a randomized control trial for breast cancer survivors. Nursing Econ 2015;33:210–8, 32.

67. Lengacher CA, Reich RR, Post-White J, et al. Mindfulness based stress reduction in post-treatment breast cancer patients: an examination of symptoms and symptom clusters. J Behav Med 2012;35:86–94.

68. Sprod LK, Fernandez ID, Janelsins MC, et al. Effects of yoga on cancer-related fatigue and global side-effect burden in older cancer survivors. J Geriatr Oncol 2015;6:8–14.

69. Wang G, Wang S, Jiang P, Zeng C. [Effect of Yoga on cancer related fatigue in breast cancer patients with chemotherapy]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2014;39:1077–82.

70. Stan DL, Croghan KA, Croghan IT, et al. Randomized pilot trial of yoga versus strengthening exercises in breast cancer survivors with cancer-related fatigue. Support Care Cancer 2016;24:4005–15.

71. Larkey LK, Roe DJ, Weihs KL, et al. Randomized controlled trial of Qigong/Tai Chi Easy on cancer-related fatigue in breast cancer survivors. Ann Behav Med 2015;49:165–76.

72. Molassiotis A, Bardy J, Finnegan-John J, et al. Acupuncture for cancer-related fatigue in patients with breast cancer: a pragmatic randomized controlled trial. J Clin Oncol 2012;30:4470–6.

73. Deng G, Chan Y, Sjoberg D, et al. Acupuncture for the treatment of post-chemotherapy chronic fatigue: a randomized, blinded, sham-controlled trial. Support Care Cancer 2013;21:1735–41.

74. Finnegan-John J, Molassiotis A, Richardson A, Ream E. A systematic review of complementary and alternative medicine interventions for the management of cancer-related fatigue. Integr Cancer Ther 2013;12:276–90.

75. Schwartz AL, Thompson JA, Masood N. Interferon-induced fatigue in patients with melanoma: a pilot study of exercise and methylphenidate. Oncol Nurs Forum 2002;29:E85–90.

76. Spathis A, Dhillan R, Booden D, et al. Modafinil for the treatment of fatigue in lung cancer: a pilot study. Palliat Med 2009;23:325–31.

77. Blackhall L, Petroni G, Shu J, et al. A pilot study evaluating the safety and efficacy of modafinal for cancer-related fatigue. J Palliat Med 2009;12:433–9.

78. Qu D, Zhang Z, Yu X, et al. Psychotropic drugs for the management of cancer-related fatigue: a systematic review and meta-analysis. Eur J Cancer Care (Engl) 2015;25:970–9.

79. Minton O, Richardson A, Sharpe M, et al. Drug therapy for the management of cancer-related fatigue. Cochrane Database Syst Rev 2010(7):CD006704.

80. Moraska AR, Sood A, Dakhil SR, et al. Phase III, randomized, double-blind, placebo-controlled study of long-acting methylphenidate for cancer-related fatigue: North Central Cancer Treatment Group NCCTG-N05C7 trial. J Clin Oncol 2010;28:3673–9.

81. Bruera E, Driver L, Barnes EA, et al. Patient-controlled methylphenidate for the management of fatigue in patients with advanced cancer: a preliminary report. J Clin Oncol 2003;21:4439–43.

82. Kerr CW, Drake J, Milch RA, et al. Effects of methylphenidate on fatigue and depression: a randomized, double-blind, placebo-controlled trial. J Pain Symptom Manage 2012;43:68–77.

83. Escalante CP, Meyers C, Reuben JM, et al. A randomized, double-blind, 2-period, placebo-controlled crossover trial of a sustained-release methylphenidate in the treatment of fatigue in cancer patients. Cancer J 2014;20:8–14.

84. Mar Fan HG, Clemons M, Xu W, et al. A randomised, placebo-controlled, double-blind trial of the effects of d-methylphenidate on fatigue and cognitive dysfunction in women undergoing adjuvant chemotherapy for breast cancer. Support Care Cancer 2008;16:577–83.

85. Butler JM Jr, Case LD, Atkins J, et al. A phase III, double-blind, placebo-controlled prospective randomized clinical trial of d-threo-methylphenidate HCl in brain tumor patients receiving radiation therapy. Int J Radiat Oncol Biol Phys 2007;69:1496–501.

86. Hovey E, de Souza P, Marx G, et al. Phase III, randomized, double-blind, placebo-controlled study of modafinil for fatigue in patients treated with docetaxel-based chemotherapy. Support Care Cancer 2014;22:1233–42.

87. Spathis A, Fife K, Blackhall F, et al. Modafinil for the treatment of fatigue in lung cancer: results of a placebo-controlled, double-blind, randomized trial. J Clin Oncol 2014;32:1882–8.

88. Berenson JR, Yellin O, Shamasunder HK, et al. A phase 3 trial of armodafinil for the treatment of cancer-related fatigue for patients with multiple myeloma. Support Care Cancer 2015;23:1503–12.

89. Boele FW, Douw L, de Groot M, et al. The effect of modafinil on fatigue, cognitive functioning, and mood in primary brain tumor patients: a multicenter randomized controlled trial. Neuro Oncol 2013;15:1420–8.

90. Jean-Pierre P, Morrow GR, Roscoe JA, et al. A phase 3 randomized, placebo-controlled, double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631 patients receiving chemotherapy: a University of Rochester Cancer Center Community Clinical Oncology Program Research base study. Cancer 2010;116:3513–20.

91. Conley CC, Kamen CS, Heckler CE, et al. Modafinil moderates the relationship between cancer-related fatigue and depression in 541 patients receiving chemotherapy. J Clin Psychopharmacol 2016;36:82–5.

92. Brattsand R, Linden M. Cytokine modulation by glucocorticoids: mechanisms and actions in cellular studies. Aliment Pharmacol Ther 1996;10 Suppl 2:81–90.

93. Yennurajalingam S, Frisbee-Hume S, Palmer JL, et al. Reduction of cancer-related fatigue with dexamethasone: a double-blind, randomized, placebo-controlled trial in patients with advanced cancer. J Clin Oncol 2013;31:3076–82.

94. Bruera E, Roca E, Cedaro L, et al. Action of oral methylprednisolone in terminal cancer patients: a prospective randomized double-blind study. Cancer Treat Rep 1985;69:751–4.

95. Pulivarthi K, Dev R, Garcia J, et al. Testosterone replacement for fatigue in male hypogonadic patients with advanced cancer: A preliminary double-blind placebo-controlled trial. J Clin Oncol 2012;30 (suppl). Abstract e19643.

96. Palesh OG, Mustian KM, Peppone LJ, et al. Impact of paroxetine on sleep problems in 426 cancer patients receiving chemotherapy: a trial from the University of Rochester Cancer Center Community Clinical Oncology Program. Sleep Med 2012;13:1184–90.

97. Thekdi SM, Trinidad A, Roth A. Psychopharmacology in Cancer. Curr Psychiatry Rep 2014;17:529.

98. Lesser GJ. Case D, Stark N, et al. A randomized, double-blind, placebo-controlled study of oral coenzyme Q10 to relieve self-reported treatment-related fatigue in newly diagnosed patients with breast cancer. J Support Oncol 2013;11:31–42.

99. Barton DL, Liu H, Dakhil SR, et al. Wisconsin Ginseng (Panax quinquefolius) to improve cancer-related fatigue: a randomized, double-blind trial, N07C2. J Natl Cancer Inst 2013;105:1230–8.

100. Yennurajalingam S, Reddy A, Tannir NM, et al. High-dose Asian ginseng (panax ginseng) for cancer-related fatigue: a preliminary report. Integr Cancer Ther 2015;14:419–27.

101. Howell D, Keller-Olaman S, Oliver TK, et al. A pan-Canadian practice guideline and algorithm: screening, assessment, and supportive care of adults with cancer-related fatigue. Curr Oncol 2013;20:e233–46.

References

1. Scherber RM, Kosiorek HE, Senyak Z, et al. Comprehensively understanding fatigue in patients with myeloproliferative neoplasms. Cancer 2016;122:477–85.

2. Neefjes EC, van der Vorst MJ, Blauwhoff-Buskermolen S, Verheul HM. Aiming for a better understanding and management of cancer-related fatigue. Oncologist 2013;18:1135–43.

3. Radbruch L, Strasser F, Elsner F, et al. Fatigue in palliative care patients -- an EAPC approach. Palliat Med 2008;22:13–32.

4. Capuron L, Pagnoni G, Demetrashvili MF, et al. Basal ganglia hypermetabolism and symptoms of fatigue during interferon-alpha therapy. Neuropsychopharmacology 2007;32:2384–92.

5. Kisiel-Sajewicz K, Siemionow V, Seyidova-Khoshknabi D, et al. Myoelectrical manifestation of fatigue less prominent in patients with cancer related fatigue. PLoS One 2013;8:e83636.

6. Bower JE, Ganz PA, Aziz N. Altered cortisol response to psychologic stress in breast cancer survivors with persistent fatigue. Psychosom Med 2005;67:277–80.

7. Barsevick A, Frost M, Zwinderman A, et al. I’m so tired: biological and genetic mechanisms of cancer-related fatigue. Qual Life Res 2010;19:1419–27.

8. Morrow GR, Hickok JT, Roscoe JA, et al. Differential effects of paroxetine on fatigue and depression: a randomized, double-blind trial from the University of Rochester Cancer Center Community Clinical Oncology Program. J Clin Oncol 2003;21:4635–41.

9. Fontes-Oliveira CC, Busquets S, Toledo M, et al. Mitochondrial and sarcoplasmic reticulum abnormalities in cancer cachexia: altered energetic efficiency? Biochim Biophys Acta 2013;1830:2770–8.

10. Agteresch HJ, Dagnelie PC, van der Gaast A, et al. Randomized clinical trial of adenosine 5’-triphosphate in patients with advanced non-small-cell lung cancer. J Natl Cancer Inst 2000;92:321–8.

11. Beijer S, Hupperets PS, van den Borne BE, et al. Randomized clinical trial on the effects of adenosine 5’-triphosphate infusions on quality of life, functional status, and fatigue in preterminal cancer patients. J Pain Symptom Manage 2010;40:520–30.

12. Kisiel-Sajewicz K, Davis MP, Siemionow V, et al. Lack of muscle contractile property changes at the time of perceived physical exhaustion suggests central mechanisms contributing to early motor task failure in patients with cancer-related fatigue. J Pain Symptom Manage 2012;44:351–61.

13. Ryan JL, Carroll JK, Ryan EP, et al. Mechanisms of cancer-related fatigue. Oncologist 2007;12 Suppl 1:22–34.

14. Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer 2008;8:887–99.

15. Wang XS, Giralt SA, Mendoza TR, et al. Clinical factors associated with cancer-related fatigue in patients being treated for leukemia and non-Hodgkin’s lymphoma. J Clin Oncol 2002;20:1319–28.

16. Collado-Hidalgo A, Bower JE, Ganz PA, et al. Inflammatory biomarkers for persistent fatigue in breast cancer survivors. Clin Cancer Res 2006;12:2759–66.

17. Wang XS, Shi Q, Williams LA, et al. Serum interleukin-6 predicts the development of multiple symptoms at nadir of allogeneic hematopoietic stem cell transplantation. Cancer 2008;113:2102–9.

18. Inagaki M, Isono M, Okuyama T, et al. Plasma interleukin-6 and fatigue in terminally ill cancer patients. J Pain Symptom Manage 2008;35:153–61.

19. Laird BJ, McMillan DC, Fayers P, et al. The systemic inflammatory response and its relationship to pain and other symptoms in advanced cancer. Oncologist 2013;18:1050–5.

20. Bower JE, Ganz PA, Irwin MR, et al. Inflammation and behavioral symptoms after breast cancer treatment: do fatigue, depression, and sleep disturbance share a common underlying mechanism? J Clin Oncol 2011;29:3517–22.

21. Whistler T, Taylor R, Craddock RC, et al. Gene expression correlates of unexplained fatigue. Pharmacogenomics 2006;7:395–405.

22. Fagundes CP, Bennett JM, Alfano CM, et al. Social support and socioeconomic status interact to predict Epstein-Barr virus latency in women awaiting diagnosis or newly diagnosed with breast cancer. Health Psychol 2012;31:11–19.

23. Landmark-Hoyvik H, Reinertsen KV, Loge JH, et al. Alterations of gene expression in blood cells associated with chronic fatigue in breast cancer survivors. Pharmacogenomics J 2009;9:333–40.

24. Smith AK, Conneely KN, Pace TW, et al. Epigenetic changes associated with inflammation in breast cancer patients treated with chemotherapy. Brain Behav Immun 2014;38:227–36.

25. Kim S, Miller BJ, Stefanek ME, Miller AH. Inflammation-induced activation of the indoleamine 2,3-dioxygenase pathway: Relevance to cancer-related fatigue. Cancer 2015;121:2129–36.

26. Mills PJ, Parker B, Dimsdale JE, et al. The relationship between fatigue and quality of life and inflammation during anthracycline-based chemotherapy in breast cancer. Biol Psychol 2005;69:85–96.

27. Jean-Pierre P, Figueroa-Moseley CD, Kohli S, et al. Assessment of cancer-related fatigue: implications for clinical diagnosis and treatment. Oncologist 2007;12 Suppl 1:11–21.

28. Wang XS, Zhao F, Fisch MJ, et al. Prevalence and characteristics of moderate to severe fatigue: a multicenter study in cancer patients and survivors. Cancer 2014;120:425–32.

29. Mendoza TR, Wang XS, Cleeland CS, et al. The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory. Cancer 1999;85:1186–96.

30. Mendoza ME, Capafons A, Gralow JR, et al. Randomized controlled trial of the Valencia model of waking hypnosis plus CBT for pain, fatigue, and sleep management in patients with cancer and cancer survivors. Psychooncology 2016 Jul 28.

31. Glaus A. Assessment of fatigue in cancer and non-cancer patients and in healthy individuals. Support Care Cancer 1993;1:305–15.

32. Seyidova-Khoshknabi D, Davis MP, Walsh D. A systematic review of cancer-related fatigue measurement questionnaires. Am J Hosp Palliat Care 2011;28:119–29.

33. Holzner B, Kemmler G, Greil R, et al. The impact of hemoglobin levels on fatigue and quality of life in cancer patients. Ann Oncol 2002;13:965–73.

34. Stein KD, Jacobsen PB, Blanchard CM, Thors C. Further validation of the multidimensional fatigue symptom inventory-short form. J Pain Symptom Manage 2004;27:14–23.

35. Hwang SS, Chang VT, Rue M, Kasimis B. Multidimensional independent predictors of cancer-related fatigue. J Pain Symptom Manage 2003;26:604–14.

36. Peterspm DR. Scope and generality of verbally defined personality factors. Psychol Rev 1965;72:48–59.

37. Berger AM, Abernethy AP, Atkinson A, et al. NCCN Clinical Practice Guidelines Cancer-related fatigue. J Natl Compr Canc Netw 2010;8:904–31.

38. Crawford J, Cella D, Cleeland CS, et al. Relationship between changes in hemoglobin level and quality of life during chemotherapy in anemic cancer patients receiving epoetin alfa therapy. Cancer 2002;95:888–95.

39. Tonia T, Mettler A, Robert N, et al. Erythropoietin or darbepoetin for patients with cancer. Cochrane Database Syst Rev 2012;12:CD003407.

40. Rizzo JD, Brouwers M, Hurley P, et al. American Society of Hematology/American Society of Clinical Oncology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. Blood 2010;116:4045–59.

41. Preston NJ, Hurlow A, Brine J, Bennett MI. Blood transfusions for anaemia in patients with advanced cancer. Cochrane Database Syst Rev 2012(2):CD009007.

42. Minton O, Stone PC. A comparison of cognitive function, sleep and activity levels in disease-free breast cancer patients with or without cancer-related fatigue syndrome. BMJ Support Palliat Care 2012;2:231–8.

43. Heckler CE, Garland SN, Peoples AR, et al. Cognitive behavioral therapy for insomnia, but not armodafinil, improves fatigue in cancer survivors with insomnia: a randomized placebo-controlled trial. Support Care Cancer 2016;24:2059–66.

44. Howell D, Oliver TK, Keller-Olaman S, et al. Sleep disturbance in adults with cancer: a systematic review of evidence for best practices in assessment and management for clinical practice. Ann Oncol 2014;25:791–800.

45. Wilt TJ, MacDonald R, Brasure M, et al. Pharmacologic treatment of insomnia disorder: an evidence report for a clinical practice guideline by the American College of Physicians. Ann Intern Med 2016;165:103–12.

46. Kuriyama A, Honda M, Hayashino Y. Ramelteon for the treatment of insomnia in adults: a systematic review and meta-analysis. Sleep Med 2014;15:385–92.

47. Strasser F, Palmer JL, Schover LR, et al. The impact of hypogonadism and autonomic dysfunction on fatigue, emotional function, and sexual desire in male patients with advanced cancer: a pilot study. Cancer 2006;107:2949–57.

48. Agasi-Idenburg SC, Thong MS, Punt CJ, et al. Comparison of symptom clusters associated with fatigue in older and younger survivors of colorectal cancer. Support Care Cancer 2017;25:625–32.

49. Miaskowski C, Aouizerat BE. Is there a biological basis for the clustering of symptoms? Semin Oncol Nurs 2007;23:99–105.

50. de Raaf PJ, de Klerk C, Timman R, et al. Systematic monitoring and treatment of physical symptoms to alleviate fatigue in patients with advanced cancer: a randomized controlled trial. J Clin Oncol 2013;31:716–23.

51. Barsevick AM, Whitmer K, Sweeney C, Nail LM. A pilot study examining energy conservation for cancer treatment-related fatigue. Cancer Nurs 2002;25:333–41.

52. Barsevick AM, Dudley W, Beck S, et a;. A randomized clinical trial of energy conservation for patients with cancer-related fatigue. Cancer 2004;100:1302–10.

53. Luciani A, Jacobsen PB, Extermann M, et al. Fatigue and functional dependence in older cancer patients. Am J Clin Oncol 2008;31:424–30.

54. Abrahams HJ, Gielissen MF, Goedendorp MM, et al. A randomized controlled trial of web-based cognitive behavioral therapy for severely fatigued breast cancer survivors (CHANGE-study): study protocol. BMC Cancer 2015;15:765.

55. Quesnel C, Savard J, Simard S, et al. Efficacy of cognitive-behavioral therapy for insomnia in women treated for nonmetastatic breast cancer. J Consult Clin Psychol 2003;71:189–200.

56. Goedendorp MM, Gielissen MF, Verhagen CA, Bleijenberg G. Psychosocial interventions for reducing fatigue during cancer treatment in adults. Cochrane Database Syst Rev 2009(1):CD006953.

57. Greiwe JS, Cheng B, Rubin DC, et al. Resistance exercise decreases skeletal muscle tumor necrosis factor alpha in frail elderly humans. FASEB J 2001;15:475–82.

58. Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev 2016;(9):CD005001.

59. Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database Syst Rev 2012;(11):CD006145.

60. Brown JC, Huedo-Medina TB, Pescatello LS, et al. Efficacy of exercise interventions in modulating cancer-related fatigue among adult cancer survivors: a meta-analysis. Cancer Epidemiol Biomarkers Prev 2011;20:123–33.

61. Steindorf K, Schmidt ME, Klassen O, et al. Randomized, controlled trial of resistance training in breast cancer patients receiving adjuvant radiotherapy: results on cancer-related fatigue and quality of life. Ann Oncol 2014;25:2237–43.

62. Bower JE, Bak K, Berger A, et al. Screening, assessment, and management of fatigue in adult survivors of cancer: an American Society of Clinical oncology clinical practice guideline adaptation. J Clin Oncol 2014;32:1840–50.

63. Kenjale AA, Hornsby WE, Crowgey T, et al. Pre-exercise participation cardiovascular screening in a heterogeneous cohort of adult cancer patients. Oncologist 2014;19:999–1005.

64. Oldervoll LM, Loge JH, Paltiel H, et al. The effect of a physical exercise program in palliative care: A phase II study. J Pain Symptom Manage 2006;31:421–30.

65. Porock D, Kristjanson LJ, Tinnelly K, et al. An exercise intervention for advanced cancer patients experiencing fatigue: a pilot study. J Palliat Care 2000;16:30–6.

66. Lengacher CA, Kip KE, Reich RR, et al. A cost-effective mindfulness stress reduction program: a randomized control trial for breast cancer survivors. Nursing Econ 2015;33:210–8, 32.

67. Lengacher CA, Reich RR, Post-White J, et al. Mindfulness based stress reduction in post-treatment breast cancer patients: an examination of symptoms and symptom clusters. J Behav Med 2012;35:86–94.

68. Sprod LK, Fernandez ID, Janelsins MC, et al. Effects of yoga on cancer-related fatigue and global side-effect burden in older cancer survivors. J Geriatr Oncol 2015;6:8–14.

69. Wang G, Wang S, Jiang P, Zeng C. [Effect of Yoga on cancer related fatigue in breast cancer patients with chemotherapy]. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2014;39:1077–82.

70. Stan DL, Croghan KA, Croghan IT, et al. Randomized pilot trial of yoga versus strengthening exercises in breast cancer survivors with cancer-related fatigue. Support Care Cancer 2016;24:4005–15.

71. Larkey LK, Roe DJ, Weihs KL, et al. Randomized controlled trial of Qigong/Tai Chi Easy on cancer-related fatigue in breast cancer survivors. Ann Behav Med 2015;49:165–76.

72. Molassiotis A, Bardy J, Finnegan-John J, et al. Acupuncture for cancer-related fatigue in patients with breast cancer: a pragmatic randomized controlled trial. J Clin Oncol 2012;30:4470–6.

73. Deng G, Chan Y, Sjoberg D, et al. Acupuncture for the treatment of post-chemotherapy chronic fatigue: a randomized, blinded, sham-controlled trial. Support Care Cancer 2013;21:1735–41.

74. Finnegan-John J, Molassiotis A, Richardson A, Ream E. A systematic review of complementary and alternative medicine interventions for the management of cancer-related fatigue. Integr Cancer Ther 2013;12:276–90.

75. Schwartz AL, Thompson JA, Masood N. Interferon-induced fatigue in patients with melanoma: a pilot study of exercise and methylphenidate. Oncol Nurs Forum 2002;29:E85–90.

76. Spathis A, Dhillan R, Booden D, et al. Modafinil for the treatment of fatigue in lung cancer: a pilot study. Palliat Med 2009;23:325–31.

77. Blackhall L, Petroni G, Shu J, et al. A pilot study evaluating the safety and efficacy of modafinal for cancer-related fatigue. J Palliat Med 2009;12:433–9.

78. Qu D, Zhang Z, Yu X, et al. Psychotropic drugs for the management of cancer-related fatigue: a systematic review and meta-analysis. Eur J Cancer Care (Engl) 2015;25:970–9.

79. Minton O, Richardson A, Sharpe M, et al. Drug therapy for the management of cancer-related fatigue. Cochrane Database Syst Rev 2010(7):CD006704.

80. Moraska AR, Sood A, Dakhil SR, et al. Phase III, randomized, double-blind, placebo-controlled study of long-acting methylphenidate for cancer-related fatigue: North Central Cancer Treatment Group NCCTG-N05C7 trial. J Clin Oncol 2010;28:3673–9.

81. Bruera E, Driver L, Barnes EA, et al. Patient-controlled methylphenidate for the management of fatigue in patients with advanced cancer: a preliminary report. J Clin Oncol 2003;21:4439–43.

82. Kerr CW, Drake J, Milch RA, et al. Effects of methylphenidate on fatigue and depression: a randomized, double-blind, placebo-controlled trial. J Pain Symptom Manage 2012;43:68–77.

83. Escalante CP, Meyers C, Reuben JM, et al. A randomized, double-blind, 2-period, placebo-controlled crossover trial of a sustained-release methylphenidate in the treatment of fatigue in cancer patients. Cancer J 2014;20:8–14.

84. Mar Fan HG, Clemons M, Xu W, et al. A randomised, placebo-controlled, double-blind trial of the effects of d-methylphenidate on fatigue and cognitive dysfunction in women undergoing adjuvant chemotherapy for breast cancer. Support Care Cancer 2008;16:577–83.

85. Butler JM Jr, Case LD, Atkins J, et al. A phase III, double-blind, placebo-controlled prospective randomized clinical trial of d-threo-methylphenidate HCl in brain tumor patients receiving radiation therapy. Int J Radiat Oncol Biol Phys 2007;69:1496–501.

86. Hovey E, de Souza P, Marx G, et al. Phase III, randomized, double-blind, placebo-controlled study of modafinil for fatigue in patients treated with docetaxel-based chemotherapy. Support Care Cancer 2014;22:1233–42.

87. Spathis A, Fife K, Blackhall F, et al. Modafinil for the treatment of fatigue in lung cancer: results of a placebo-controlled, double-blind, randomized trial. J Clin Oncol 2014;32:1882–8.

88. Berenson JR, Yellin O, Shamasunder HK, et al. A phase 3 trial of armodafinil for the treatment of cancer-related fatigue for patients with multiple myeloma. Support Care Cancer 2015;23:1503–12.

89. Boele FW, Douw L, de Groot M, et al. The effect of modafinil on fatigue, cognitive functioning, and mood in primary brain tumor patients: a multicenter randomized controlled trial. Neuro Oncol 2013;15:1420–8.

90. Jean-Pierre P, Morrow GR, Roscoe JA, et al. A phase 3 randomized, placebo-controlled, double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631 patients receiving chemotherapy: a University of Rochester Cancer Center Community Clinical Oncology Program Research base study. Cancer 2010;116:3513–20.

91. Conley CC, Kamen CS, Heckler CE, et al. Modafinil moderates the relationship between cancer-related fatigue and depression in 541 patients receiving chemotherapy. J Clin Psychopharmacol 2016;36:82–5.

92. Brattsand R, Linden M. Cytokine modulation by glucocorticoids: mechanisms and actions in cellular studies. Aliment Pharmacol Ther 1996;10 Suppl 2:81–90.

93. Yennurajalingam S, Frisbee-Hume S, Palmer JL, et al. Reduction of cancer-related fatigue with dexamethasone: a double-blind, randomized, placebo-controlled trial in patients with advanced cancer. J Clin Oncol 2013;31:3076–82.

94. Bruera E, Roca E, Cedaro L, et al. Action of oral methylprednisolone in terminal cancer patients: a prospective randomized double-blind study. Cancer Treat Rep 1985;69:751–4.

95. Pulivarthi K, Dev R, Garcia J, et al. Testosterone replacement for fatigue in male hypogonadic patients with advanced cancer: A preliminary double-blind placebo-controlled trial. J Clin Oncol 2012;30 (suppl). Abstract e19643.

96. Palesh OG, Mustian KM, Peppone LJ, et al. Impact of paroxetine on sleep problems in 426 cancer patients receiving chemotherapy: a trial from the University of Rochester Cancer Center Community Clinical Oncology Program. Sleep Med 2012;13:1184–90.

97. Thekdi SM, Trinidad A, Roth A. Psychopharmacology in Cancer. Curr Psychiatry Rep 2014;17:529.

98. Lesser GJ. Case D, Stark N, et al. A randomized, double-blind, placebo-controlled study of oral coenzyme Q10 to relieve self-reported treatment-related fatigue in newly diagnosed patients with breast cancer. J Support Oncol 2013;11:31–42.

99. Barton DL, Liu H, Dakhil SR, et al. Wisconsin Ginseng (Panax quinquefolius) to improve cancer-related fatigue: a randomized, double-blind trial, N07C2. J Natl Cancer Inst 2013;105:1230–8.

100. Yennurajalingam S, Reddy A, Tannir NM, et al. High-dose Asian ginseng (panax ginseng) for cancer-related fatigue: a preliminary report. Integr Cancer Ther 2015;14:419–27.

101. Howell D, Keller-Olaman S, Oliver TK, et al. A pan-Canadian practice guideline and algorithm: screening, assessment, and supportive care of adults with cancer-related fatigue. Curr Oncol 2013;20:e233–46.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Publications
Article Type
Display Headline
Clinical Assessment and Management of Cancer-Related Fatigue
Display Headline
Clinical Assessment and Management of Cancer-Related Fatigue
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Making Quality Real for Physicians

Article Type
Changed
Thu, 03/28/2019 - 14:52
Display Headline
Making Quality Real for Physicians

From the Department of Medicine Quality Program, Brigham and Women’s Hospital (Drs. Pennant, McElrath, Coblyn, and Desai) and the Institute for Relevant Clinical Data Analytics, Boston Children’s Hospital, (Drs. Szent-Gyorgyi and Greenberg), Boston, MA.

 

Abstract

  • Objectives: To describe the department of medicine quality program (DOMQP) at Brigham and Women’s Hospital (BWH).
  • Methods: The program began in 2007 to engage physicians in local, specialty-specific quality efforts. The program has broadened its scope to include government mandates and hospital-wide priorities, such as maintenance of certification (MOC), “meaningful use (MU),” and medication reconciliation. More recently, we have evolved into a project-based program focusing on both chronic disease management and optimizing care pathways for high-risk inpatient conditions. Our key strategies are developing metrics, raising awareness, distilling information to front-line staff, highlighting relevant action items, and bringing feedback from front-line staff to hospital leadership.
  • Results: We have developed 21 metrics across 13 clinical divisions, with performance improvement seen in > 50% of metrics. In 2014, we leveraged our quality metrics to earn MOC credit, with 100 physicians across 10 divisions earning MOC points through the Practice Assessment option. Additionally, department physicians achieved 90% compliance with our institutional medication reconciliation policy. The percentage of physicians achieving stage 1 MU was 98% in 2013, 99% in 2014, and 100% achieved stage 2 MU in 2015.
  • Conclusion: Over the past 10 years, the DOMQP has played a unique role in promoting quality and serves as a model for QI within the hospital. We are well positioned to provide support to physicians and their practices as the health care environment continues to evolve.

Key words: quality improvement; quality measurement.

 

Within the past several years, the health care landscape in the United States has shifted considerably. New financial risk and quality-related incentive structures have been put in place, such as financial incentives to adopt electronic health records (EHRs) and to demonstrate “meaningful use (MU)” of these EHRs [1]. There is greater focus on value based payments, and accountable care organizations (ACOs) are proliferating [2–4]. Certification and training requirements have changed and require completion of performance improvement projects [5,6]. Upcoming changes to quality measurement and improvement through the Quality Payment Program will bring further changes to how clinicians are both monitored and incentivized or penalized [7]. The confluence of these efforts provides an impetus to incorporate quality measurement and improvement into the day to day practice of medicine.

In 2007, the department of medicine (DOM) at Brigham and Women’s Hospital (BWH), a teaching affiliate of Harvard Medical School and a founding member of Partners Healthcare, began a quality program (DOMQP) to engage physicians in local, specialty-specific quality efforts [8]. The program began by focusing on internally developed performance metrics to drive physician engagement. Later, the program expanded its focus to include more externally focused mandates from federal government and other accreditation bodies [9,10]. In this article, we discuss our efforts, including our early stage work and more recent focus in the areas of meaningful use, medication reconciliation, and maintenance of certification as well as our ongoing projects focused on chronic disease management and high-risk inpatient conditions.

Setting

The DOM has approximately 1400 physician faculty members and is the largest clinical department at BWH. The DOMQP is comprised of a medical director (0.35 FTE) and 2 project coordinators (2 FTE) who operate in a consultative capacity to liaison between the various levels of clinical leadership and frontline staff. The DOMQP serves 13 divisions: 11 medical specialties, primary care, and the hospitalist service.

Internally Driven, Specialty-Specific Quality Metrics

The early stages of our efforts focused on engaging clinical leadership and physicians, navigating the hospital’s information systems (IS) to develop quality reports within the EHR and implementing strategies for improvement. Until 30 May 2015, BWH utilized an internally developed EHR comprising a myriad of individual systems (eg, billing, electronic medication administration, clinical repository) that did not interface easily with one another. To resolve the IS challenges, we engaged many levels of the organization’s IS structure and requested updates or developed workaround solutions leading to the integration of the various datasets for comprehensive reporting. This resulted in a new agreement between the hospital and Partners that allowed for billing data to be sent to the same repository that housed the clinical data.

We developed at least one data report for each specialty division. The divisions selected a quality measure, and we worked with clinical leadership to define the numerator and denominator and identify any additional information they wanted in the report, eg, demographics, visit dates, labs. To produce the report, an IS consultant compiles the data elements on an excel spreadsheet. This spreadsheet is then manually chart reviewed by the DOMQP for accuracy before it is converted into a report. The report is then reviewed by the DOMQP to ensure the information is presented correctly and is easily interpreted, and then shared with the division champion(s), who determine how to message and introduce a proposed improvement effort before it is shared with the entire division.

Reports display a graph showing overall performance of the division and individual provider rates (an example is shown in Figure 1). Currently, most reports are sent unblinded; previously it was left to the discretion of the division chief. Patients are attributed to the provider they have seen most often during the reporting period. If there is a tie between 2 providers the patient is attributed to the most recent provider. Provider-level reports are also developed to show the physician’s performance over time as compared to the division’s overall performance. The reports list the provider’s patients and their performance status on the metric. We found that identifying patients not meeting the metric are essential to gaining clinician buy-in [11]. Physicians can be skeptical of baseline data if the rationale and details around how measurement was performed are not clearly communicated. Concrete examples with their own patient panel provide the proof they need and the relevant caveats and limitations of the data, in order to work toward improvement. We have seen clinic improvement on many metrics, with some clinics improving from 40% to over 88% in 2 to 3 years [12,13] (Table 1).

With accurate reports in place, we leverage all staff levels, from practice assistants to clinicians, whenever possible to improve by evaluating the performance data and setting goals. We use plan-do-study-act (PDSA) cycles to iterate performance improvement plans and tailor our efforts to the unique staffing levels and patient populations of each division [14,15]. For example, we have worked for years on improving blood pressure control among chronic kidney disease patients in our renal division. Initially, our first PDSA cycle involved mostly education and data sharing. When improvements were not seen, we did another PDSA cycle adding a brief form for physicians to fill out in the clinic for patients who had at least 3 blood pressure measurements that were not at goal. The sheet clearly displayed the 3 elevated blood pressures and asked the provider to note that day’s blood pressure as well as if the medication was adjusted or if the patient is not taking the medication(s) prescribed. Data from these worksheets informed efforts to focus on non–office-based blood pressure measurements [16]. Through a grant the renal division was able to give free blood pressure cuffs to patients for at-home monitoring.

In an effort to improve influenza and pneumococcal vaccination rates, we worked closely with front-line clinicians and staff to create improvement strategies tailored to the patient population and clinic staffing structure in 4 divisions: allergy (order and document flu vaccinations for asthma patients), rheumatology (point-of-care standing pneumococcal paper-orders for immunosuppressed patients), infectious diseases (a nurse-driven influenza protocol was implemented for HIV patients), and pulmonary (letters sent to chronic lung disease patients asking them to bring documentation of prior pneumococcal vaccination to their next visit). We saw increases in vaccination rates across all 4 divisions using varied approaches to reach our goals [17].

 

 

Meaningful Use

Our team was responsible for ensuring that the DOM succeeded in the federal government’s MU program. Millions of dollars were at risk to the hospital and to physicians based on performance on various MU metrics. The hospital made this a priority and set a goal for 90% of eligible providers to successfully certify in Stage 1 as meaningful users in the first year of MU.

To meet MU requirements, we categorized metrics by those requiring physician effort and those relying on clinic staff effort. We worked with division and departmental leadership to develop and implement an approach to improve performance on each metric. Monthly emails were sent with the current performance data, explanations of low-performing metrics, and actions to take for improvement (Figure 2). This information was reinforced at faculty presentations.

While working with the clinics to achieve meaningful use, we recognized that a range of staff members played a critical role in helping physicians meet MU metric targets. We worked with the clinic and departmental administrative leadership to set up a one-time bonus payment in 2013 to all DOM clinic staff, including medical assistants, licensed practical nurses, practice assistants, for appreciation of their significant efforts to help the physicians and the hospital achieve their MU goals. As health care delivery continues to rely more heavily on highly functional teams, acknowledgment of the efforts of non-clinical staff in helping front-line clinicians in achieving MU can help promote teamwork around a common goal.

Our partnership with the hospital MU team lead to a final tally of 98% of eligible providers meeting stage 1 in 2013, 99% in 2014 and 100% for the modified stage 2 in 2015 and 99% in 2016 for DOM specialty divisions, representing over 250 physicians.

Medication Reconciliation

The impetus for the hospital’s focus on medication reconciliation was the Joint Commission requirement that medications listed as prescribed in the EHR be reconciled with the patient at each visit [10]. The hospital’s quality and safety team created a hospital-wide metric aligned with hospital policy to measure how providers were reconciling medication lists during office visits. The medication reconciliation metric tallied visits when a medication change occurred (denominator) in which all of the medications originally prescribed by a physician in that specialty clinic were reconciled (numerator). This made it easier for specialists to buy-in to the metric because they were only responsible for the medications they prescribed and not the entire list. To meet the metric, physicians are required to take an action on each of the medications they prescribed by clicking taking, not taking, taking differently, or discontinuing. If the clinic has additional staff within the clinic to assist in the medication review process with the patient, physicians would receive credit for their actions once they had reviewed the medication list.

The metric BWH established to measure medication reconciliation was distinct and more rigorous than the medication reconciliation metric used to meet MU requirements. This presented a challenge in both physician communication regarding requirements and data sharing to drive performance improvement. Since the DOM is the largest and most prescribing department, we had to work with clinical and administrative leaders at the divisional level so that all staff understood the exact requirements and how to achieve them. When presenting to faculty we encountered many questions, and general resistance to more clicks in the EHR, as there was no universal “reconcile” button in the extant EHR. After breaking down the process into discrete components with staff and analyzing the data, we targeted outreach to the specific divisions and clinics that needed additional education and support. In our monthly email communication to division leadership, we displayed comparative data on all divisions within the DOM.

In late 2013, medication reconciliation was named as one of the main metrics hospital-wide that would be tied to a financial incentive for departments. The incentive was for money to be returned to the departments (not physicians) and the goal was common across the hospital. The hospital set a goal that 90% of visits with a medication change have all medications originally prescribed by that clinic reconciled. This led to a larger hospital-wide push to achieve success in medication reconciliation, including presentation of department-level data to senior leadership, such as the hospital president, chief medical officer, chief nursing officer, chief operating officer, and the various department chairs. Though the initial progress for this metric was slow, progress became evident across all divisions by the spring of 2014 (Figure 3) [18].

After we transitioned to a new vendor-based EHR in 2015, we developed a new medication reconciliation metric, aimed to align with Stage 2 MU requirements, and ensured all divisions had processes in place to review and reconcile medications. In our new EHR, medications lists are shared and viewable between ambulatory and inpatient environments and discharge summaries contain changes made to medications while inpatient. We have found that our performance on our new metric is 70.5% for February 2017 and we are continuing to educate physicians and staff on our new EHR functionality, our new electronic measurement and on workflows to help assist with improving performance with a short-term goal of 80%. We have found that our new EHR functionality does not match the ease of our old EHR functionality, which has made improvement on this metric a significant challenge for our clinicians and staff.

 

 

Maintenance of Certification

As of 2013, physicians who were board certified by the ABIM or ABMS were required to attain 20 practice assessment points showing participation in quality improvement activities for maintenance of certification (MOC). Although ABIM suspended the practice assessment requirement in 2015, it is likely that some component of quality improvement will be assessed in the future [5,19]. Prior to the changes in 2015, we were approached by the hospital leadership to roll out a new, streamlined process to leverage our existing quality metrics and established performance improvement plans in order to allow physicians to gain MOC credit. Each metric/project was vetted with 3 separate groups: the DOMQP, hospital leadership leading the MOC efforts, and Partners Healthcare, our overarching local health care system. We worked with our division physician champions and the physicians preparing for board re-certification to complete the one-time documentation required by the hospital for MOC. This process took approximately 6 months per project. Once the project was approved by all 3 channels, all physicians participating in the quality effort could get the practice assessment points by completing a simple attestation form.

For example, the DOMQP had already developed a metric with our allergy division measuring use of the Asthma Control Test during office-based visits. During one of our faculty meeting presentations, we shared that Partners now had a pilot program to count current QI projects at the hospital for MOC points. We asked for interested volunteers and worked with 1 or 2 physicians who most often were up for recertification that year. DOMQP provided the data that we already managed and prepopulated the required application and PowerPoint. The physician adds text regarding the clinical relevance of the project, how the improvement strategies in clinic evolved over time, and barriers faced in achieving their goal. The DOMQP sends the documents to Partners for review, and once the project is accepted, the division chief forwards attestation forms to participating physicians. To be eligible, a physician had to have data present in our monthly reports with at least 10 patients in their denominator. Some division chiefs included the additional criterion that a provider also had to be at goal performance set by the division in order to attest and receive MOC points. To date we have 15 projects approved with 206 physician attestations submitted for MOC points (Table 2). Through the aid of the Partners Healthcare Office of Continuing Professional Development we are now able to offer physicians assistants AMA PRA Category 1 credit for participation in division projects. Having an easy process that builds on existing DOMQP efforts has been a highly valued resource to our faculty. As the MOC process was de-emphasized by ABIM, we have shifted our focus to the changes surrounding the Quality Payment Program and are aligning our project-based work to support quality measurement and improvement.

Current Focus

After the stabilization of our new EHR and the re-creation of former reports, the program began to engage divisions in new measures. We still focus on chronic disease management and vaccinations but instead try to create a unified approach across multiple divisions within the DOM. Building upon our previous work in the renal division, over the past year we convened a hyper-tension work group comprising physician leads from endocrine, cardiology, renal, primary care, gerontology, neurology, pharmacy, and obstetrics. The goal of these meetings is to optimize blood pressure management across different patient populations by creating a centralized hospital approach with an algorithm agreed upon by the physician workgroup. We were able to secure additional internal grant funding to develop a pilot project where bluetooth blood pressure cuffs are given to eligible hypertensive patients in our pilot ambulatory practices. The daily blood pressure readings are transmitted into the EHR and a nurse practitioner or pharmacist contacts the patient at defined intervals to address any barriers and titrate medications as necessary. Analysis of the outcomes will be presented this fall. Similarly with vaccinations we are creating an automated order form within the EHR that will appear whenever a specialist places an order to start immunosuppressive medications. This will prompt the provider to order appropriate labs and vaccinations recommended for the course of treatment.

In addition to expanding upon previous metrics, we have expanded our scope to focus on patient safety measures, specifically, missed and delayed cancer diagnoses of the lung and colon. We are working on processes to track every patient with an abnormal finding from point of notification to completion of recommended follow-up at the appropriate intervals. Also we have now have 3 projects in the inpatient setting: chronic obstructive pulmonary disease (COPD) readmissions, and 2 standardized clinical assessment and management pathways (SCAMPs), one on acute kidney injury and the other on congestive heart failure [20]. The COPD project aims to have every patient admitted with COPD receive a pulmonary and respiratory consult during their stay and a follow-up visit with a pulmonologist. The goal of any SCAMP is to standardize care in an area where there is a lot of variability through the use of clinical pathways.

Communication Strategy

In order for the DOMQP to ensure that multiple quality requirements are met by all divisions, we have established a robust communication strategy with the goal of clear, concise, and relevant information-sharing with physicians and staff. We engage physicians through direct meetings, regular emails, and data reporting. The purpose of our outreach to the division faculty is threefold: (1) to educate physicians about hospital-wide programs, (2) to orient them to specific action items required for success, and (3) to funnel questions back to project leaders to ensure that the feedback of clinicians was incorporated into hospital wide quality initiatives. Our first challenge is to provide context for physicians about the project, be it based on accreditation, credentialing or a federal mandate. We work with the hospital project leaders to learn as much as possible about the efforts they are promoting so we can work in concert with them to highlight key messages to physicians.

Next, we establish a schedule to present at each clinical specialty faculty meeting on a regular basis (semi-annually to quarterly). At these meetings we present an overall picture of the key initiatives relevant to the division, identify milestones, offer clear timelines and prioritization of these projects, and narrow the focus of work to a few bulleted action items for all levels of the clinic staff to incorporate into its workflow. We then listen to questions and concerns and bring these back to the initiative leaders so that systematic changes can be made. Answers and updates are communicated back to the divisions, thus closing the communication loop. We interface with practice managers and clinical support staff to identify opportunities for them to support physicians in meeting initiative requirements [21,22].

In addition to presenting at faculty meetings, we present updates to departmental and hospital leadership, including vice chairs and division chiefs. These meetings include high-level data on performance as well as an opportunity to discuss the challenges we identified through our discussions with individual specialties. These forums are a good place to discuss overarching process issues or to disseminate answers to previous questions.

An important part of our communication plan involves our comprehensive monthly emails. For each initiative, we receive department level data on a monthly basis from the project leadership. We deconstruct these reports to enable us to evaluate our 13 divisions individually. We show performance at the physician level and highlight general areas where improvement is needed. We send monthly e-mails to division clinical and administrative leadership to apprise them of their division’s performance and inform them of areas that require concentrated effort. Depending on the initiative, we present data as a snapshot in time or trend over time. From 2012–2017 the DOMQP has helped to bridge the gap between large-scale rollouts of the new initiatives and the vast number of DOM physicians who required more specific education and tools to meet these new requirements.

 

 

Conclusion

The DOMQP has been working on quality within the department of medicine for the past 10 years. We have moved from initiating an internal quality program among the specialty providers, which required education among faculty and resolve to overcome many IS challenges, to serving as a resource for hospital-wide quality-related initiatives. We have developed a successful architecture for disseminating information and guiding faculty and administrative support toward success on a multitude of metrics that have implications on both finances and sound patient care. We have navigated the significant challenges associated with the large-scale change due to the EHR transition we underwent in 2015, including clinician burnout and fatigue, new EHR functionality, and the development of a new data reporting infrastructure and governance. The DOMQP continues to demonstrate that quality has a multifaceted role to play within a hospital.

As the health care environment continues to evolve, the need for this level of support for clinics will increase. The DOMQP is well positioned to provide continuing support to physicians and their practices in measuring and improving quality, with attention paid to such areas as coordination and efficacy of care, patient-reported outcomes, patient safety, and population health management. We believe that the DOMQP can serve as a model of a departmental quality program.

 

Acknowledgements: We would like to thank the department of medicine administration, faculty and support staff for their continuous effort and support in the work of clinical quality improvement.

Corresponding author: Sonali Desai, MD, MPH, Brigham & Women’s Hospital, 45 Francis St., PBB-B3, Boston, MA 02115, [email protected].

Financial disclosures: None.

References

1. Electronic health records (EHR) incentive programs. Accessed at www.cms.gov/Regulations-and Guidance/Legislation/EHRIncentivePrograms/index.html?redirect=/EHRIncentiveprograms/.

2. Accountable care organizations (ACO). Accessed at www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/ACO/index.html?redirect=/aco.

3. Quinn K. The 8 basic payment methods in health care. Ann Intern Med 2015;163:300–6.

4. Muhlestein D, McClellan M. Accountable care organizations in 2016: private and public-sector growth and dispersion. Health Aff blog 21 Apr 2016. Accessed at http://healthaffairs.org/blog/2016/04/21/accountable-care-organizations-in-2016-private-and-public-sector-growth-and-dispersion/.

5. ABIM. Maintenance of certification. Accessed 6 Oct 2015 at www.abim.org/maintenance-of-certification/.

6. Accreditation Council for Graduate Medical Education. CLER pathways to excellence. 2014. Accessed 6 Oct 2015 at www.acgme.org/acgmeweb/Portals/0/PDFs/CLER/CLER_Brochure.pdf.

7. Centers for Medicare and Medicaid Services. Quality payment program. Accessed 31 Mar 2017 at https://qpp.cms.gov/measures/quality.

8. Szent-Gyorgyi LE, Coblyn J, Turchin A, et al. Building a departmental quality program: a patient-based and provider-led approach. Acad Med 2011;86:314–20.

9. Brook RH, McGlynn EA, Shekelle PG. Defining and measuring quality of care: a perspective from US researchers. Int J Qual Health Care 2000;12:281–95.

10. The Joint Commision. National patient safety goals. Accessed at www.jointcommission.org/standards_information/npsgs.aspx.

11. Conway PH, Mostashari F, Clancy C. The future of quality measurement for improvement and accountability. JAMA 2013;309:2215–6.

12. Desai SP, Turchin A, Szent-Gyorgyi LE, et al. Routinely measuring and reporting pneumococcal vaccination among immunosuppressed rheumatology outpatients: the first step in improving quality. Rheumatology (Oxford) 2011;50:366–72.

13. Desai SP, Lu B, Szent-Gyorgyi LE, et al. Increasing pneumococcal vaccination for immunosuppressed patients: a cluster quality improvement trial. Arthritis Rheum 2013;65:39–47.

14. Deming Institute. PDSA cycle. Accessed at www.deming.org/theman/theories/pdsacycle.

15. McGlynn EA, Schneider EC, Kerr EA. Reimagining quality measurement. N Engl J Med 2014;371:2150–3.

16. Greenberg JO, Vakharia N, Szent-Gyorgyi LE, et al. Meaningful measurement: developing a measurement system to improve blood pressure control in patients with chronic kidney disease. J Am Med Inform Assoc 2013;20:e97–e101.

17. Pennant KN, Costa JJ, Fuhlbrigge AL, et al. Improving influenza and pneumococcal vaccination rates in ambulatory specialty practices. Open Forum Infect Dis 2015;2:ofv119.

18. Keogh C, Kachalia A, Fiumara K, et al. Ambulatory medication reconciliation: using a collaborative approach to process improvement at an academic medical center. Jt Comm J Qual Patient Saf 2016;42:186–94.

19. ABIM. Maintenance of certification FAQ. Accessed at www.abim.org/maintenance-of-certification/moc-faq/default.aspx.

20. Mendu ML, Ciociolo GR Jr, McLaughlin SR, et al. A decision-making algorithm for initiation and discontinuation of RRT in severe AKI. Clin J Am Soc Nephrol 2017;12:228–36.

21. Galvin RS, McGlynn EA. Using performance measurement to drive improvement: a road map for change. Med Care 2003;41:I48–60.

22. Glaser J, Hess R. Leveraging healthcare IT to improve operational performance. Healthc Financ Manage 2011;65:82–5.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Topics
Sections

From the Department of Medicine Quality Program, Brigham and Women’s Hospital (Drs. Pennant, McElrath, Coblyn, and Desai) and the Institute for Relevant Clinical Data Analytics, Boston Children’s Hospital, (Drs. Szent-Gyorgyi and Greenberg), Boston, MA.

 

Abstract

  • Objectives: To describe the department of medicine quality program (DOMQP) at Brigham and Women’s Hospital (BWH).
  • Methods: The program began in 2007 to engage physicians in local, specialty-specific quality efforts. The program has broadened its scope to include government mandates and hospital-wide priorities, such as maintenance of certification (MOC), “meaningful use (MU),” and medication reconciliation. More recently, we have evolved into a project-based program focusing on both chronic disease management and optimizing care pathways for high-risk inpatient conditions. Our key strategies are developing metrics, raising awareness, distilling information to front-line staff, highlighting relevant action items, and bringing feedback from front-line staff to hospital leadership.
  • Results: We have developed 21 metrics across 13 clinical divisions, with performance improvement seen in > 50% of metrics. In 2014, we leveraged our quality metrics to earn MOC credit, with 100 physicians across 10 divisions earning MOC points through the Practice Assessment option. Additionally, department physicians achieved 90% compliance with our institutional medication reconciliation policy. The percentage of physicians achieving stage 1 MU was 98% in 2013, 99% in 2014, and 100% achieved stage 2 MU in 2015.
  • Conclusion: Over the past 10 years, the DOMQP has played a unique role in promoting quality and serves as a model for QI within the hospital. We are well positioned to provide support to physicians and their practices as the health care environment continues to evolve.

Key words: quality improvement; quality measurement.

 

Within the past several years, the health care landscape in the United States has shifted considerably. New financial risk and quality-related incentive structures have been put in place, such as financial incentives to adopt electronic health records (EHRs) and to demonstrate “meaningful use (MU)” of these EHRs [1]. There is greater focus on value based payments, and accountable care organizations (ACOs) are proliferating [2–4]. Certification and training requirements have changed and require completion of performance improvement projects [5,6]. Upcoming changes to quality measurement and improvement through the Quality Payment Program will bring further changes to how clinicians are both monitored and incentivized or penalized [7]. The confluence of these efforts provides an impetus to incorporate quality measurement and improvement into the day to day practice of medicine.

In 2007, the department of medicine (DOM) at Brigham and Women’s Hospital (BWH), a teaching affiliate of Harvard Medical School and a founding member of Partners Healthcare, began a quality program (DOMQP) to engage physicians in local, specialty-specific quality efforts [8]. The program began by focusing on internally developed performance metrics to drive physician engagement. Later, the program expanded its focus to include more externally focused mandates from federal government and other accreditation bodies [9,10]. In this article, we discuss our efforts, including our early stage work and more recent focus in the areas of meaningful use, medication reconciliation, and maintenance of certification as well as our ongoing projects focused on chronic disease management and high-risk inpatient conditions.

Setting

The DOM has approximately 1400 physician faculty members and is the largest clinical department at BWH. The DOMQP is comprised of a medical director (0.35 FTE) and 2 project coordinators (2 FTE) who operate in a consultative capacity to liaison between the various levels of clinical leadership and frontline staff. The DOMQP serves 13 divisions: 11 medical specialties, primary care, and the hospitalist service.

Internally Driven, Specialty-Specific Quality Metrics

The early stages of our efforts focused on engaging clinical leadership and physicians, navigating the hospital’s information systems (IS) to develop quality reports within the EHR and implementing strategies for improvement. Until 30 May 2015, BWH utilized an internally developed EHR comprising a myriad of individual systems (eg, billing, electronic medication administration, clinical repository) that did not interface easily with one another. To resolve the IS challenges, we engaged many levels of the organization’s IS structure and requested updates or developed workaround solutions leading to the integration of the various datasets for comprehensive reporting. This resulted in a new agreement between the hospital and Partners that allowed for billing data to be sent to the same repository that housed the clinical data.

We developed at least one data report for each specialty division. The divisions selected a quality measure, and we worked with clinical leadership to define the numerator and denominator and identify any additional information they wanted in the report, eg, demographics, visit dates, labs. To produce the report, an IS consultant compiles the data elements on an excel spreadsheet. This spreadsheet is then manually chart reviewed by the DOMQP for accuracy before it is converted into a report. The report is then reviewed by the DOMQP to ensure the information is presented correctly and is easily interpreted, and then shared with the division champion(s), who determine how to message and introduce a proposed improvement effort before it is shared with the entire division.

Reports display a graph showing overall performance of the division and individual provider rates (an example is shown in Figure 1). Currently, most reports are sent unblinded; previously it was left to the discretion of the division chief. Patients are attributed to the provider they have seen most often during the reporting period. If there is a tie between 2 providers the patient is attributed to the most recent provider. Provider-level reports are also developed to show the physician’s performance over time as compared to the division’s overall performance. The reports list the provider’s patients and their performance status on the metric. We found that identifying patients not meeting the metric are essential to gaining clinician buy-in [11]. Physicians can be skeptical of baseline data if the rationale and details around how measurement was performed are not clearly communicated. Concrete examples with their own patient panel provide the proof they need and the relevant caveats and limitations of the data, in order to work toward improvement. We have seen clinic improvement on many metrics, with some clinics improving from 40% to over 88% in 2 to 3 years [12,13] (Table 1).

With accurate reports in place, we leverage all staff levels, from practice assistants to clinicians, whenever possible to improve by evaluating the performance data and setting goals. We use plan-do-study-act (PDSA) cycles to iterate performance improvement plans and tailor our efforts to the unique staffing levels and patient populations of each division [14,15]. For example, we have worked for years on improving blood pressure control among chronic kidney disease patients in our renal division. Initially, our first PDSA cycle involved mostly education and data sharing. When improvements were not seen, we did another PDSA cycle adding a brief form for physicians to fill out in the clinic for patients who had at least 3 blood pressure measurements that were not at goal. The sheet clearly displayed the 3 elevated blood pressures and asked the provider to note that day’s blood pressure as well as if the medication was adjusted or if the patient is not taking the medication(s) prescribed. Data from these worksheets informed efforts to focus on non–office-based blood pressure measurements [16]. Through a grant the renal division was able to give free blood pressure cuffs to patients for at-home monitoring.

In an effort to improve influenza and pneumococcal vaccination rates, we worked closely with front-line clinicians and staff to create improvement strategies tailored to the patient population and clinic staffing structure in 4 divisions: allergy (order and document flu vaccinations for asthma patients), rheumatology (point-of-care standing pneumococcal paper-orders for immunosuppressed patients), infectious diseases (a nurse-driven influenza protocol was implemented for HIV patients), and pulmonary (letters sent to chronic lung disease patients asking them to bring documentation of prior pneumococcal vaccination to their next visit). We saw increases in vaccination rates across all 4 divisions using varied approaches to reach our goals [17].

 

 

Meaningful Use

Our team was responsible for ensuring that the DOM succeeded in the federal government’s MU program. Millions of dollars were at risk to the hospital and to physicians based on performance on various MU metrics. The hospital made this a priority and set a goal for 90% of eligible providers to successfully certify in Stage 1 as meaningful users in the first year of MU.

To meet MU requirements, we categorized metrics by those requiring physician effort and those relying on clinic staff effort. We worked with division and departmental leadership to develop and implement an approach to improve performance on each metric. Monthly emails were sent with the current performance data, explanations of low-performing metrics, and actions to take for improvement (Figure 2). This information was reinforced at faculty presentations.

While working with the clinics to achieve meaningful use, we recognized that a range of staff members played a critical role in helping physicians meet MU metric targets. We worked with the clinic and departmental administrative leadership to set up a one-time bonus payment in 2013 to all DOM clinic staff, including medical assistants, licensed practical nurses, practice assistants, for appreciation of their significant efforts to help the physicians and the hospital achieve their MU goals. As health care delivery continues to rely more heavily on highly functional teams, acknowledgment of the efforts of non-clinical staff in helping front-line clinicians in achieving MU can help promote teamwork around a common goal.

Our partnership with the hospital MU team lead to a final tally of 98% of eligible providers meeting stage 1 in 2013, 99% in 2014 and 100% for the modified stage 2 in 2015 and 99% in 2016 for DOM specialty divisions, representing over 250 physicians.

Medication Reconciliation

The impetus for the hospital’s focus on medication reconciliation was the Joint Commission requirement that medications listed as prescribed in the EHR be reconciled with the patient at each visit [10]. The hospital’s quality and safety team created a hospital-wide metric aligned with hospital policy to measure how providers were reconciling medication lists during office visits. The medication reconciliation metric tallied visits when a medication change occurred (denominator) in which all of the medications originally prescribed by a physician in that specialty clinic were reconciled (numerator). This made it easier for specialists to buy-in to the metric because they were only responsible for the medications they prescribed and not the entire list. To meet the metric, physicians are required to take an action on each of the medications they prescribed by clicking taking, not taking, taking differently, or discontinuing. If the clinic has additional staff within the clinic to assist in the medication review process with the patient, physicians would receive credit for their actions once they had reviewed the medication list.

The metric BWH established to measure medication reconciliation was distinct and more rigorous than the medication reconciliation metric used to meet MU requirements. This presented a challenge in both physician communication regarding requirements and data sharing to drive performance improvement. Since the DOM is the largest and most prescribing department, we had to work with clinical and administrative leaders at the divisional level so that all staff understood the exact requirements and how to achieve them. When presenting to faculty we encountered many questions, and general resistance to more clicks in the EHR, as there was no universal “reconcile” button in the extant EHR. After breaking down the process into discrete components with staff and analyzing the data, we targeted outreach to the specific divisions and clinics that needed additional education and support. In our monthly email communication to division leadership, we displayed comparative data on all divisions within the DOM.

In late 2013, medication reconciliation was named as one of the main metrics hospital-wide that would be tied to a financial incentive for departments. The incentive was for money to be returned to the departments (not physicians) and the goal was common across the hospital. The hospital set a goal that 90% of visits with a medication change have all medications originally prescribed by that clinic reconciled. This led to a larger hospital-wide push to achieve success in medication reconciliation, including presentation of department-level data to senior leadership, such as the hospital president, chief medical officer, chief nursing officer, chief operating officer, and the various department chairs. Though the initial progress for this metric was slow, progress became evident across all divisions by the spring of 2014 (Figure 3) [18].

After we transitioned to a new vendor-based EHR in 2015, we developed a new medication reconciliation metric, aimed to align with Stage 2 MU requirements, and ensured all divisions had processes in place to review and reconcile medications. In our new EHR, medications lists are shared and viewable between ambulatory and inpatient environments and discharge summaries contain changes made to medications while inpatient. We have found that our performance on our new metric is 70.5% for February 2017 and we are continuing to educate physicians and staff on our new EHR functionality, our new electronic measurement and on workflows to help assist with improving performance with a short-term goal of 80%. We have found that our new EHR functionality does not match the ease of our old EHR functionality, which has made improvement on this metric a significant challenge for our clinicians and staff.

 

 

Maintenance of Certification

As of 2013, physicians who were board certified by the ABIM or ABMS were required to attain 20 practice assessment points showing participation in quality improvement activities for maintenance of certification (MOC). Although ABIM suspended the practice assessment requirement in 2015, it is likely that some component of quality improvement will be assessed in the future [5,19]. Prior to the changes in 2015, we were approached by the hospital leadership to roll out a new, streamlined process to leverage our existing quality metrics and established performance improvement plans in order to allow physicians to gain MOC credit. Each metric/project was vetted with 3 separate groups: the DOMQP, hospital leadership leading the MOC efforts, and Partners Healthcare, our overarching local health care system. We worked with our division physician champions and the physicians preparing for board re-certification to complete the one-time documentation required by the hospital for MOC. This process took approximately 6 months per project. Once the project was approved by all 3 channels, all physicians participating in the quality effort could get the practice assessment points by completing a simple attestation form.

For example, the DOMQP had already developed a metric with our allergy division measuring use of the Asthma Control Test during office-based visits. During one of our faculty meeting presentations, we shared that Partners now had a pilot program to count current QI projects at the hospital for MOC points. We asked for interested volunteers and worked with 1 or 2 physicians who most often were up for recertification that year. DOMQP provided the data that we already managed and prepopulated the required application and PowerPoint. The physician adds text regarding the clinical relevance of the project, how the improvement strategies in clinic evolved over time, and barriers faced in achieving their goal. The DOMQP sends the documents to Partners for review, and once the project is accepted, the division chief forwards attestation forms to participating physicians. To be eligible, a physician had to have data present in our monthly reports with at least 10 patients in their denominator. Some division chiefs included the additional criterion that a provider also had to be at goal performance set by the division in order to attest and receive MOC points. To date we have 15 projects approved with 206 physician attestations submitted for MOC points (Table 2). Through the aid of the Partners Healthcare Office of Continuing Professional Development we are now able to offer physicians assistants AMA PRA Category 1 credit for participation in division projects. Having an easy process that builds on existing DOMQP efforts has been a highly valued resource to our faculty. As the MOC process was de-emphasized by ABIM, we have shifted our focus to the changes surrounding the Quality Payment Program and are aligning our project-based work to support quality measurement and improvement.

Current Focus

After the stabilization of our new EHR and the re-creation of former reports, the program began to engage divisions in new measures. We still focus on chronic disease management and vaccinations but instead try to create a unified approach across multiple divisions within the DOM. Building upon our previous work in the renal division, over the past year we convened a hyper-tension work group comprising physician leads from endocrine, cardiology, renal, primary care, gerontology, neurology, pharmacy, and obstetrics. The goal of these meetings is to optimize blood pressure management across different patient populations by creating a centralized hospital approach with an algorithm agreed upon by the physician workgroup. We were able to secure additional internal grant funding to develop a pilot project where bluetooth blood pressure cuffs are given to eligible hypertensive patients in our pilot ambulatory practices. The daily blood pressure readings are transmitted into the EHR and a nurse practitioner or pharmacist contacts the patient at defined intervals to address any barriers and titrate medications as necessary. Analysis of the outcomes will be presented this fall. Similarly with vaccinations we are creating an automated order form within the EHR that will appear whenever a specialist places an order to start immunosuppressive medications. This will prompt the provider to order appropriate labs and vaccinations recommended for the course of treatment.

In addition to expanding upon previous metrics, we have expanded our scope to focus on patient safety measures, specifically, missed and delayed cancer diagnoses of the lung and colon. We are working on processes to track every patient with an abnormal finding from point of notification to completion of recommended follow-up at the appropriate intervals. Also we have now have 3 projects in the inpatient setting: chronic obstructive pulmonary disease (COPD) readmissions, and 2 standardized clinical assessment and management pathways (SCAMPs), one on acute kidney injury and the other on congestive heart failure [20]. The COPD project aims to have every patient admitted with COPD receive a pulmonary and respiratory consult during their stay and a follow-up visit with a pulmonologist. The goal of any SCAMP is to standardize care in an area where there is a lot of variability through the use of clinical pathways.

Communication Strategy

In order for the DOMQP to ensure that multiple quality requirements are met by all divisions, we have established a robust communication strategy with the goal of clear, concise, and relevant information-sharing with physicians and staff. We engage physicians through direct meetings, regular emails, and data reporting. The purpose of our outreach to the division faculty is threefold: (1) to educate physicians about hospital-wide programs, (2) to orient them to specific action items required for success, and (3) to funnel questions back to project leaders to ensure that the feedback of clinicians was incorporated into hospital wide quality initiatives. Our first challenge is to provide context for physicians about the project, be it based on accreditation, credentialing or a federal mandate. We work with the hospital project leaders to learn as much as possible about the efforts they are promoting so we can work in concert with them to highlight key messages to physicians.

Next, we establish a schedule to present at each clinical specialty faculty meeting on a regular basis (semi-annually to quarterly). At these meetings we present an overall picture of the key initiatives relevant to the division, identify milestones, offer clear timelines and prioritization of these projects, and narrow the focus of work to a few bulleted action items for all levels of the clinic staff to incorporate into its workflow. We then listen to questions and concerns and bring these back to the initiative leaders so that systematic changes can be made. Answers and updates are communicated back to the divisions, thus closing the communication loop. We interface with practice managers and clinical support staff to identify opportunities for them to support physicians in meeting initiative requirements [21,22].

In addition to presenting at faculty meetings, we present updates to departmental and hospital leadership, including vice chairs and division chiefs. These meetings include high-level data on performance as well as an opportunity to discuss the challenges we identified through our discussions with individual specialties. These forums are a good place to discuss overarching process issues or to disseminate answers to previous questions.

An important part of our communication plan involves our comprehensive monthly emails. For each initiative, we receive department level data on a monthly basis from the project leadership. We deconstruct these reports to enable us to evaluate our 13 divisions individually. We show performance at the physician level and highlight general areas where improvement is needed. We send monthly e-mails to division clinical and administrative leadership to apprise them of their division’s performance and inform them of areas that require concentrated effort. Depending on the initiative, we present data as a snapshot in time or trend over time. From 2012–2017 the DOMQP has helped to bridge the gap between large-scale rollouts of the new initiatives and the vast number of DOM physicians who required more specific education and tools to meet these new requirements.

 

 

Conclusion

The DOMQP has been working on quality within the department of medicine for the past 10 years. We have moved from initiating an internal quality program among the specialty providers, which required education among faculty and resolve to overcome many IS challenges, to serving as a resource for hospital-wide quality-related initiatives. We have developed a successful architecture for disseminating information and guiding faculty and administrative support toward success on a multitude of metrics that have implications on both finances and sound patient care. We have navigated the significant challenges associated with the large-scale change due to the EHR transition we underwent in 2015, including clinician burnout and fatigue, new EHR functionality, and the development of a new data reporting infrastructure and governance. The DOMQP continues to demonstrate that quality has a multifaceted role to play within a hospital.

As the health care environment continues to evolve, the need for this level of support for clinics will increase. The DOMQP is well positioned to provide continuing support to physicians and their practices in measuring and improving quality, with attention paid to such areas as coordination and efficacy of care, patient-reported outcomes, patient safety, and population health management. We believe that the DOMQP can serve as a model of a departmental quality program.

 

Acknowledgements: We would like to thank the department of medicine administration, faculty and support staff for their continuous effort and support in the work of clinical quality improvement.

Corresponding author: Sonali Desai, MD, MPH, Brigham & Women’s Hospital, 45 Francis St., PBB-B3, Boston, MA 02115, [email protected].

Financial disclosures: None.

From the Department of Medicine Quality Program, Brigham and Women’s Hospital (Drs. Pennant, McElrath, Coblyn, and Desai) and the Institute for Relevant Clinical Data Analytics, Boston Children’s Hospital, (Drs. Szent-Gyorgyi and Greenberg), Boston, MA.

 

Abstract

  • Objectives: To describe the department of medicine quality program (DOMQP) at Brigham and Women’s Hospital (BWH).
  • Methods: The program began in 2007 to engage physicians in local, specialty-specific quality efforts. The program has broadened its scope to include government mandates and hospital-wide priorities, such as maintenance of certification (MOC), “meaningful use (MU),” and medication reconciliation. More recently, we have evolved into a project-based program focusing on both chronic disease management and optimizing care pathways for high-risk inpatient conditions. Our key strategies are developing metrics, raising awareness, distilling information to front-line staff, highlighting relevant action items, and bringing feedback from front-line staff to hospital leadership.
  • Results: We have developed 21 metrics across 13 clinical divisions, with performance improvement seen in > 50% of metrics. In 2014, we leveraged our quality metrics to earn MOC credit, with 100 physicians across 10 divisions earning MOC points through the Practice Assessment option. Additionally, department physicians achieved 90% compliance with our institutional medication reconciliation policy. The percentage of physicians achieving stage 1 MU was 98% in 2013, 99% in 2014, and 100% achieved stage 2 MU in 2015.
  • Conclusion: Over the past 10 years, the DOMQP has played a unique role in promoting quality and serves as a model for QI within the hospital. We are well positioned to provide support to physicians and their practices as the health care environment continues to evolve.

Key words: quality improvement; quality measurement.

 

Within the past several years, the health care landscape in the United States has shifted considerably. New financial risk and quality-related incentive structures have been put in place, such as financial incentives to adopt electronic health records (EHRs) and to demonstrate “meaningful use (MU)” of these EHRs [1]. There is greater focus on value based payments, and accountable care organizations (ACOs) are proliferating [2–4]. Certification and training requirements have changed and require completion of performance improvement projects [5,6]. Upcoming changes to quality measurement and improvement through the Quality Payment Program will bring further changes to how clinicians are both monitored and incentivized or penalized [7]. The confluence of these efforts provides an impetus to incorporate quality measurement and improvement into the day to day practice of medicine.

In 2007, the department of medicine (DOM) at Brigham and Women’s Hospital (BWH), a teaching affiliate of Harvard Medical School and a founding member of Partners Healthcare, began a quality program (DOMQP) to engage physicians in local, specialty-specific quality efforts [8]. The program began by focusing on internally developed performance metrics to drive physician engagement. Later, the program expanded its focus to include more externally focused mandates from federal government and other accreditation bodies [9,10]. In this article, we discuss our efforts, including our early stage work and more recent focus in the areas of meaningful use, medication reconciliation, and maintenance of certification as well as our ongoing projects focused on chronic disease management and high-risk inpatient conditions.

Setting

The DOM has approximately 1400 physician faculty members and is the largest clinical department at BWH. The DOMQP is comprised of a medical director (0.35 FTE) and 2 project coordinators (2 FTE) who operate in a consultative capacity to liaison between the various levels of clinical leadership and frontline staff. The DOMQP serves 13 divisions: 11 medical specialties, primary care, and the hospitalist service.

Internally Driven, Specialty-Specific Quality Metrics

The early stages of our efforts focused on engaging clinical leadership and physicians, navigating the hospital’s information systems (IS) to develop quality reports within the EHR and implementing strategies for improvement. Until 30 May 2015, BWH utilized an internally developed EHR comprising a myriad of individual systems (eg, billing, electronic medication administration, clinical repository) that did not interface easily with one another. To resolve the IS challenges, we engaged many levels of the organization’s IS structure and requested updates or developed workaround solutions leading to the integration of the various datasets for comprehensive reporting. This resulted in a new agreement between the hospital and Partners that allowed for billing data to be sent to the same repository that housed the clinical data.

We developed at least one data report for each specialty division. The divisions selected a quality measure, and we worked with clinical leadership to define the numerator and denominator and identify any additional information they wanted in the report, eg, demographics, visit dates, labs. To produce the report, an IS consultant compiles the data elements on an excel spreadsheet. This spreadsheet is then manually chart reviewed by the DOMQP for accuracy before it is converted into a report. The report is then reviewed by the DOMQP to ensure the information is presented correctly and is easily interpreted, and then shared with the division champion(s), who determine how to message and introduce a proposed improvement effort before it is shared with the entire division.

Reports display a graph showing overall performance of the division and individual provider rates (an example is shown in Figure 1). Currently, most reports are sent unblinded; previously it was left to the discretion of the division chief. Patients are attributed to the provider they have seen most often during the reporting period. If there is a tie between 2 providers the patient is attributed to the most recent provider. Provider-level reports are also developed to show the physician’s performance over time as compared to the division’s overall performance. The reports list the provider’s patients and their performance status on the metric. We found that identifying patients not meeting the metric are essential to gaining clinician buy-in [11]. Physicians can be skeptical of baseline data if the rationale and details around how measurement was performed are not clearly communicated. Concrete examples with their own patient panel provide the proof they need and the relevant caveats and limitations of the data, in order to work toward improvement. We have seen clinic improvement on many metrics, with some clinics improving from 40% to over 88% in 2 to 3 years [12,13] (Table 1).

With accurate reports in place, we leverage all staff levels, from practice assistants to clinicians, whenever possible to improve by evaluating the performance data and setting goals. We use plan-do-study-act (PDSA) cycles to iterate performance improvement plans and tailor our efforts to the unique staffing levels and patient populations of each division [14,15]. For example, we have worked for years on improving blood pressure control among chronic kidney disease patients in our renal division. Initially, our first PDSA cycle involved mostly education and data sharing. When improvements were not seen, we did another PDSA cycle adding a brief form for physicians to fill out in the clinic for patients who had at least 3 blood pressure measurements that were not at goal. The sheet clearly displayed the 3 elevated blood pressures and asked the provider to note that day’s blood pressure as well as if the medication was adjusted or if the patient is not taking the medication(s) prescribed. Data from these worksheets informed efforts to focus on non–office-based blood pressure measurements [16]. Through a grant the renal division was able to give free blood pressure cuffs to patients for at-home monitoring.

In an effort to improve influenza and pneumococcal vaccination rates, we worked closely with front-line clinicians and staff to create improvement strategies tailored to the patient population and clinic staffing structure in 4 divisions: allergy (order and document flu vaccinations for asthma patients), rheumatology (point-of-care standing pneumococcal paper-orders for immunosuppressed patients), infectious diseases (a nurse-driven influenza protocol was implemented for HIV patients), and pulmonary (letters sent to chronic lung disease patients asking them to bring documentation of prior pneumococcal vaccination to their next visit). We saw increases in vaccination rates across all 4 divisions using varied approaches to reach our goals [17].

 

 

Meaningful Use

Our team was responsible for ensuring that the DOM succeeded in the federal government’s MU program. Millions of dollars were at risk to the hospital and to physicians based on performance on various MU metrics. The hospital made this a priority and set a goal for 90% of eligible providers to successfully certify in Stage 1 as meaningful users in the first year of MU.

To meet MU requirements, we categorized metrics by those requiring physician effort and those relying on clinic staff effort. We worked with division and departmental leadership to develop and implement an approach to improve performance on each metric. Monthly emails were sent with the current performance data, explanations of low-performing metrics, and actions to take for improvement (Figure 2). This information was reinforced at faculty presentations.

While working with the clinics to achieve meaningful use, we recognized that a range of staff members played a critical role in helping physicians meet MU metric targets. We worked with the clinic and departmental administrative leadership to set up a one-time bonus payment in 2013 to all DOM clinic staff, including medical assistants, licensed practical nurses, practice assistants, for appreciation of their significant efforts to help the physicians and the hospital achieve their MU goals. As health care delivery continues to rely more heavily on highly functional teams, acknowledgment of the efforts of non-clinical staff in helping front-line clinicians in achieving MU can help promote teamwork around a common goal.

Our partnership with the hospital MU team lead to a final tally of 98% of eligible providers meeting stage 1 in 2013, 99% in 2014 and 100% for the modified stage 2 in 2015 and 99% in 2016 for DOM specialty divisions, representing over 250 physicians.

Medication Reconciliation

The impetus for the hospital’s focus on medication reconciliation was the Joint Commission requirement that medications listed as prescribed in the EHR be reconciled with the patient at each visit [10]. The hospital’s quality and safety team created a hospital-wide metric aligned with hospital policy to measure how providers were reconciling medication lists during office visits. The medication reconciliation metric tallied visits when a medication change occurred (denominator) in which all of the medications originally prescribed by a physician in that specialty clinic were reconciled (numerator). This made it easier for specialists to buy-in to the metric because they were only responsible for the medications they prescribed and not the entire list. To meet the metric, physicians are required to take an action on each of the medications they prescribed by clicking taking, not taking, taking differently, or discontinuing. If the clinic has additional staff within the clinic to assist in the medication review process with the patient, physicians would receive credit for their actions once they had reviewed the medication list.

The metric BWH established to measure medication reconciliation was distinct and more rigorous than the medication reconciliation metric used to meet MU requirements. This presented a challenge in both physician communication regarding requirements and data sharing to drive performance improvement. Since the DOM is the largest and most prescribing department, we had to work with clinical and administrative leaders at the divisional level so that all staff understood the exact requirements and how to achieve them. When presenting to faculty we encountered many questions, and general resistance to more clicks in the EHR, as there was no universal “reconcile” button in the extant EHR. After breaking down the process into discrete components with staff and analyzing the data, we targeted outreach to the specific divisions and clinics that needed additional education and support. In our monthly email communication to division leadership, we displayed comparative data on all divisions within the DOM.

In late 2013, medication reconciliation was named as one of the main metrics hospital-wide that would be tied to a financial incentive for departments. The incentive was for money to be returned to the departments (not physicians) and the goal was common across the hospital. The hospital set a goal that 90% of visits with a medication change have all medications originally prescribed by that clinic reconciled. This led to a larger hospital-wide push to achieve success in medication reconciliation, including presentation of department-level data to senior leadership, such as the hospital president, chief medical officer, chief nursing officer, chief operating officer, and the various department chairs. Though the initial progress for this metric was slow, progress became evident across all divisions by the spring of 2014 (Figure 3) [18].

After we transitioned to a new vendor-based EHR in 2015, we developed a new medication reconciliation metric, aimed to align with Stage 2 MU requirements, and ensured all divisions had processes in place to review and reconcile medications. In our new EHR, medications lists are shared and viewable between ambulatory and inpatient environments and discharge summaries contain changes made to medications while inpatient. We have found that our performance on our new metric is 70.5% for February 2017 and we are continuing to educate physicians and staff on our new EHR functionality, our new electronic measurement and on workflows to help assist with improving performance with a short-term goal of 80%. We have found that our new EHR functionality does not match the ease of our old EHR functionality, which has made improvement on this metric a significant challenge for our clinicians and staff.

 

 

Maintenance of Certification

As of 2013, physicians who were board certified by the ABIM or ABMS were required to attain 20 practice assessment points showing participation in quality improvement activities for maintenance of certification (MOC). Although ABIM suspended the practice assessment requirement in 2015, it is likely that some component of quality improvement will be assessed in the future [5,19]. Prior to the changes in 2015, we were approached by the hospital leadership to roll out a new, streamlined process to leverage our existing quality metrics and established performance improvement plans in order to allow physicians to gain MOC credit. Each metric/project was vetted with 3 separate groups: the DOMQP, hospital leadership leading the MOC efforts, and Partners Healthcare, our overarching local health care system. We worked with our division physician champions and the physicians preparing for board re-certification to complete the one-time documentation required by the hospital for MOC. This process took approximately 6 months per project. Once the project was approved by all 3 channels, all physicians participating in the quality effort could get the practice assessment points by completing a simple attestation form.

For example, the DOMQP had already developed a metric with our allergy division measuring use of the Asthma Control Test during office-based visits. During one of our faculty meeting presentations, we shared that Partners now had a pilot program to count current QI projects at the hospital for MOC points. We asked for interested volunteers and worked with 1 or 2 physicians who most often were up for recertification that year. DOMQP provided the data that we already managed and prepopulated the required application and PowerPoint. The physician adds text regarding the clinical relevance of the project, how the improvement strategies in clinic evolved over time, and barriers faced in achieving their goal. The DOMQP sends the documents to Partners for review, and once the project is accepted, the division chief forwards attestation forms to participating physicians. To be eligible, a physician had to have data present in our monthly reports with at least 10 patients in their denominator. Some division chiefs included the additional criterion that a provider also had to be at goal performance set by the division in order to attest and receive MOC points. To date we have 15 projects approved with 206 physician attestations submitted for MOC points (Table 2). Through the aid of the Partners Healthcare Office of Continuing Professional Development we are now able to offer physicians assistants AMA PRA Category 1 credit for participation in division projects. Having an easy process that builds on existing DOMQP efforts has been a highly valued resource to our faculty. As the MOC process was de-emphasized by ABIM, we have shifted our focus to the changes surrounding the Quality Payment Program and are aligning our project-based work to support quality measurement and improvement.

Current Focus

After the stabilization of our new EHR and the re-creation of former reports, the program began to engage divisions in new measures. We still focus on chronic disease management and vaccinations but instead try to create a unified approach across multiple divisions within the DOM. Building upon our previous work in the renal division, over the past year we convened a hyper-tension work group comprising physician leads from endocrine, cardiology, renal, primary care, gerontology, neurology, pharmacy, and obstetrics. The goal of these meetings is to optimize blood pressure management across different patient populations by creating a centralized hospital approach with an algorithm agreed upon by the physician workgroup. We were able to secure additional internal grant funding to develop a pilot project where bluetooth blood pressure cuffs are given to eligible hypertensive patients in our pilot ambulatory practices. The daily blood pressure readings are transmitted into the EHR and a nurse practitioner or pharmacist contacts the patient at defined intervals to address any barriers and titrate medications as necessary. Analysis of the outcomes will be presented this fall. Similarly with vaccinations we are creating an automated order form within the EHR that will appear whenever a specialist places an order to start immunosuppressive medications. This will prompt the provider to order appropriate labs and vaccinations recommended for the course of treatment.

In addition to expanding upon previous metrics, we have expanded our scope to focus on patient safety measures, specifically, missed and delayed cancer diagnoses of the lung and colon. We are working on processes to track every patient with an abnormal finding from point of notification to completion of recommended follow-up at the appropriate intervals. Also we have now have 3 projects in the inpatient setting: chronic obstructive pulmonary disease (COPD) readmissions, and 2 standardized clinical assessment and management pathways (SCAMPs), one on acute kidney injury and the other on congestive heart failure [20]. The COPD project aims to have every patient admitted with COPD receive a pulmonary and respiratory consult during their stay and a follow-up visit with a pulmonologist. The goal of any SCAMP is to standardize care in an area where there is a lot of variability through the use of clinical pathways.

Communication Strategy

In order for the DOMQP to ensure that multiple quality requirements are met by all divisions, we have established a robust communication strategy with the goal of clear, concise, and relevant information-sharing with physicians and staff. We engage physicians through direct meetings, regular emails, and data reporting. The purpose of our outreach to the division faculty is threefold: (1) to educate physicians about hospital-wide programs, (2) to orient them to specific action items required for success, and (3) to funnel questions back to project leaders to ensure that the feedback of clinicians was incorporated into hospital wide quality initiatives. Our first challenge is to provide context for physicians about the project, be it based on accreditation, credentialing or a federal mandate. We work with the hospital project leaders to learn as much as possible about the efforts they are promoting so we can work in concert with them to highlight key messages to physicians.

Next, we establish a schedule to present at each clinical specialty faculty meeting on a regular basis (semi-annually to quarterly). At these meetings we present an overall picture of the key initiatives relevant to the division, identify milestones, offer clear timelines and prioritization of these projects, and narrow the focus of work to a few bulleted action items for all levels of the clinic staff to incorporate into its workflow. We then listen to questions and concerns and bring these back to the initiative leaders so that systematic changes can be made. Answers and updates are communicated back to the divisions, thus closing the communication loop. We interface with practice managers and clinical support staff to identify opportunities for them to support physicians in meeting initiative requirements [21,22].

In addition to presenting at faculty meetings, we present updates to departmental and hospital leadership, including vice chairs and division chiefs. These meetings include high-level data on performance as well as an opportunity to discuss the challenges we identified through our discussions with individual specialties. These forums are a good place to discuss overarching process issues or to disseminate answers to previous questions.

An important part of our communication plan involves our comprehensive monthly emails. For each initiative, we receive department level data on a monthly basis from the project leadership. We deconstruct these reports to enable us to evaluate our 13 divisions individually. We show performance at the physician level and highlight general areas where improvement is needed. We send monthly e-mails to division clinical and administrative leadership to apprise them of their division’s performance and inform them of areas that require concentrated effort. Depending on the initiative, we present data as a snapshot in time or trend over time. From 2012–2017 the DOMQP has helped to bridge the gap between large-scale rollouts of the new initiatives and the vast number of DOM physicians who required more specific education and tools to meet these new requirements.

 

 

Conclusion

The DOMQP has been working on quality within the department of medicine for the past 10 years. We have moved from initiating an internal quality program among the specialty providers, which required education among faculty and resolve to overcome many IS challenges, to serving as a resource for hospital-wide quality-related initiatives. We have developed a successful architecture for disseminating information and guiding faculty and administrative support toward success on a multitude of metrics that have implications on both finances and sound patient care. We have navigated the significant challenges associated with the large-scale change due to the EHR transition we underwent in 2015, including clinician burnout and fatigue, new EHR functionality, and the development of a new data reporting infrastructure and governance. The DOMQP continues to demonstrate that quality has a multifaceted role to play within a hospital.

As the health care environment continues to evolve, the need for this level of support for clinics will increase. The DOMQP is well positioned to provide continuing support to physicians and their practices in measuring and improving quality, with attention paid to such areas as coordination and efficacy of care, patient-reported outcomes, patient safety, and population health management. We believe that the DOMQP can serve as a model of a departmental quality program.

 

Acknowledgements: We would like to thank the department of medicine administration, faculty and support staff for their continuous effort and support in the work of clinical quality improvement.

Corresponding author: Sonali Desai, MD, MPH, Brigham & Women’s Hospital, 45 Francis St., PBB-B3, Boston, MA 02115, [email protected].

Financial disclosures: None.

References

1. Electronic health records (EHR) incentive programs. Accessed at www.cms.gov/Regulations-and Guidance/Legislation/EHRIncentivePrograms/index.html?redirect=/EHRIncentiveprograms/.

2. Accountable care organizations (ACO). Accessed at www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/ACO/index.html?redirect=/aco.

3. Quinn K. The 8 basic payment methods in health care. Ann Intern Med 2015;163:300–6.

4. Muhlestein D, McClellan M. Accountable care organizations in 2016: private and public-sector growth and dispersion. Health Aff blog 21 Apr 2016. Accessed at http://healthaffairs.org/blog/2016/04/21/accountable-care-organizations-in-2016-private-and-public-sector-growth-and-dispersion/.

5. ABIM. Maintenance of certification. Accessed 6 Oct 2015 at www.abim.org/maintenance-of-certification/.

6. Accreditation Council for Graduate Medical Education. CLER pathways to excellence. 2014. Accessed 6 Oct 2015 at www.acgme.org/acgmeweb/Portals/0/PDFs/CLER/CLER_Brochure.pdf.

7. Centers for Medicare and Medicaid Services. Quality payment program. Accessed 31 Mar 2017 at https://qpp.cms.gov/measures/quality.

8. Szent-Gyorgyi LE, Coblyn J, Turchin A, et al. Building a departmental quality program: a patient-based and provider-led approach. Acad Med 2011;86:314–20.

9. Brook RH, McGlynn EA, Shekelle PG. Defining and measuring quality of care: a perspective from US researchers. Int J Qual Health Care 2000;12:281–95.

10. The Joint Commision. National patient safety goals. Accessed at www.jointcommission.org/standards_information/npsgs.aspx.

11. Conway PH, Mostashari F, Clancy C. The future of quality measurement for improvement and accountability. JAMA 2013;309:2215–6.

12. Desai SP, Turchin A, Szent-Gyorgyi LE, et al. Routinely measuring and reporting pneumococcal vaccination among immunosuppressed rheumatology outpatients: the first step in improving quality. Rheumatology (Oxford) 2011;50:366–72.

13. Desai SP, Lu B, Szent-Gyorgyi LE, et al. Increasing pneumococcal vaccination for immunosuppressed patients: a cluster quality improvement trial. Arthritis Rheum 2013;65:39–47.

14. Deming Institute. PDSA cycle. Accessed at www.deming.org/theman/theories/pdsacycle.

15. McGlynn EA, Schneider EC, Kerr EA. Reimagining quality measurement. N Engl J Med 2014;371:2150–3.

16. Greenberg JO, Vakharia N, Szent-Gyorgyi LE, et al. Meaningful measurement: developing a measurement system to improve blood pressure control in patients with chronic kidney disease. J Am Med Inform Assoc 2013;20:e97–e101.

17. Pennant KN, Costa JJ, Fuhlbrigge AL, et al. Improving influenza and pneumococcal vaccination rates in ambulatory specialty practices. Open Forum Infect Dis 2015;2:ofv119.

18. Keogh C, Kachalia A, Fiumara K, et al. Ambulatory medication reconciliation: using a collaborative approach to process improvement at an academic medical center. Jt Comm J Qual Patient Saf 2016;42:186–94.

19. ABIM. Maintenance of certification FAQ. Accessed at www.abim.org/maintenance-of-certification/moc-faq/default.aspx.

20. Mendu ML, Ciociolo GR Jr, McLaughlin SR, et al. A decision-making algorithm for initiation and discontinuation of RRT in severe AKI. Clin J Am Soc Nephrol 2017;12:228–36.

21. Galvin RS, McGlynn EA. Using performance measurement to drive improvement: a road map for change. Med Care 2003;41:I48–60.

22. Glaser J, Hess R. Leveraging healthcare IT to improve operational performance. Healthc Financ Manage 2011;65:82–5.

References

1. Electronic health records (EHR) incentive programs. Accessed at www.cms.gov/Regulations-and Guidance/Legislation/EHRIncentivePrograms/index.html?redirect=/EHRIncentiveprograms/.

2. Accountable care organizations (ACO). Accessed at www.cms.gov/Medicare/Medicare-Fee-for-Service-Payment/ACO/index.html?redirect=/aco.

3. Quinn K. The 8 basic payment methods in health care. Ann Intern Med 2015;163:300–6.

4. Muhlestein D, McClellan M. Accountable care organizations in 2016: private and public-sector growth and dispersion. Health Aff blog 21 Apr 2016. Accessed at http://healthaffairs.org/blog/2016/04/21/accountable-care-organizations-in-2016-private-and-public-sector-growth-and-dispersion/.

5. ABIM. Maintenance of certification. Accessed 6 Oct 2015 at www.abim.org/maintenance-of-certification/.

6. Accreditation Council for Graduate Medical Education. CLER pathways to excellence. 2014. Accessed 6 Oct 2015 at www.acgme.org/acgmeweb/Portals/0/PDFs/CLER/CLER_Brochure.pdf.

7. Centers for Medicare and Medicaid Services. Quality payment program. Accessed 31 Mar 2017 at https://qpp.cms.gov/measures/quality.

8. Szent-Gyorgyi LE, Coblyn J, Turchin A, et al. Building a departmental quality program: a patient-based and provider-led approach. Acad Med 2011;86:314–20.

9. Brook RH, McGlynn EA, Shekelle PG. Defining and measuring quality of care: a perspective from US researchers. Int J Qual Health Care 2000;12:281–95.

10. The Joint Commision. National patient safety goals. Accessed at www.jointcommission.org/standards_information/npsgs.aspx.

11. Conway PH, Mostashari F, Clancy C. The future of quality measurement for improvement and accountability. JAMA 2013;309:2215–6.

12. Desai SP, Turchin A, Szent-Gyorgyi LE, et al. Routinely measuring and reporting pneumococcal vaccination among immunosuppressed rheumatology outpatients: the first step in improving quality. Rheumatology (Oxford) 2011;50:366–72.

13. Desai SP, Lu B, Szent-Gyorgyi LE, et al. Increasing pneumococcal vaccination for immunosuppressed patients: a cluster quality improvement trial. Arthritis Rheum 2013;65:39–47.

14. Deming Institute. PDSA cycle. Accessed at www.deming.org/theman/theories/pdsacycle.

15. McGlynn EA, Schneider EC, Kerr EA. Reimagining quality measurement. N Engl J Med 2014;371:2150–3.

16. Greenberg JO, Vakharia N, Szent-Gyorgyi LE, et al. Meaningful measurement: developing a measurement system to improve blood pressure control in patients with chronic kidney disease. J Am Med Inform Assoc 2013;20:e97–e101.

17. Pennant KN, Costa JJ, Fuhlbrigge AL, et al. Improving influenza and pneumococcal vaccination rates in ambulatory specialty practices. Open Forum Infect Dis 2015;2:ofv119.

18. Keogh C, Kachalia A, Fiumara K, et al. Ambulatory medication reconciliation: using a collaborative approach to process improvement at an academic medical center. Jt Comm J Qual Patient Saf 2016;42:186–94.

19. ABIM. Maintenance of certification FAQ. Accessed at www.abim.org/maintenance-of-certification/moc-faq/default.aspx.

20. Mendu ML, Ciociolo GR Jr, McLaughlin SR, et al. A decision-making algorithm for initiation and discontinuation of RRT in severe AKI. Clin J Am Soc Nephrol 2017;12:228–36.

21. Galvin RS, McGlynn EA. Using performance measurement to drive improvement: a road map for change. Med Care 2003;41:I48–60.

22. Glaser J, Hess R. Leveraging healthcare IT to improve operational performance. Healthc Financ Manage 2011;65:82–5.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Publications
Topics
Article Type
Display Headline
Making Quality Real for Physicians
Display Headline
Making Quality Real for Physicians
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default
Use ProPublica

Intensive Outpatient Care for High-Need Patients Did Not Impact Utilization or Costs

Article Type
Changed
Wed, 02/28/2018 - 13:55
Display Headline
Intensive Outpatient Care for High-Need Patients Did Not Impact Utilization or Costs

Study Overview

Objective. To determine the effect of an intensive out-patient program for high-need patients in a Veterans Affairs patient-centered medical home.

Design. Randomized controlled trial.

Setting and participants. The study was conducted at a single VA health care facility. Participants were 583 patients whose health care costs were in the top 5% for the facility during a 9-month eligibility period or whose risk for 1-year hospitalization risk as determined by the Care Assessment Need risk prediction algorithm [1] was in the top 5% for the facility. Patients were excluded if they were enrolled in mental health intensive case management program, home-based primary program or palliative care program, or if they were in an inpatient setting for more than half of the eligibility period. 150 patients were randomly assigned to the intensive outpatient group and the rest were assigned to receive standard VA-based primary care, which uses the patient-centered medical home model [2].

Intervention. The intensive outpatient care group received care from a multidisciplinary team comprising a nurse practitioner, physician, social worker, and recreational therapist. The enhanced care included comprehensive patient assessment, identification and tracking of patients’ health-related goals and priorities, assessment of physical function, cognitive function, social support, medical adherence and level of patient activation, and care management for medical and social needs. Frequent contacts using telephone lines and in-person visits as needed, weekly team discussions of high-acuity patients, and coordination of care with VA and non-VA clinicians also occurred. Additionally, the program offered interventions to support patients’ and caregivers’ quality of life, such as recreation therapy.

Main outcome measures. The main outcome measures were health care costs and utilization. Total health care costs included inpatient, outpatient, and fee-basis care provided outside the VA. Utilization measures included hospitalization frequency, hospital length of stay, and number of outpatient and emergency room visits. The study team examined cost and utilization patterns during the 16 months prior to initiation of the program (baseline period) and the 17 months after initiation of the program (follow-up period). The study also evaluated patient care experience in the intensive care group via survey at baseline and at 6 months after enrollment. The survey included items from the Patient Satisfaction questionnaire, the Patient Activation Measures tool, and questions about satisfaction with the intensive care program and the likelihood to recommend the program to others.

Main results. Of the 150 patients assigned to the intervention, 140 patients were included in the analysis after excluding those who were ineligible or died before the intervention began; there were 405 in the usual care group. Among the 140 patients, 96 engaged in the program and 60 completed the follow-up survey. The average patient age was 66 years and over 90% were male, with the majority living in an urban area. The average number of chronic conditions was approximately 10, and about two-thirds had a mental health diagnosis. In the follow-up period, patients in the intensive outpatient care group had a higher number of outpatient primary care visits (average of 21.8 visits [SD 17.4]) compared with the usual care group (average of 7.4 visits [SD 7.5]). The number of acute medical or surgical hospitalizations in the follow-up period was similar between the 2 groups, as was the number of emergency room visits. There were also no significant differences on other inpatient or outpatient health care utilization measures. The intensive outpatient care program was not associated with reduced costs of care when compared with usual care. For measurements on patient experience, the majority of patients who completed the survey (92%) indicated that they would recommend the program to others and 70% indicated that they were extremely satisfied with the program’s medical care.

Conclusions. Intensive outpatient care for high-need patients in this VA setting was not associated with a decrease in acute health care utilization or reduced costs. Patients in the intensive outpatient care program indicated that they were satisfied with the program and would recommend the program to others.

Commentary

Management of high-risk, high-cost patients continues to be a challenge for the health care system. High-users account for a disproportionate amount of health care costs. It would seem reasonable that attending to these patients’ complex needs by providing lower-cost supplemental primary care services early would reduce the need for more expensive care (eg, hospitalization) down-stream.

In this study, researchers examined the impact of an intensive outpatient care program targeting high-need veterans on health care utilization and costs. Although patients liked the program, the results demonstrated no reduction in either acute care utilization, including inpatient hospitalization or emergency room visits, or costs. The findings are consistent with a number of prior studies that have demonstrated limited impact of care coordination programs on cost and utilization [3] albeit demonstrating impact on other clinically relevant outcomes, including patient experience.

The study authors proposed a few factors that may have contributed to this finding. One was that a longer follow-up period may be needed to demonstrate improved outcomes. Another was that there may be a mismatch between the patients’ needs and the services offered by the program. In addition, the intensive out-patient services may have uncovered unmet needs that led to appropriate care, which could increase costs. The role of these factors might be examined using process measures, or with ongoing collection of administrative data, perhaps in a future study.

In interpreting this study, it is important to point out certain differences between this study and the typical randomized clinical trial. In this study, patients were not enrolled in a clinical trial at the time of the intensive outpatient care program—it was considered a quality improvement initiative at the time when the program was started. Thus, the study subjects may be different from the subjects likely to be included in a randomized clinical trial, where subjects must agree to participate in research in order to be part of the study. The patients in this study therefore likely resemble the patient population in a clinical setting rather than in a research study setting.

The other difference is that in addition to examining the impact of the intervention, the study tests the targeting strategy of the intervention—in this case, targeting patients with high need using algorithms already embedded in the VA. This strategy contrasts with a number of outpatient collaborative care interventions [4,5] that target specific medical conditions. While targeting high-utilizers makes sense from an economic point of view, such a group may be more diverse and have more diverse needs than a study population with a condition-specific profile, eg, patients with chronic disease and depression [4]. Two thirds of the study population had a mental health diagnosis, but the team did not include specific mental health personnel or care protocols for mental health management.

Because of its design as a quality improvement project, the study suffers from a number of shortcomings that may threaten its internal validity, namely, the low follow-up rate, the lack of a comparison group for some outcomes, and perhaps, less assurance that participants were treated equally except for the study intervention.

Applications for Clinical Practice

The study adds to the current literature on interventions for improving care and reducing costs for patients with high health care needs. As health care costs continue to escalate, implementing strategies to improve efficiency continues to be a priority. The intensive outpatient care program may not be the solution for curbing costs for the study population at this time; perhaps follow-up studies that assess its impact on other relevant clinical outcomes with longer follow-up may tell a different story.

 

—William W. Hung, MD, MPH

References

1. Wang L, Porter B, Maynard C, et al. Predicting risk of hospitalization or death among patients receiving primary care in the Veterans Health Administration. Med Care 2013;51:368–73.

2. Yano EM, Bair MJ, Carrasquillo O, et al. Patient Aligned Care Teams (PACT): VA’s journey to implement patient-centered medical homes. J Gen Intern Med 2014;29 Suppl 2:S547–9.

3. Brown RS, Peikes D, Peterson G, et al. Six features of Medicare coordinated care demonstration programs that cut hospital admissions of high-risk patients. Health Aff (Millwood) 2012;31:1156–66.

4. Katon WJ, Lin EHB, Von Korff M, et al. Collaborative care for patients with depression and chronic illnesses. N Engl J Med 2010;363:2611–20.

5. Callahan CM, Boustani MA, Unverzagt FW, et al. Effectiveness of collaborative care for older adults with Alzheimer disease in primary care: a randomized controlled trial. JAMA 2006;295:2148–57.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Topics
Sections

Study Overview

Objective. To determine the effect of an intensive out-patient program for high-need patients in a Veterans Affairs patient-centered medical home.

Design. Randomized controlled trial.

Setting and participants. The study was conducted at a single VA health care facility. Participants were 583 patients whose health care costs were in the top 5% for the facility during a 9-month eligibility period or whose risk for 1-year hospitalization risk as determined by the Care Assessment Need risk prediction algorithm [1] was in the top 5% for the facility. Patients were excluded if they were enrolled in mental health intensive case management program, home-based primary program or palliative care program, or if they were in an inpatient setting for more than half of the eligibility period. 150 patients were randomly assigned to the intensive outpatient group and the rest were assigned to receive standard VA-based primary care, which uses the patient-centered medical home model [2].

Intervention. The intensive outpatient care group received care from a multidisciplinary team comprising a nurse practitioner, physician, social worker, and recreational therapist. The enhanced care included comprehensive patient assessment, identification and tracking of patients’ health-related goals and priorities, assessment of physical function, cognitive function, social support, medical adherence and level of patient activation, and care management for medical and social needs. Frequent contacts using telephone lines and in-person visits as needed, weekly team discussions of high-acuity patients, and coordination of care with VA and non-VA clinicians also occurred. Additionally, the program offered interventions to support patients’ and caregivers’ quality of life, such as recreation therapy.

Main outcome measures. The main outcome measures were health care costs and utilization. Total health care costs included inpatient, outpatient, and fee-basis care provided outside the VA. Utilization measures included hospitalization frequency, hospital length of stay, and number of outpatient and emergency room visits. The study team examined cost and utilization patterns during the 16 months prior to initiation of the program (baseline period) and the 17 months after initiation of the program (follow-up period). The study also evaluated patient care experience in the intensive care group via survey at baseline and at 6 months after enrollment. The survey included items from the Patient Satisfaction questionnaire, the Patient Activation Measures tool, and questions about satisfaction with the intensive care program and the likelihood to recommend the program to others.

Main results. Of the 150 patients assigned to the intervention, 140 patients were included in the analysis after excluding those who were ineligible or died before the intervention began; there were 405 in the usual care group. Among the 140 patients, 96 engaged in the program and 60 completed the follow-up survey. The average patient age was 66 years and over 90% were male, with the majority living in an urban area. The average number of chronic conditions was approximately 10, and about two-thirds had a mental health diagnosis. In the follow-up period, patients in the intensive outpatient care group had a higher number of outpatient primary care visits (average of 21.8 visits [SD 17.4]) compared with the usual care group (average of 7.4 visits [SD 7.5]). The number of acute medical or surgical hospitalizations in the follow-up period was similar between the 2 groups, as was the number of emergency room visits. There were also no significant differences on other inpatient or outpatient health care utilization measures. The intensive outpatient care program was not associated with reduced costs of care when compared with usual care. For measurements on patient experience, the majority of patients who completed the survey (92%) indicated that they would recommend the program to others and 70% indicated that they were extremely satisfied with the program’s medical care.

Conclusions. Intensive outpatient care for high-need patients in this VA setting was not associated with a decrease in acute health care utilization or reduced costs. Patients in the intensive outpatient care program indicated that they were satisfied with the program and would recommend the program to others.

Commentary

Management of high-risk, high-cost patients continues to be a challenge for the health care system. High-users account for a disproportionate amount of health care costs. It would seem reasonable that attending to these patients’ complex needs by providing lower-cost supplemental primary care services early would reduce the need for more expensive care (eg, hospitalization) down-stream.

In this study, researchers examined the impact of an intensive outpatient care program targeting high-need veterans on health care utilization and costs. Although patients liked the program, the results demonstrated no reduction in either acute care utilization, including inpatient hospitalization or emergency room visits, or costs. The findings are consistent with a number of prior studies that have demonstrated limited impact of care coordination programs on cost and utilization [3] albeit demonstrating impact on other clinically relevant outcomes, including patient experience.

The study authors proposed a few factors that may have contributed to this finding. One was that a longer follow-up period may be needed to demonstrate improved outcomes. Another was that there may be a mismatch between the patients’ needs and the services offered by the program. In addition, the intensive out-patient services may have uncovered unmet needs that led to appropriate care, which could increase costs. The role of these factors might be examined using process measures, or with ongoing collection of administrative data, perhaps in a future study.

In interpreting this study, it is important to point out certain differences between this study and the typical randomized clinical trial. In this study, patients were not enrolled in a clinical trial at the time of the intensive outpatient care program—it was considered a quality improvement initiative at the time when the program was started. Thus, the study subjects may be different from the subjects likely to be included in a randomized clinical trial, where subjects must agree to participate in research in order to be part of the study. The patients in this study therefore likely resemble the patient population in a clinical setting rather than in a research study setting.

The other difference is that in addition to examining the impact of the intervention, the study tests the targeting strategy of the intervention—in this case, targeting patients with high need using algorithms already embedded in the VA. This strategy contrasts with a number of outpatient collaborative care interventions [4,5] that target specific medical conditions. While targeting high-utilizers makes sense from an economic point of view, such a group may be more diverse and have more diverse needs than a study population with a condition-specific profile, eg, patients with chronic disease and depression [4]. Two thirds of the study population had a mental health diagnosis, but the team did not include specific mental health personnel or care protocols for mental health management.

Because of its design as a quality improvement project, the study suffers from a number of shortcomings that may threaten its internal validity, namely, the low follow-up rate, the lack of a comparison group for some outcomes, and perhaps, less assurance that participants were treated equally except for the study intervention.

Applications for Clinical Practice

The study adds to the current literature on interventions for improving care and reducing costs for patients with high health care needs. As health care costs continue to escalate, implementing strategies to improve efficiency continues to be a priority. The intensive outpatient care program may not be the solution for curbing costs for the study population at this time; perhaps follow-up studies that assess its impact on other relevant clinical outcomes with longer follow-up may tell a different story.

 

—William W. Hung, MD, MPH

Study Overview

Objective. To determine the effect of an intensive out-patient program for high-need patients in a Veterans Affairs patient-centered medical home.

Design. Randomized controlled trial.

Setting and participants. The study was conducted at a single VA health care facility. Participants were 583 patients whose health care costs were in the top 5% for the facility during a 9-month eligibility period or whose risk for 1-year hospitalization risk as determined by the Care Assessment Need risk prediction algorithm [1] was in the top 5% for the facility. Patients were excluded if they were enrolled in mental health intensive case management program, home-based primary program or palliative care program, or if they were in an inpatient setting for more than half of the eligibility period. 150 patients were randomly assigned to the intensive outpatient group and the rest were assigned to receive standard VA-based primary care, which uses the patient-centered medical home model [2].

Intervention. The intensive outpatient care group received care from a multidisciplinary team comprising a nurse practitioner, physician, social worker, and recreational therapist. The enhanced care included comprehensive patient assessment, identification and tracking of patients’ health-related goals and priorities, assessment of physical function, cognitive function, social support, medical adherence and level of patient activation, and care management for medical and social needs. Frequent contacts using telephone lines and in-person visits as needed, weekly team discussions of high-acuity patients, and coordination of care with VA and non-VA clinicians also occurred. Additionally, the program offered interventions to support patients’ and caregivers’ quality of life, such as recreation therapy.

Main outcome measures. The main outcome measures were health care costs and utilization. Total health care costs included inpatient, outpatient, and fee-basis care provided outside the VA. Utilization measures included hospitalization frequency, hospital length of stay, and number of outpatient and emergency room visits. The study team examined cost and utilization patterns during the 16 months prior to initiation of the program (baseline period) and the 17 months after initiation of the program (follow-up period). The study also evaluated patient care experience in the intensive care group via survey at baseline and at 6 months after enrollment. The survey included items from the Patient Satisfaction questionnaire, the Patient Activation Measures tool, and questions about satisfaction with the intensive care program and the likelihood to recommend the program to others.

Main results. Of the 150 patients assigned to the intervention, 140 patients were included in the analysis after excluding those who were ineligible or died before the intervention began; there were 405 in the usual care group. Among the 140 patients, 96 engaged in the program and 60 completed the follow-up survey. The average patient age was 66 years and over 90% were male, with the majority living in an urban area. The average number of chronic conditions was approximately 10, and about two-thirds had a mental health diagnosis. In the follow-up period, patients in the intensive outpatient care group had a higher number of outpatient primary care visits (average of 21.8 visits [SD 17.4]) compared with the usual care group (average of 7.4 visits [SD 7.5]). The number of acute medical or surgical hospitalizations in the follow-up period was similar between the 2 groups, as was the number of emergency room visits. There were also no significant differences on other inpatient or outpatient health care utilization measures. The intensive outpatient care program was not associated with reduced costs of care when compared with usual care. For measurements on patient experience, the majority of patients who completed the survey (92%) indicated that they would recommend the program to others and 70% indicated that they were extremely satisfied with the program’s medical care.

Conclusions. Intensive outpatient care for high-need patients in this VA setting was not associated with a decrease in acute health care utilization or reduced costs. Patients in the intensive outpatient care program indicated that they were satisfied with the program and would recommend the program to others.

Commentary

Management of high-risk, high-cost patients continues to be a challenge for the health care system. High-users account for a disproportionate amount of health care costs. It would seem reasonable that attending to these patients’ complex needs by providing lower-cost supplemental primary care services early would reduce the need for more expensive care (eg, hospitalization) down-stream.

In this study, researchers examined the impact of an intensive outpatient care program targeting high-need veterans on health care utilization and costs. Although patients liked the program, the results demonstrated no reduction in either acute care utilization, including inpatient hospitalization or emergency room visits, or costs. The findings are consistent with a number of prior studies that have demonstrated limited impact of care coordination programs on cost and utilization [3] albeit demonstrating impact on other clinically relevant outcomes, including patient experience.

The study authors proposed a few factors that may have contributed to this finding. One was that a longer follow-up period may be needed to demonstrate improved outcomes. Another was that there may be a mismatch between the patients’ needs and the services offered by the program. In addition, the intensive out-patient services may have uncovered unmet needs that led to appropriate care, which could increase costs. The role of these factors might be examined using process measures, or with ongoing collection of administrative data, perhaps in a future study.

In interpreting this study, it is important to point out certain differences between this study and the typical randomized clinical trial. In this study, patients were not enrolled in a clinical trial at the time of the intensive outpatient care program—it was considered a quality improvement initiative at the time when the program was started. Thus, the study subjects may be different from the subjects likely to be included in a randomized clinical trial, where subjects must agree to participate in research in order to be part of the study. The patients in this study therefore likely resemble the patient population in a clinical setting rather than in a research study setting.

The other difference is that in addition to examining the impact of the intervention, the study tests the targeting strategy of the intervention—in this case, targeting patients with high need using algorithms already embedded in the VA. This strategy contrasts with a number of outpatient collaborative care interventions [4,5] that target specific medical conditions. While targeting high-utilizers makes sense from an economic point of view, such a group may be more diverse and have more diverse needs than a study population with a condition-specific profile, eg, patients with chronic disease and depression [4]. Two thirds of the study population had a mental health diagnosis, but the team did not include specific mental health personnel or care protocols for mental health management.

Because of its design as a quality improvement project, the study suffers from a number of shortcomings that may threaten its internal validity, namely, the low follow-up rate, the lack of a comparison group for some outcomes, and perhaps, less assurance that participants were treated equally except for the study intervention.

Applications for Clinical Practice

The study adds to the current literature on interventions for improving care and reducing costs for patients with high health care needs. As health care costs continue to escalate, implementing strategies to improve efficiency continues to be a priority. The intensive outpatient care program may not be the solution for curbing costs for the study population at this time; perhaps follow-up studies that assess its impact on other relevant clinical outcomes with longer follow-up may tell a different story.

 

—William W. Hung, MD, MPH

References

1. Wang L, Porter B, Maynard C, et al. Predicting risk of hospitalization or death among patients receiving primary care in the Veterans Health Administration. Med Care 2013;51:368–73.

2. Yano EM, Bair MJ, Carrasquillo O, et al. Patient Aligned Care Teams (PACT): VA’s journey to implement patient-centered medical homes. J Gen Intern Med 2014;29 Suppl 2:S547–9.

3. Brown RS, Peikes D, Peterson G, et al. Six features of Medicare coordinated care demonstration programs that cut hospital admissions of high-risk patients. Health Aff (Millwood) 2012;31:1156–66.

4. Katon WJ, Lin EHB, Von Korff M, et al. Collaborative care for patients with depression and chronic illnesses. N Engl J Med 2010;363:2611–20.

5. Callahan CM, Boustani MA, Unverzagt FW, et al. Effectiveness of collaborative care for older adults with Alzheimer disease in primary care: a randomized controlled trial. JAMA 2006;295:2148–57.

References

1. Wang L, Porter B, Maynard C, et al. Predicting risk of hospitalization or death among patients receiving primary care in the Veterans Health Administration. Med Care 2013;51:368–73.

2. Yano EM, Bair MJ, Carrasquillo O, et al. Patient Aligned Care Teams (PACT): VA’s journey to implement patient-centered medical homes. J Gen Intern Med 2014;29 Suppl 2:S547–9.

3. Brown RS, Peikes D, Peterson G, et al. Six features of Medicare coordinated care demonstration programs that cut hospital admissions of high-risk patients. Health Aff (Millwood) 2012;31:1156–66.

4. Katon WJ, Lin EHB, Von Korff M, et al. Collaborative care for patients with depression and chronic illnesses. N Engl J Med 2010;363:2611–20.

5. Callahan CM, Boustani MA, Unverzagt FW, et al. Effectiveness of collaborative care for older adults with Alzheimer disease in primary care: a randomized controlled trial. JAMA 2006;295:2148–57.

Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Issue
Journal of Clinical Outcomes Management - May 2017, Vol. 24, No. 5
Publications
Publications
Topics
Article Type
Display Headline
Intensive Outpatient Care for High-Need Patients Did Not Impact Utilization or Costs
Display Headline
Intensive Outpatient Care for High-Need Patients Did Not Impact Utilization or Costs
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Disqus Comments
Default

Cutaneous Metastasis of a Pulmonary Carcinoid Tumor

Article Type
Changed
Thu, 01/10/2019 - 13:41
Display Headline
Cutaneous Metastasis of a Pulmonary Carcinoid Tumor

Case Report

A 72-year-old white man with a history of pancreatic adenocarcinoma presented for Mohs micrographic surgery of a basal cell carcinoma on the right helix. On the day of the surgery, the patient reported a new, rapidly growing, exquisitely painful lesion on the cheek of 3 to 4 weeks’ duration. Physical examination revealed a 0.8×0.8×0.8-cm, extremely tender, firm, pink papule on the right preauricular cheek. A horizontal deep shave excision was done and the histopathology was remarkable for neoplastic cells with necrosis in the dermis. We observed dermal cellular infiltrates in the form of sheets and nodules, some showing central necrosis (Figure 1). At higher magnification, a trabecular arrangement of cells was seen. These cells had a moderate amount of cytoplasm with eccentric nuclei and rare nucleoli (Figure 2). Mitotic figures were seen at higher magnification (Figure 3). Immunohistochemistry of the neoplastic cells exhibited similar positive staining for the neuroendocrine markers chromogranin A and synaptophysin (Figure 4). Staining of the neoplastic cells also was positive for thyroid transcription factor 1 (TTF-1) and cancer antigen 19-9. Villin and caudal type homeobox 2 stains were negative. These results were consistent with cutaneous metastasis from a known pulmonary carcinoid tumor.

Figure 1. Cutaneous metastasis of a pulmonary carcinoid tumor. A dermal cellular infiltrate of sheets and nodules, some showing central necrosis (H&E, original magnification ×40).

Figure 2. Cutaneous metastasis of a pulmonary carcinoid tumor. A trabecular arrangement of cells with a moderate amount of cytoplasm with eccentric nuclei and rare nucleoli (H&E, original magnification ×400).

Figure 3. Cutaneous metastasis of a pulmonary carcinoid tumor. Mitotic figures can be seen (H&E, original magnification ×400).

Figure 4. Cutaneous metastasis of a pulmonary carcinoid tumor. Immunohistochemistry was positive for chromogranin A (original magnification ×400).

On further review of the patient’s medical history, it was discovered that he had undergone a Whipple procedure with adjuvant chemotherapy and radiation for pancreatic adenocarcinoma approximately 4 years prior to the current presentation. He was then followed by oncology, and 3 years later a chest computed tomography suggested possible disease progression with a new pulmonary metastasis. This pulmonary lesion was biopsied and immunologic staining was consistent with a primary neuroendocrine neoplasm of the lung, a new carcinoid tumor. The tissue was positive for cytokeratin (CK) 7,TTF-1, cancer antigen 19-9, CD56, synaptophysin, and chromogranin A, and was negative for villin and CK20. By the time he was seen in our clinic, several trials of chemotherapy had failed. Serial computed tomography subsequently demonstrated progression of the lung disease and he later developed malignant pleural effusions. Approximately 6 months after the cutaneous carcinoid metastasis was diagnosed, the patient died of respiratory failure.

 

 

Comment

Carcinoid tumors are uncommon neoplasms of neuroendocrine origin that generally arise in the gastrointestinal or bronchopulmonary tracts. Metastases from these primary neoplasms more commonly affect the regional lymph nodes or viscera, with rare reports of cutaneous metastases to the skin. The true incidence of carcinoid tumors with metastasis to the skin is unknown because it is limited to single case reports in the literature.

The clinical presentation of cutaneous carcinoid metastases has been reported most commonly as firm papules of varying sizes with no specific site predilection.1 The color of these lesions has ranged from erythematous to violaceous to brown.2 Several of the reported cases were noted to be extremely tender and painful, while other reports of lesions were noted to be asymptomatic or only mildly pruritic.3-7

Carcinoid syndrome is more common with neoplasms present within the gastrointestinal tract, but it also has been reported with large bronchial carcinoid tumors and with metastatic disease.8,9 Paroxysmal flushing is the most prominent cutaneous manifestation of this syndrome, occurring in 75% of patients.10,11 Other common symptoms include patchy cyanosis, telangiectasia, and pellagralike skin lesions.3 Carcinoid syndrome secondary to bronchial adenomas is thought to differ from gastrointestinal carcinoid neoplasms in that it has prolonged flushing (hours to days instead of minutes) and is characterized by marked anxiety, fever, disorientation, sweating, and lacrimation.8,9

Many cases of cutaneous carcinoid metastases have been accompanied by reports of exquisite tenderness,7 similar to our patient. The pathogenesis of the pain in these lesions is still unclear, but several hypotheses have been established. It has been postulated that perineural invasion by the tumor is responsible for the pain; however, this finding has been inconsistent, as neural involvement also has been present in nonpainful lesions.2,5,7,12 Another theory for the pain is that it is secondary to the release of vasoactive substances and peptide hormones from the carcinoid cells, such as kallikrein and serotonin. Lastly, local tissue necrosis and fibrosis also have been suggested as possible etiologies.7

The histology of cutaneous carcinoid metastases typically resembles the primary lesion and may demonstrate fascicles of spindle cells with focal areas of necrosis, mild atypia, and a relatively low mitotic rate.10 Other neoplasms such as Merkel cell carcinoma and carcinoidlike sebaceous carcinoma should be considered in the differential diagnosis. A primary malignant peripheral primitive neuroectodermal tumor or a primary cutaneous carcinoid tumor is less common but should be considered. Differing from carcinoid tumors, Merkel cell carcinomas usually have a higher mitotic rate and positive staining for CK20. The sebaceous neoplasms with a carcinoidlike pattern may appear histologically similar, requiring immunohistochemical evaluation with monoclonal antibodies such as D2-40.13 A diffuse granular cytoplasmic reaction to chromogranin A is characteristic of carcinoid tumors. Synaptophysin and TTF-1 also are positive in carcinoid tumors, with TTF-1 being highly specific for neuroendocrine tumors of the lung.10

Cutaneous metastases of internal malignancies are more common from carcinomas of the lungs, gastrointestinal tract, and breasts.5 Occasionally, the cutaneous metastasis will develop directly over the underlying malignancy. Our case of cutaneous metastasis of a carcinoid tumor presented as an exquisitely tender and painful papule on the cheek. The histology of the lesion was consistent with the known carcinoid tumor of the lung. Because these lesions are extremely uncommon, it is imperative to obtain an accurate clinical history and use the appropriate immunohistochemical panel to correctly diagnose these metastases.

References
  1. Blochin E, Stein JA, Wang NS. Atypical carcinoid metastasis to the skin. Am J Dermatopathol. 2010;32:735-739.
  2. Rodriguez G, Villamizar R. Carcinoid tumor with skin metastasis. Am J Dermatopathol. 1992;14:263-269.
  3. Archer CB, Rauch HJ, Allen MH, et al. Ultrastructural features of metastatic cutaneous carcinoid. J Cutan Pathol. 1984;11:485-490.
  4. Archer CB, Wells RS, MacDonald DM. Metastatic cutaneous carcinoid. J Am Acad Dermatol. 1985;13(2, pt 2):363-366.
  5. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis:a meta-analysis of data. South Med J. 2003;96:164-167.
  6. Oleksowicz L, Morris JC, Phelps RG, et al. Pulmonary carcinoid presenting as multiple subcutaneous nodules. Tumori. 1990;76:44-47.
  7. Zuetenhorst JM, van Velthuysen ML, Rutgers EJ, et al. Pathogenesis and treatment of pain caused by skin metastases in neuroendocrine tumours. Neth J Med. 2002;60:207-211.
  8. Melmon KL. Kinins: one of the many mediators of the carcinoid spectrum. Gastroenterology. 1968;55:545-548.
  9. Zuetenhorst JM, Taal BG. Metastatic carcinoid tumors: a clinical review. Oncologist. 2005;10:123-131.
  10. Sabir S, James WD, Schuchter LM. Cutaneous manifestations of cancer. Curr Opin Oncol. 1999;11:139-144.
  11. Braverman IM. Skin manifestations of internal malignancy. Clin Geriatr Med. 2002;18:1-19.
  12. Santi R, Massi D, Mazzoni F, et al. Skin metastasis from typical carcinoid tumor of the lung. J Cutan Pathol. 2008;35:418-422.
  13. Kazakov DV, Kutzner H, Rütten A, et al. Carcinoid-like pattern in sebaceous neoplasms. another distinctive, previously unrecognized pattern in extraocular sebaceous carcinoma and sebaceoma. Am J Dermatopathol. 2005;27:195-203.
Article PDF
Author and Disclosure Information

Dr. Falto-Aizpurua is from the Department of Dermatology, University of Puerto Rico, Medical Sciences Campus, San Juan. Drs. Seyfer, Krishnan, and Orengo are from Baylor College of Medicine, Houston, Texas.

The authors report no conflict of interest.

Correspondence: Ida Orengo, MD, 1977 Butler Blvd, Ste E6.200, Houston, TX 77030 ([email protected]).

Issue
Cutis - 99(5)
Publications
Topics
Page Number
E13-E15
Sections
Author and Disclosure Information

Dr. Falto-Aizpurua is from the Department of Dermatology, University of Puerto Rico, Medical Sciences Campus, San Juan. Drs. Seyfer, Krishnan, and Orengo are from Baylor College of Medicine, Houston, Texas.

The authors report no conflict of interest.

Correspondence: Ida Orengo, MD, 1977 Butler Blvd, Ste E6.200, Houston, TX 77030 ([email protected]).

Author and Disclosure Information

Dr. Falto-Aizpurua is from the Department of Dermatology, University of Puerto Rico, Medical Sciences Campus, San Juan. Drs. Seyfer, Krishnan, and Orengo are from Baylor College of Medicine, Houston, Texas.

The authors report no conflict of interest.

Correspondence: Ida Orengo, MD, 1977 Butler Blvd, Ste E6.200, Houston, TX 77030 ([email protected]).

Article PDF
Article PDF

Case Report

A 72-year-old white man with a history of pancreatic adenocarcinoma presented for Mohs micrographic surgery of a basal cell carcinoma on the right helix. On the day of the surgery, the patient reported a new, rapidly growing, exquisitely painful lesion on the cheek of 3 to 4 weeks’ duration. Physical examination revealed a 0.8×0.8×0.8-cm, extremely tender, firm, pink papule on the right preauricular cheek. A horizontal deep shave excision was done and the histopathology was remarkable for neoplastic cells with necrosis in the dermis. We observed dermal cellular infiltrates in the form of sheets and nodules, some showing central necrosis (Figure 1). At higher magnification, a trabecular arrangement of cells was seen. These cells had a moderate amount of cytoplasm with eccentric nuclei and rare nucleoli (Figure 2). Mitotic figures were seen at higher magnification (Figure 3). Immunohistochemistry of the neoplastic cells exhibited similar positive staining for the neuroendocrine markers chromogranin A and synaptophysin (Figure 4). Staining of the neoplastic cells also was positive for thyroid transcription factor 1 (TTF-1) and cancer antigen 19-9. Villin and caudal type homeobox 2 stains were negative. These results were consistent with cutaneous metastasis from a known pulmonary carcinoid tumor.

Figure 1. Cutaneous metastasis of a pulmonary carcinoid tumor. A dermal cellular infiltrate of sheets and nodules, some showing central necrosis (H&E, original magnification ×40).

Figure 2. Cutaneous metastasis of a pulmonary carcinoid tumor. A trabecular arrangement of cells with a moderate amount of cytoplasm with eccentric nuclei and rare nucleoli (H&E, original magnification ×400).

Figure 3. Cutaneous metastasis of a pulmonary carcinoid tumor. Mitotic figures can be seen (H&E, original magnification ×400).

Figure 4. Cutaneous metastasis of a pulmonary carcinoid tumor. Immunohistochemistry was positive for chromogranin A (original magnification ×400).

On further review of the patient’s medical history, it was discovered that he had undergone a Whipple procedure with adjuvant chemotherapy and radiation for pancreatic adenocarcinoma approximately 4 years prior to the current presentation. He was then followed by oncology, and 3 years later a chest computed tomography suggested possible disease progression with a new pulmonary metastasis. This pulmonary lesion was biopsied and immunologic staining was consistent with a primary neuroendocrine neoplasm of the lung, a new carcinoid tumor. The tissue was positive for cytokeratin (CK) 7,TTF-1, cancer antigen 19-9, CD56, synaptophysin, and chromogranin A, and was negative for villin and CK20. By the time he was seen in our clinic, several trials of chemotherapy had failed. Serial computed tomography subsequently demonstrated progression of the lung disease and he later developed malignant pleural effusions. Approximately 6 months after the cutaneous carcinoid metastasis was diagnosed, the patient died of respiratory failure.

 

 

Comment

Carcinoid tumors are uncommon neoplasms of neuroendocrine origin that generally arise in the gastrointestinal or bronchopulmonary tracts. Metastases from these primary neoplasms more commonly affect the regional lymph nodes or viscera, with rare reports of cutaneous metastases to the skin. The true incidence of carcinoid tumors with metastasis to the skin is unknown because it is limited to single case reports in the literature.

The clinical presentation of cutaneous carcinoid metastases has been reported most commonly as firm papules of varying sizes with no specific site predilection.1 The color of these lesions has ranged from erythematous to violaceous to brown.2 Several of the reported cases were noted to be extremely tender and painful, while other reports of lesions were noted to be asymptomatic or only mildly pruritic.3-7

Carcinoid syndrome is more common with neoplasms present within the gastrointestinal tract, but it also has been reported with large bronchial carcinoid tumors and with metastatic disease.8,9 Paroxysmal flushing is the most prominent cutaneous manifestation of this syndrome, occurring in 75% of patients.10,11 Other common symptoms include patchy cyanosis, telangiectasia, and pellagralike skin lesions.3 Carcinoid syndrome secondary to bronchial adenomas is thought to differ from gastrointestinal carcinoid neoplasms in that it has prolonged flushing (hours to days instead of minutes) and is characterized by marked anxiety, fever, disorientation, sweating, and lacrimation.8,9

Many cases of cutaneous carcinoid metastases have been accompanied by reports of exquisite tenderness,7 similar to our patient. The pathogenesis of the pain in these lesions is still unclear, but several hypotheses have been established. It has been postulated that perineural invasion by the tumor is responsible for the pain; however, this finding has been inconsistent, as neural involvement also has been present in nonpainful lesions.2,5,7,12 Another theory for the pain is that it is secondary to the release of vasoactive substances and peptide hormones from the carcinoid cells, such as kallikrein and serotonin. Lastly, local tissue necrosis and fibrosis also have been suggested as possible etiologies.7

The histology of cutaneous carcinoid metastases typically resembles the primary lesion and may demonstrate fascicles of spindle cells with focal areas of necrosis, mild atypia, and a relatively low mitotic rate.10 Other neoplasms such as Merkel cell carcinoma and carcinoidlike sebaceous carcinoma should be considered in the differential diagnosis. A primary malignant peripheral primitive neuroectodermal tumor or a primary cutaneous carcinoid tumor is less common but should be considered. Differing from carcinoid tumors, Merkel cell carcinomas usually have a higher mitotic rate and positive staining for CK20. The sebaceous neoplasms with a carcinoidlike pattern may appear histologically similar, requiring immunohistochemical evaluation with monoclonal antibodies such as D2-40.13 A diffuse granular cytoplasmic reaction to chromogranin A is characteristic of carcinoid tumors. Synaptophysin and TTF-1 also are positive in carcinoid tumors, with TTF-1 being highly specific for neuroendocrine tumors of the lung.10

Cutaneous metastases of internal malignancies are more common from carcinomas of the lungs, gastrointestinal tract, and breasts.5 Occasionally, the cutaneous metastasis will develop directly over the underlying malignancy. Our case of cutaneous metastasis of a carcinoid tumor presented as an exquisitely tender and painful papule on the cheek. The histology of the lesion was consistent with the known carcinoid tumor of the lung. Because these lesions are extremely uncommon, it is imperative to obtain an accurate clinical history and use the appropriate immunohistochemical panel to correctly diagnose these metastases.

Case Report

A 72-year-old white man with a history of pancreatic adenocarcinoma presented for Mohs micrographic surgery of a basal cell carcinoma on the right helix. On the day of the surgery, the patient reported a new, rapidly growing, exquisitely painful lesion on the cheek of 3 to 4 weeks’ duration. Physical examination revealed a 0.8×0.8×0.8-cm, extremely tender, firm, pink papule on the right preauricular cheek. A horizontal deep shave excision was done and the histopathology was remarkable for neoplastic cells with necrosis in the dermis. We observed dermal cellular infiltrates in the form of sheets and nodules, some showing central necrosis (Figure 1). At higher magnification, a trabecular arrangement of cells was seen. These cells had a moderate amount of cytoplasm with eccentric nuclei and rare nucleoli (Figure 2). Mitotic figures were seen at higher magnification (Figure 3). Immunohistochemistry of the neoplastic cells exhibited similar positive staining for the neuroendocrine markers chromogranin A and synaptophysin (Figure 4). Staining of the neoplastic cells also was positive for thyroid transcription factor 1 (TTF-1) and cancer antigen 19-9. Villin and caudal type homeobox 2 stains were negative. These results were consistent with cutaneous metastasis from a known pulmonary carcinoid tumor.

Figure 1. Cutaneous metastasis of a pulmonary carcinoid tumor. A dermal cellular infiltrate of sheets and nodules, some showing central necrosis (H&E, original magnification ×40).

Figure 2. Cutaneous metastasis of a pulmonary carcinoid tumor. A trabecular arrangement of cells with a moderate amount of cytoplasm with eccentric nuclei and rare nucleoli (H&E, original magnification ×400).

Figure 3. Cutaneous metastasis of a pulmonary carcinoid tumor. Mitotic figures can be seen (H&E, original magnification ×400).

Figure 4. Cutaneous metastasis of a pulmonary carcinoid tumor. Immunohistochemistry was positive for chromogranin A (original magnification ×400).

On further review of the patient’s medical history, it was discovered that he had undergone a Whipple procedure with adjuvant chemotherapy and radiation for pancreatic adenocarcinoma approximately 4 years prior to the current presentation. He was then followed by oncology, and 3 years later a chest computed tomography suggested possible disease progression with a new pulmonary metastasis. This pulmonary lesion was biopsied and immunologic staining was consistent with a primary neuroendocrine neoplasm of the lung, a new carcinoid tumor. The tissue was positive for cytokeratin (CK) 7,TTF-1, cancer antigen 19-9, CD56, synaptophysin, and chromogranin A, and was negative for villin and CK20. By the time he was seen in our clinic, several trials of chemotherapy had failed. Serial computed tomography subsequently demonstrated progression of the lung disease and he later developed malignant pleural effusions. Approximately 6 months after the cutaneous carcinoid metastasis was diagnosed, the patient died of respiratory failure.

 

 

Comment

Carcinoid tumors are uncommon neoplasms of neuroendocrine origin that generally arise in the gastrointestinal or bronchopulmonary tracts. Metastases from these primary neoplasms more commonly affect the regional lymph nodes or viscera, with rare reports of cutaneous metastases to the skin. The true incidence of carcinoid tumors with metastasis to the skin is unknown because it is limited to single case reports in the literature.

The clinical presentation of cutaneous carcinoid metastases has been reported most commonly as firm papules of varying sizes with no specific site predilection.1 The color of these lesions has ranged from erythematous to violaceous to brown.2 Several of the reported cases were noted to be extremely tender and painful, while other reports of lesions were noted to be asymptomatic or only mildly pruritic.3-7

Carcinoid syndrome is more common with neoplasms present within the gastrointestinal tract, but it also has been reported with large bronchial carcinoid tumors and with metastatic disease.8,9 Paroxysmal flushing is the most prominent cutaneous manifestation of this syndrome, occurring in 75% of patients.10,11 Other common symptoms include patchy cyanosis, telangiectasia, and pellagralike skin lesions.3 Carcinoid syndrome secondary to bronchial adenomas is thought to differ from gastrointestinal carcinoid neoplasms in that it has prolonged flushing (hours to days instead of minutes) and is characterized by marked anxiety, fever, disorientation, sweating, and lacrimation.8,9

Many cases of cutaneous carcinoid metastases have been accompanied by reports of exquisite tenderness,7 similar to our patient. The pathogenesis of the pain in these lesions is still unclear, but several hypotheses have been established. It has been postulated that perineural invasion by the tumor is responsible for the pain; however, this finding has been inconsistent, as neural involvement also has been present in nonpainful lesions.2,5,7,12 Another theory for the pain is that it is secondary to the release of vasoactive substances and peptide hormones from the carcinoid cells, such as kallikrein and serotonin. Lastly, local tissue necrosis and fibrosis also have been suggested as possible etiologies.7

The histology of cutaneous carcinoid metastases typically resembles the primary lesion and may demonstrate fascicles of spindle cells with focal areas of necrosis, mild atypia, and a relatively low mitotic rate.10 Other neoplasms such as Merkel cell carcinoma and carcinoidlike sebaceous carcinoma should be considered in the differential diagnosis. A primary malignant peripheral primitive neuroectodermal tumor or a primary cutaneous carcinoid tumor is less common but should be considered. Differing from carcinoid tumors, Merkel cell carcinomas usually have a higher mitotic rate and positive staining for CK20. The sebaceous neoplasms with a carcinoidlike pattern may appear histologically similar, requiring immunohistochemical evaluation with monoclonal antibodies such as D2-40.13 A diffuse granular cytoplasmic reaction to chromogranin A is characteristic of carcinoid tumors. Synaptophysin and TTF-1 also are positive in carcinoid tumors, with TTF-1 being highly specific for neuroendocrine tumors of the lung.10

Cutaneous metastases of internal malignancies are more common from carcinomas of the lungs, gastrointestinal tract, and breasts.5 Occasionally, the cutaneous metastasis will develop directly over the underlying malignancy. Our case of cutaneous metastasis of a carcinoid tumor presented as an exquisitely tender and painful papule on the cheek. The histology of the lesion was consistent with the known carcinoid tumor of the lung. Because these lesions are extremely uncommon, it is imperative to obtain an accurate clinical history and use the appropriate immunohistochemical panel to correctly diagnose these metastases.

References
  1. Blochin E, Stein JA, Wang NS. Atypical carcinoid metastasis to the skin. Am J Dermatopathol. 2010;32:735-739.
  2. Rodriguez G, Villamizar R. Carcinoid tumor with skin metastasis. Am J Dermatopathol. 1992;14:263-269.
  3. Archer CB, Rauch HJ, Allen MH, et al. Ultrastructural features of metastatic cutaneous carcinoid. J Cutan Pathol. 1984;11:485-490.
  4. Archer CB, Wells RS, MacDonald DM. Metastatic cutaneous carcinoid. J Am Acad Dermatol. 1985;13(2, pt 2):363-366.
  5. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis:a meta-analysis of data. South Med J. 2003;96:164-167.
  6. Oleksowicz L, Morris JC, Phelps RG, et al. Pulmonary carcinoid presenting as multiple subcutaneous nodules. Tumori. 1990;76:44-47.
  7. Zuetenhorst JM, van Velthuysen ML, Rutgers EJ, et al. Pathogenesis and treatment of pain caused by skin metastases in neuroendocrine tumours. Neth J Med. 2002;60:207-211.
  8. Melmon KL. Kinins: one of the many mediators of the carcinoid spectrum. Gastroenterology. 1968;55:545-548.
  9. Zuetenhorst JM, Taal BG. Metastatic carcinoid tumors: a clinical review. Oncologist. 2005;10:123-131.
  10. Sabir S, James WD, Schuchter LM. Cutaneous manifestations of cancer. Curr Opin Oncol. 1999;11:139-144.
  11. Braverman IM. Skin manifestations of internal malignancy. Clin Geriatr Med. 2002;18:1-19.
  12. Santi R, Massi D, Mazzoni F, et al. Skin metastasis from typical carcinoid tumor of the lung. J Cutan Pathol. 2008;35:418-422.
  13. Kazakov DV, Kutzner H, Rütten A, et al. Carcinoid-like pattern in sebaceous neoplasms. another distinctive, previously unrecognized pattern in extraocular sebaceous carcinoma and sebaceoma. Am J Dermatopathol. 2005;27:195-203.
References
  1. Blochin E, Stein JA, Wang NS. Atypical carcinoid metastasis to the skin. Am J Dermatopathol. 2010;32:735-739.
  2. Rodriguez G, Villamizar R. Carcinoid tumor with skin metastasis. Am J Dermatopathol. 1992;14:263-269.
  3. Archer CB, Rauch HJ, Allen MH, et al. Ultrastructural features of metastatic cutaneous carcinoid. J Cutan Pathol. 1984;11:485-490.
  4. Archer CB, Wells RS, MacDonald DM. Metastatic cutaneous carcinoid. J Am Acad Dermatol. 1985;13(2, pt 2):363-366.
  5. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis:a meta-analysis of data. South Med J. 2003;96:164-167.
  6. Oleksowicz L, Morris JC, Phelps RG, et al. Pulmonary carcinoid presenting as multiple subcutaneous nodules. Tumori. 1990;76:44-47.
  7. Zuetenhorst JM, van Velthuysen ML, Rutgers EJ, et al. Pathogenesis and treatment of pain caused by skin metastases in neuroendocrine tumours. Neth J Med. 2002;60:207-211.
  8. Melmon KL. Kinins: one of the many mediators of the carcinoid spectrum. Gastroenterology. 1968;55:545-548.
  9. Zuetenhorst JM, Taal BG. Metastatic carcinoid tumors: a clinical review. Oncologist. 2005;10:123-131.
  10. Sabir S, James WD, Schuchter LM. Cutaneous manifestations of cancer. Curr Opin Oncol. 1999;11:139-144.
  11. Braverman IM. Skin manifestations of internal malignancy. Clin Geriatr Med. 2002;18:1-19.
  12. Santi R, Massi D, Mazzoni F, et al. Skin metastasis from typical carcinoid tumor of the lung. J Cutan Pathol. 2008;35:418-422.
  13. Kazakov DV, Kutzner H, Rütten A, et al. Carcinoid-like pattern in sebaceous neoplasms. another distinctive, previously unrecognized pattern in extraocular sebaceous carcinoma and sebaceoma. Am J Dermatopathol. 2005;27:195-203.
Issue
Cutis - 99(5)
Issue
Cutis - 99(5)
Page Number
E13-E15
Page Number
E13-E15
Publications
Publications
Topics
Article Type
Display Headline
Cutaneous Metastasis of a Pulmonary Carcinoid Tumor
Display Headline
Cutaneous Metastasis of a Pulmonary Carcinoid Tumor
Sections
Inside the Article

Practice Points

  • Cutaneous metastases of carcinoid tumors are extremely rare, and clinical presentation can vary. They can present as firm papules ranging in color from pink to brown, can be painful, and could occur at any site.
  • It is imperative to obtain an accurate clinical history and use the appropriate immunohistochemical panel to correctly diagnose cutaneous metastases of carcinoid tumors.
  • Neoplasms within the gastrointestinal tract commonly present with carcinoid syndrome, but it also has been observed with bronchial carcinoid tumors and with metastatic disease.
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Are E-cigarettes effective for smoking cessation?

Article Type
Changed
Fri, 09/28/2018 - 10:58
Display Headline
Are E-cigarettes effective for smoking cessation?

Author and Disclosure Information

Natassia V. Gaznick, MD, PhD,
Resident Physician
Department of Psychiatry
University of California, San Diego, Health Sciences
San Diego, California

Publications
Topics
Sections
Author and Disclosure Information

Natassia V. Gaznick, MD, PhD,
Resident Physician
Department of Psychiatry
University of California, San Diego, Health Sciences
San Diego, California

Author and Disclosure Information

Natassia V. Gaznick, MD, PhD,
Resident Physician
Department of Psychiatry
University of California, San Diego, Health Sciences
San Diego, California

Publications
Publications
Topics
Article Type
Display Headline
Are E-cigarettes effective for smoking cessation?
Display Headline
Are E-cigarettes effective for smoking cessation?
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Use ProPublica

Primary Total Knee Arthroplasty for Distal Femur Fractures: A Systematic Review of Indications, Implants, Techniques, and Results

Article Type
Changed
Thu, 09/19/2019 - 13:21
Display Headline
Primary Total Knee Arthroplasty for Distal Femur Fractures: A Systematic Review of Indications, Implants, Techniques, and Results

Take-Home Points

  • Arthroplasty is a rarely utilized and, therefore, a rarely reported treatment for distal femur fractures.
  • Arthroplasty carries certain advantages over fixation, including earlier weight-bearing, a benefit for elderly individuals.
  • Arthroplasty is more often described in situations of comminution, often necessitating constrained prostheses.
  • It is not unreasonable to utilize arthroplasty in extra-articular fractures in poor-quality bone, which can take the form of unconstrained prosthesis and supplemental fixation.
  • The true complication rate is unclear, given that the few papers reporting high complication rates were in sicker populations.

Distal femur fractures (DFFs) in the elderly historically were difficult to treat because of osteoporotic bone, comminution, and intra-articular involvement. DFFs in minimally ambulatory patients were once treated nonoperatively, with traction or immobilization,1,2 but surgery is now considered for displaced and unstable fractures, even in myelopathic and nonambulatory patients, to provide pain relief, ease mobility, and decrease the risks associated with prolonged bed rest.1 Options are constantly evolving, but poor knee function, malunion, nonunion, prolonged immobilization, implant failure, and high morbidity and mortality rates have been reported in several studies regardless of fixation method.

Arthritis after DFF has been reported at rates of 36% to 50% by long-term follow-up.3-5 However, total knee arthroplasty (TKA) for posttraumatic arthritis is more complex because of scarring, arthrofibrosis, malunion, nonunion, and the frequent need for hardware removal. These cases have a higher incidence of infection, aseptic loosening, stiffness,6 and skin necrosis.Primary TKA is a rarely used treatment for acute DFF. Several authors have recommended primary TKA for patients with intra-articular DFFs and preexisting osteoarthritis or rheumatoid arthritis, severe comminution, or poor bone stock.7-22 Compared with open reduction and internal fixation (ORIF), primary TKA may allow for earlier mobility and weight-bearing and thereby reduce the rates of complications (eg, respiratory failure, deep vein thrombosis, pulmonary embolism) associated with prolonged immobilization.23As the literature on TKA for acute DFF is scant, and to our knowledge there are no clear indications or guidelines, we performed a systematic review to determine whether TKA has been successful in relieving pain and restoring knee function. In this article, we discuss the indications, implant options, technical considerations, complications, and results (eg, range of motion [ROM], ambulatory status) associated with these procedures.

Methods

On December 1, 2015, we searched the major databases Medline, EMBASE (Excerpta Medica dataBASE), and the Cochrane Library for articles published since 1950. In our searches, we used the conjoint term knee arthroplasty with femur fracture, and knee replacement with femur fracture. Specifically, we queried: ((“knee replacement” OR “knee arthroplasty”) AND (intercondylar OR supracondylar OR femoral OR femur) AND fracture) NOT arthrodesis NOT periprosthetic NOT “posttraumatic arthritis” NOT osteotomy. We also hand-searched the current website of JBJS [Journal of Bone and Joint Surgery] Case Connector, a major case-report repository that was launched in 2011 but is not currently indexed by Medline.

All citations were imported to RefWorks for management and for removal of duplicates. Each article underwent screening and review by Dr. Chen and Dr. Li. Articles were included if titles were relevant to arthroplasty as treatment for acute (within 1 month) DFF. Articles and cases were excluded if they were reviews, published in languages other than English, animal studies, studies regarding nonacute (>3 months or nonunion) DFFs or periprosthetic fractures, or studies that considered only treatments other than TKA (ie, plate osteosynthesis).

Full-text publications were obtained and independently reviewed by Dr. Chen and Dr. Li for relevance and satisfaction of inclusion criteria. Disagreements were resolved by discussion. Given the rarity of publications on the treatment, all study designs from level I to level IV were included.

The same 2 reviewers extracted the data into prearranged summary tables. Data included study size, patient demographics, AO/OTA (Arbeitsgemeinschaft für Osteosynthesefragen/Orthopaedic Trauma Association) fracture type either reported or assessed by description and imaging (33A, extra-articular; 33B, partial articular with 1 intact condyle; 33C, complete articular with both condyles involved), baseline comorbidity, implant used and fracture treatment (if separate from arthroplasty), postoperative regimen, respective outcomes, and complication rates.

Results

We identified 728 articles: 389 through Medline, 294 through EMBASE, and 45 through the Cochrane Library (Figure 1).

Figure 1.
After duplicates were removed, 476 articles remained. After titles and abstracts were reviewed, 22 articles met the screening criteria. Five series included patients with TKA-treated acute DFF but did not report their specific outcomes (these were described separately).
Table 1.

The current evidence regarding primary TKA for acute DFF is primarily level IV (Table 1). Only 1 level III study16 compared TKA with ORIF. Three case series11,19,24 met our inclusion criteria (Table 1, Table 2). In addition, 5 case series involved patients who met our criteria, but these studies did not separately report results for DFFs and proximal tibia fractures,9,20-22 or separately for acute fractures and nonunions or ORIF failures.8

Table 2.
These studies were considered level IV and were tabulated separately (Table 3). Specific patient characteristics and management strategies varied significantly between studies, though many studies augmented 33A fractures with internal fixation, whereas 33C fractures more often underwent resection and placement of highly constrained implants. Of 117 acute DFFs reviewed, 20% were 33A fractures, 7% were 33B fractures, and 73% were 33C fractures (Table 1). Of the studies that specified, there were 8 cases of rheumatoid arthritis and 18 cases of osteoarthritis (Table 2).
Table 3.


Modular, hinged, and tumor-type arthroplasty designs accounted for 83% of the treatments included in this review. Trade names are listed in Table 4. Authors who used these implants took a more aggressive approach, often resecting the entire femoral epiphyseal-metaphyseal area, menisci, and collateral ligaments.9,13,15,16,18 The majority of patients who underwent resection had 33C fractures (Tables 1, 3).
Table 4.
Figures 2A-2D show an aggressive resection example.Authors who used less constrained arthroplasty designs focused on bone preservation, augmentation with graft, and internal fixation.7,20 In and colleagues25 thought that if the cruciate and collateral ligaments are found to be intact, then resecting these ligaments and performing the deep cuts necessary for linked prostheses are too aggressive. Their internal fixation methods included use of cannulated screws, Dall-Miles cabling (Stryker), and plate osteosynthesis. Choi and colleagues19 took a similar approach but also used stem extensions in 6 of 8 fractures assessed to be unstable (Figures 3A-3H).
Figure 2.
Yoshino and colleagues7 used posterior-stabilized implants with femoral stem extensions (Figures 4A-4C).
Figure 3.
Intraoperative use of an external fixator to align and stabilize a comminuted fracture before insertion of an intramedullary guide and during femoral cutting has also been described.19 All 33B and many 33A fractures were treated in this fashion.

The majority of authors who treated fractures with resection and modular implants allowed their patients full weight-bearing soon after surgery (Table 1),11,12,15-18,24 whereas authors who treated their patients partly with fracture fixation often had to delay weight-bearing (Table 1).
Figure 4.
Overall, results were encouraging, with most studies finding between 90° and 135° of flexion to near full extension after each type of treatment. At follow-up, most survivors achieved full weight-bearing and were capable of walking up and down stairs.

Cement use was universally described in the literature. Some authors avoided placing cement in the fracture site (to reduce the risk of nonunion),7,19 whereas others used bone cement to fill metaphyseal defects that remained after fracture resection and implantation.11,24Complication rates were modest, and there were no reports specifically on implant loosening or fracture nonunion.7,10,12-19 The majority of complications were recorded in 2 studies that used megaprostheses in sicker populations: Bell and colleagues11 noted debilitating illnesses in all their patients, and Appleton and colleagues24 included 9 nonambulatory patients and 36 patients who required 2 assistants to ambulate. All deaths were attributed to medical comorbidities and disseminated malignancy. Contrarily, studies by Pearse and colleagues16 and Choi and colleagues19 included previously ambulatory patients and reported no deaths or complications (Table 2). Likewise, in studies that combined results of DFFs and proximal tibia fractures, death and complication rates varied from 7% to 31% (Table 3).

 

 

Discussion

DFFs in the elderly historically were difficult to treat. Reported outcomes are largely favorable, but, even with newer plate designs, catastrophic failures still occur in the absence of bony union.26,27 After ORIF, patients’ weight-bearing is often restricted for 12 weeks or longer28—a protocol that is undesirable in elderly patients, especially given that the rate of mortality 1 year after these fractures has been found to be as high as 25%.29

Arthroplasty for DFFs—performed either with ORIF, or independently with a constrained implant—is a documented treatment modality, but the evidence is poor, and results have been mixed. Patients who received hinged TKA with major fracture resection had higher complication rates.8,11,22,24 However, the problems were mostly medical, not associated with surgical technique. Appleton and colleagues24 found a higher than expected 1-year mortality rate, 41%, but used an unhealthy baseline population (44% cognitive impairment, 17% nonambulatory before injury).Although Boureau and colleagues22 found a 1-year mortality rate of 30%, only 1 in 10 deaths was attributable to a perioperative complication. Among the remaining cases involving resection and megaprostheses for previously ambulatory patients, only 1 perioperative death was recorded (Table 2).11,12,16,18 Therefore, the risks associated with patients’ baseline health and ambulatory status must be weighed against the benefits of aggressive arthroplasty.

An overwhelming majority of 33C fractures were treated with megaprostheses—a finding perhaps attributable to the higher likelihood that patients with osteoporosis have intra-articular, comminuted injuries. In addition, surgeons may have been more likely to indicate 33C fractures for joint replacement, whereas 33A and 33B patterns were more amenable to fracture fixation.17,18 Interestingly, few type B fractures (0 in primary analysis and only 9 of 67 cases in Table 3) were treated with megaprostheses. In these situations, 1 condyle and ligamentous constraint remain intact, reducing the need for a constrained implant.

There were no reports of atraumatic or aseptic loosening, though use of rotating platforms with linked prostheses helps minimize this complication. Also surprising is the lack of nonunions in any of the reviewed studies, as nonunion is one of the most devastating complications of ORIF. Only 1 superficial and 2 deep infections were reported in all of the literature—representing 1.8% of all cases, which is comparable to the rate for elective primary TKA.30In elderly patients with significant comorbidities, the main surgical goals are to minimize operative time and reduce time to mobility. It is therefore imperative to keep in mind that arthroplasty is elective. However, functional results of primary TKA for DFF may be more encouraging for healthier patients, as many can achieve satisfactory ROM and early weight-bearing. Therefore, TKA for DFF may benefit healthy and ambulatory patients in the setting of intra-articular comminution. Whether this treatment affects mortality rates remains to be seen.

There were several limitations to this study. First, the literature on the topic is scant. Second, exclusion criteria were kept lax to allow for inclusion of all treatments. This came at a cost to internal validity, given the heterogeneous population and differences in comorbidities between studies. Fracture classification was inconsistent as well: Although AO/OTA classification was dominant, descriptive classifications were used in several cases7,10,12 (these descriptions, however, were sufficient for assigning equivalent AO/OTA classes). Details on preoperative functional status and comorbidity status and on postoperative protocols were also limited, though ROM and ambulatory status were provided in most studies. Last, most of these studies were single case reports or case series, so there may be reporting bias in the body of the literature, as reflected in the discrepancies between encouraging case reports and concerning case series with longer follow-up. Such bias can be avoided with larger, controlled sampling and adequate follow-up.

TKA should be considered for acute DFF in patients who have knee arthritis and are able to tolerate the physiological load of the surgery. In the choice of implant design, several factors should be considered, including bone quality, articular involvement, degree of comminution, and ligamentous injury. Unconstrained knee designs should be considered in cases in which the fracture pattern appears stable and the collateral ligaments are intact (eg, 33A and 33BB fractures). Megaprostheses, which may allow for immediate weight-bearing but require considerable bone resection, would be beneficial in 33C fractures and in fractures with ligamentous compromise. However, their complication rates are unclear, and comparative studies are needed to investigate whether the rates are higher for these patients than for patients treated more traditionally.

Am J Orthop. 2017;46(3):E163-E171. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

 

 

References

1. Cass J, Sems SA. Operative versus nonoperative management of distal femur fracture in myelopathic, nonambulatory patients. Orthopedics. 2008;31(11):1091.

2. Eichenholtz SN. Management of long-bone fracture in paraplegic patients. J Bone Joint Surg Am. 1963;45(2):299-310.

3. Thomson AB, Driver R, Kregor PJ, Obremskey WT. Long-term functional outcomes after intra-articular distal femur fractures: ORIF versus retrograde intramedullary nailing. Orthopedics. 2008;31(8):748-750.

4. Rademakers MV, Kerkhoffs GM, Sierevelt IN, Raaymakers EL, Marti RK. Intra-articular fractures of the distal femur: a long-term follow-up study of surgically treated patients. J Orthop Trauma. 2004;18(4):213-219.

5. Schenker ML, Mauck RL, Ahn J, Mehta S. Pathogenesis and prevention of posttraumatic osteoarthritis after intra-articular fracture. J Am Acad Orthop Surg. 2014;22(1):20-28.

6. Papadopoulos EC, Parvizi J, Lai CH, Lewallen DG. Total knee arthroplasty following prior distal femoral fracture. Knee. 2002;9(4):267-274.

7. Yoshino N, Takai S, Watanabe Y, Fujiwara H, Ohshima Y, Hirasawa Y. Primary total knee arthroplasty for supracondylar/condylar femoral fracture in osteoarthritic knees. J Arthroplasty. 2001;16(4):471-475.

8. Rosen AL, Strauss E. Primary total knee arthroplasty for complex distal femur fractures in elderly patients. Clin Orthop Relat Res. 2004;(425):101-105.

9. Malviya A, Reed MR, Partington PF. Acute primary total knee arthroplasty for peri-articular knee fractures in patients over 65 years of age. Injury. 2011;42(11):1368-1371.

10. Wolfgang GL. Primary total knee arthroplasty for intercondylar fracture of the femur in a rheumatoid arthritic patient. A case report. Clin Orthop Relat Res. 1982;(171):80-82.

11. Bell KM, Johnstone AJ, Court-Brown CM, Hughes SP. Primary knee arthroplasty for distal femoral fractures in elderly patients. J Bone Joint Surg Br. 1992;74(3):400-402.

12. Shah A, Asirvatham R, Sudlow RA. Primary resection total knee arthroplasty for complicated fracture of the distal femur with an arthritic knee joint. Contemp Orthop. 1993;26(5):463-467.

13. Freedman EL, Hak DJ, Johnson EE, Eckardt JJ. Total knee replacement including a modular distal femoral component in elderly patients with acute fracture or nonunion. J Orthop Trauma. 1995;9(3):231-237.

14. Patterson RH, Earll M. Repair of supracondylar femur fracture and unilateral knee replacement at the same surgery. J Orthop Trauma. 1999;13(5):388-390.

15. Nau T, Pflegerl E, Erhart J, Vecsei V. Primary total knee arthroplasty for periarticular fractures. J Arthroplasty. 2003;18(8):968-971.

16. Pearse EO, Klass B, Bendall SP, Railton GT. Stanmore total knee replacement versus internal fixation for supracondylar fractures of the distal femur in elderly patients. Injury. 2005;36(1):163-168.

17. Mounasamy V, Ma SY, Schoderbek RJ, Mihalko WM, Saleh KJ, Brown TE. Primary total knee arthroplasty with condylar allograft and MCL reconstruction for a comminuted medial condyle fracture in an arthritic knee—a case report. Knee. 2006;13(5):400-403.

18. Mounasamy V, Cui Q, Brown TE, Saleh KJ, Mihalko WM. Primary total knee arthroplasty for a complex distal femur fracture in the elderly: a case report. Eur J Orthop Surg Traumatol. 2007;17(5):491-494.

19. Choi NY, Sohn JM, Cho SG, Kim SC, In Y. Primary total knee arthroplasty for simple distal femoral fractures in elderly patients with knee osteoarthritis. Knee Surg Relat Res. 2013;25(3):141-146.

20. Parratte S, Bonnevialle P, Pietu G, Saragaglia D, Cherrier B, Lafosse JM. Primary total knee arthroplasty in the management of epiphyseal fracture around the knee. Orthop Traumatol Surg Res. 2011;97(6 suppl):S87-S94.

21. Benazzo F, Rossi SM, Ghiara M, Zanardi A, Perticarini L, Combi A. Total knee replacement in acute and chronic traumatic events. Injury. 2014;45(suppl 6):S98-S104.

22. Boureau F, Benad K, Putman S, Dereudre G, Kern G, Chantelot C. Does primary total knee arthroplasty for acute knee joint fracture maintain autonomy in the elderly? A retrospective study of 21 cases. Orthop Traumatol Surg Res. 2015;101(8):947-951.

23. Bishop JA, Suarez P, Diponio L, Ota D, Curtin CM. Surgical versus nonsurgical treatment of femur fractures in people with spinal cord injury: an administrative analysis of risks. Arch Phys Med Rehabil. 2013;94(12):2357-2364.

24. Appleton P, Moran M, Houshian S, Robinson CM. Distal femoral fractures treated by hinged total knee replacement in elderly patients. J Bone Joint Surg Br. 2006;88(8):1065-1070.

25. In Y, Koh HS, Kim SJ. Cruciate-retaining stemmed total knee arthroplasty for supracondylar-intercondylar femoral fractures in elderly patients: a report of three cases. J Arthroplasty. 2006;21(7):1074-1079.

26. Kregor PJ, Stannard JA, Zlowodzki M, Cole PA. Treatment of distal femur fractures using the less invasive stabilization system: surgical experience and early clinical results in 103 fractures. J Orthop Trauma. 2004;18(8):509-520.

27. Vallier HA, Hennessey TA, Sontich JK, Patterson BM. Failure of LCP condylar plate fixation in the distal part of the femur. A report of six cases. J Bone Joint Surg Am. 2006;88(4):846-853.

28. Gwathmey FW Jr, Jones-Quaidoo SM, Kahler D, Hurwitz S, Cui Q. Distal femoral fractures: current concepts. J Am Acad Orthop Surg. 2010;18(10):597-607.

29. Streubel PN, Ricci WM, Wong A, Gardner MJ. Mortality after distal femur fractures in elderly patients. Clin Orthop Relat Res. 2011;469(4):1188-1196.

30. Peersman G, Laskin R, Davis J, Peterson M. Infection in total knee replacement: a retrospective review of 6489 total knee replacements. Clin Orthop Relat Res. 2001;(392):15-23.

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(3)
Publications
Topics
Page Number
E163-E171
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

  • Arthroplasty is a rarely utilized and, therefore, a rarely reported treatment for distal femur fractures.
  • Arthroplasty carries certain advantages over fixation, including earlier weight-bearing, a benefit for elderly individuals.
  • Arthroplasty is more often described in situations of comminution, often necessitating constrained prostheses.
  • It is not unreasonable to utilize arthroplasty in extra-articular fractures in poor-quality bone, which can take the form of unconstrained prosthesis and supplemental fixation.
  • The true complication rate is unclear, given that the few papers reporting high complication rates were in sicker populations.

Distal femur fractures (DFFs) in the elderly historically were difficult to treat because of osteoporotic bone, comminution, and intra-articular involvement. DFFs in minimally ambulatory patients were once treated nonoperatively, with traction or immobilization,1,2 but surgery is now considered for displaced and unstable fractures, even in myelopathic and nonambulatory patients, to provide pain relief, ease mobility, and decrease the risks associated with prolonged bed rest.1 Options are constantly evolving, but poor knee function, malunion, nonunion, prolonged immobilization, implant failure, and high morbidity and mortality rates have been reported in several studies regardless of fixation method.

Arthritis after DFF has been reported at rates of 36% to 50% by long-term follow-up.3-5 However, total knee arthroplasty (TKA) for posttraumatic arthritis is more complex because of scarring, arthrofibrosis, malunion, nonunion, and the frequent need for hardware removal. These cases have a higher incidence of infection, aseptic loosening, stiffness,6 and skin necrosis.Primary TKA is a rarely used treatment for acute DFF. Several authors have recommended primary TKA for patients with intra-articular DFFs and preexisting osteoarthritis or rheumatoid arthritis, severe comminution, or poor bone stock.7-22 Compared with open reduction and internal fixation (ORIF), primary TKA may allow for earlier mobility and weight-bearing and thereby reduce the rates of complications (eg, respiratory failure, deep vein thrombosis, pulmonary embolism) associated with prolonged immobilization.23As the literature on TKA for acute DFF is scant, and to our knowledge there are no clear indications or guidelines, we performed a systematic review to determine whether TKA has been successful in relieving pain and restoring knee function. In this article, we discuss the indications, implant options, technical considerations, complications, and results (eg, range of motion [ROM], ambulatory status) associated with these procedures.

Methods

On December 1, 2015, we searched the major databases Medline, EMBASE (Excerpta Medica dataBASE), and the Cochrane Library for articles published since 1950. In our searches, we used the conjoint term knee arthroplasty with femur fracture, and knee replacement with femur fracture. Specifically, we queried: ((“knee replacement” OR “knee arthroplasty”) AND (intercondylar OR supracondylar OR femoral OR femur) AND fracture) NOT arthrodesis NOT periprosthetic NOT “posttraumatic arthritis” NOT osteotomy. We also hand-searched the current website of JBJS [Journal of Bone and Joint Surgery] Case Connector, a major case-report repository that was launched in 2011 but is not currently indexed by Medline.

All citations were imported to RefWorks for management and for removal of duplicates. Each article underwent screening and review by Dr. Chen and Dr. Li. Articles were included if titles were relevant to arthroplasty as treatment for acute (within 1 month) DFF. Articles and cases were excluded if they were reviews, published in languages other than English, animal studies, studies regarding nonacute (>3 months or nonunion) DFFs or periprosthetic fractures, or studies that considered only treatments other than TKA (ie, plate osteosynthesis).

Full-text publications were obtained and independently reviewed by Dr. Chen and Dr. Li for relevance and satisfaction of inclusion criteria. Disagreements were resolved by discussion. Given the rarity of publications on the treatment, all study designs from level I to level IV were included.

The same 2 reviewers extracted the data into prearranged summary tables. Data included study size, patient demographics, AO/OTA (Arbeitsgemeinschaft für Osteosynthesefragen/Orthopaedic Trauma Association) fracture type either reported or assessed by description and imaging (33A, extra-articular; 33B, partial articular with 1 intact condyle; 33C, complete articular with both condyles involved), baseline comorbidity, implant used and fracture treatment (if separate from arthroplasty), postoperative regimen, respective outcomes, and complication rates.

Results

We identified 728 articles: 389 through Medline, 294 through EMBASE, and 45 through the Cochrane Library (Figure 1).

Figure 1.
After duplicates were removed, 476 articles remained. After titles and abstracts were reviewed, 22 articles met the screening criteria. Five series included patients with TKA-treated acute DFF but did not report their specific outcomes (these were described separately).
Table 1.

The current evidence regarding primary TKA for acute DFF is primarily level IV (Table 1). Only 1 level III study16 compared TKA with ORIF. Three case series11,19,24 met our inclusion criteria (Table 1, Table 2). In addition, 5 case series involved patients who met our criteria, but these studies did not separately report results for DFFs and proximal tibia fractures,9,20-22 or separately for acute fractures and nonunions or ORIF failures.8

Table 2.
These studies were considered level IV and were tabulated separately (Table 3). Specific patient characteristics and management strategies varied significantly between studies, though many studies augmented 33A fractures with internal fixation, whereas 33C fractures more often underwent resection and placement of highly constrained implants. Of 117 acute DFFs reviewed, 20% were 33A fractures, 7% were 33B fractures, and 73% were 33C fractures (Table 1). Of the studies that specified, there were 8 cases of rheumatoid arthritis and 18 cases of osteoarthritis (Table 2).
Table 3.


Modular, hinged, and tumor-type arthroplasty designs accounted for 83% of the treatments included in this review. Trade names are listed in Table 4. Authors who used these implants took a more aggressive approach, often resecting the entire femoral epiphyseal-metaphyseal area, menisci, and collateral ligaments.9,13,15,16,18 The majority of patients who underwent resection had 33C fractures (Tables 1, 3).
Table 4.
Figures 2A-2D show an aggressive resection example.Authors who used less constrained arthroplasty designs focused on bone preservation, augmentation with graft, and internal fixation.7,20 In and colleagues25 thought that if the cruciate and collateral ligaments are found to be intact, then resecting these ligaments and performing the deep cuts necessary for linked prostheses are too aggressive. Their internal fixation methods included use of cannulated screws, Dall-Miles cabling (Stryker), and plate osteosynthesis. Choi and colleagues19 took a similar approach but also used stem extensions in 6 of 8 fractures assessed to be unstable (Figures 3A-3H).
Figure 2.
Yoshino and colleagues7 used posterior-stabilized implants with femoral stem extensions (Figures 4A-4C).
Figure 3.
Intraoperative use of an external fixator to align and stabilize a comminuted fracture before insertion of an intramedullary guide and during femoral cutting has also been described.19 All 33B and many 33A fractures were treated in this fashion.

The majority of authors who treated fractures with resection and modular implants allowed their patients full weight-bearing soon after surgery (Table 1),11,12,15-18,24 whereas authors who treated their patients partly with fracture fixation often had to delay weight-bearing (Table 1).
Figure 4.
Overall, results were encouraging, with most studies finding between 90° and 135° of flexion to near full extension after each type of treatment. At follow-up, most survivors achieved full weight-bearing and were capable of walking up and down stairs.

Cement use was universally described in the literature. Some authors avoided placing cement in the fracture site (to reduce the risk of nonunion),7,19 whereas others used bone cement to fill metaphyseal defects that remained after fracture resection and implantation.11,24Complication rates were modest, and there were no reports specifically on implant loosening or fracture nonunion.7,10,12-19 The majority of complications were recorded in 2 studies that used megaprostheses in sicker populations: Bell and colleagues11 noted debilitating illnesses in all their patients, and Appleton and colleagues24 included 9 nonambulatory patients and 36 patients who required 2 assistants to ambulate. All deaths were attributed to medical comorbidities and disseminated malignancy. Contrarily, studies by Pearse and colleagues16 and Choi and colleagues19 included previously ambulatory patients and reported no deaths or complications (Table 2). Likewise, in studies that combined results of DFFs and proximal tibia fractures, death and complication rates varied from 7% to 31% (Table 3).

 

 

Discussion

DFFs in the elderly historically were difficult to treat. Reported outcomes are largely favorable, but, even with newer plate designs, catastrophic failures still occur in the absence of bony union.26,27 After ORIF, patients’ weight-bearing is often restricted for 12 weeks or longer28—a protocol that is undesirable in elderly patients, especially given that the rate of mortality 1 year after these fractures has been found to be as high as 25%.29

Arthroplasty for DFFs—performed either with ORIF, or independently with a constrained implant—is a documented treatment modality, but the evidence is poor, and results have been mixed. Patients who received hinged TKA with major fracture resection had higher complication rates.8,11,22,24 However, the problems were mostly medical, not associated with surgical technique. Appleton and colleagues24 found a higher than expected 1-year mortality rate, 41%, but used an unhealthy baseline population (44% cognitive impairment, 17% nonambulatory before injury).Although Boureau and colleagues22 found a 1-year mortality rate of 30%, only 1 in 10 deaths was attributable to a perioperative complication. Among the remaining cases involving resection and megaprostheses for previously ambulatory patients, only 1 perioperative death was recorded (Table 2).11,12,16,18 Therefore, the risks associated with patients’ baseline health and ambulatory status must be weighed against the benefits of aggressive arthroplasty.

An overwhelming majority of 33C fractures were treated with megaprostheses—a finding perhaps attributable to the higher likelihood that patients with osteoporosis have intra-articular, comminuted injuries. In addition, surgeons may have been more likely to indicate 33C fractures for joint replacement, whereas 33A and 33B patterns were more amenable to fracture fixation.17,18 Interestingly, few type B fractures (0 in primary analysis and only 9 of 67 cases in Table 3) were treated with megaprostheses. In these situations, 1 condyle and ligamentous constraint remain intact, reducing the need for a constrained implant.

There were no reports of atraumatic or aseptic loosening, though use of rotating platforms with linked prostheses helps minimize this complication. Also surprising is the lack of nonunions in any of the reviewed studies, as nonunion is one of the most devastating complications of ORIF. Only 1 superficial and 2 deep infections were reported in all of the literature—representing 1.8% of all cases, which is comparable to the rate for elective primary TKA.30In elderly patients with significant comorbidities, the main surgical goals are to minimize operative time and reduce time to mobility. It is therefore imperative to keep in mind that arthroplasty is elective. However, functional results of primary TKA for DFF may be more encouraging for healthier patients, as many can achieve satisfactory ROM and early weight-bearing. Therefore, TKA for DFF may benefit healthy and ambulatory patients in the setting of intra-articular comminution. Whether this treatment affects mortality rates remains to be seen.

There were several limitations to this study. First, the literature on the topic is scant. Second, exclusion criteria were kept lax to allow for inclusion of all treatments. This came at a cost to internal validity, given the heterogeneous population and differences in comorbidities between studies. Fracture classification was inconsistent as well: Although AO/OTA classification was dominant, descriptive classifications were used in several cases7,10,12 (these descriptions, however, were sufficient for assigning equivalent AO/OTA classes). Details on preoperative functional status and comorbidity status and on postoperative protocols were also limited, though ROM and ambulatory status were provided in most studies. Last, most of these studies were single case reports or case series, so there may be reporting bias in the body of the literature, as reflected in the discrepancies between encouraging case reports and concerning case series with longer follow-up. Such bias can be avoided with larger, controlled sampling and adequate follow-up.

TKA should be considered for acute DFF in patients who have knee arthritis and are able to tolerate the physiological load of the surgery. In the choice of implant design, several factors should be considered, including bone quality, articular involvement, degree of comminution, and ligamentous injury. Unconstrained knee designs should be considered in cases in which the fracture pattern appears stable and the collateral ligaments are intact (eg, 33A and 33BB fractures). Megaprostheses, which may allow for immediate weight-bearing but require considerable bone resection, would be beneficial in 33C fractures and in fractures with ligamentous compromise. However, their complication rates are unclear, and comparative studies are needed to investigate whether the rates are higher for these patients than for patients treated more traditionally.

Am J Orthop. 2017;46(3):E163-E171. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

 

 

Take-Home Points

  • Arthroplasty is a rarely utilized and, therefore, a rarely reported treatment for distal femur fractures.
  • Arthroplasty carries certain advantages over fixation, including earlier weight-bearing, a benefit for elderly individuals.
  • Arthroplasty is more often described in situations of comminution, often necessitating constrained prostheses.
  • It is not unreasonable to utilize arthroplasty in extra-articular fractures in poor-quality bone, which can take the form of unconstrained prosthesis and supplemental fixation.
  • The true complication rate is unclear, given that the few papers reporting high complication rates were in sicker populations.

Distal femur fractures (DFFs) in the elderly historically were difficult to treat because of osteoporotic bone, comminution, and intra-articular involvement. DFFs in minimally ambulatory patients were once treated nonoperatively, with traction or immobilization,1,2 but surgery is now considered for displaced and unstable fractures, even in myelopathic and nonambulatory patients, to provide pain relief, ease mobility, and decrease the risks associated with prolonged bed rest.1 Options are constantly evolving, but poor knee function, malunion, nonunion, prolonged immobilization, implant failure, and high morbidity and mortality rates have been reported in several studies regardless of fixation method.

Arthritis after DFF has been reported at rates of 36% to 50% by long-term follow-up.3-5 However, total knee arthroplasty (TKA) for posttraumatic arthritis is more complex because of scarring, arthrofibrosis, malunion, nonunion, and the frequent need for hardware removal. These cases have a higher incidence of infection, aseptic loosening, stiffness,6 and skin necrosis.Primary TKA is a rarely used treatment for acute DFF. Several authors have recommended primary TKA for patients with intra-articular DFFs and preexisting osteoarthritis or rheumatoid arthritis, severe comminution, or poor bone stock.7-22 Compared with open reduction and internal fixation (ORIF), primary TKA may allow for earlier mobility and weight-bearing and thereby reduce the rates of complications (eg, respiratory failure, deep vein thrombosis, pulmonary embolism) associated with prolonged immobilization.23As the literature on TKA for acute DFF is scant, and to our knowledge there are no clear indications or guidelines, we performed a systematic review to determine whether TKA has been successful in relieving pain and restoring knee function. In this article, we discuss the indications, implant options, technical considerations, complications, and results (eg, range of motion [ROM], ambulatory status) associated with these procedures.

Methods

On December 1, 2015, we searched the major databases Medline, EMBASE (Excerpta Medica dataBASE), and the Cochrane Library for articles published since 1950. In our searches, we used the conjoint term knee arthroplasty with femur fracture, and knee replacement with femur fracture. Specifically, we queried: ((“knee replacement” OR “knee arthroplasty”) AND (intercondylar OR supracondylar OR femoral OR femur) AND fracture) NOT arthrodesis NOT periprosthetic NOT “posttraumatic arthritis” NOT osteotomy. We also hand-searched the current website of JBJS [Journal of Bone and Joint Surgery] Case Connector, a major case-report repository that was launched in 2011 but is not currently indexed by Medline.

All citations were imported to RefWorks for management and for removal of duplicates. Each article underwent screening and review by Dr. Chen and Dr. Li. Articles were included if titles were relevant to arthroplasty as treatment for acute (within 1 month) DFF. Articles and cases were excluded if they were reviews, published in languages other than English, animal studies, studies regarding nonacute (>3 months or nonunion) DFFs or periprosthetic fractures, or studies that considered only treatments other than TKA (ie, plate osteosynthesis).

Full-text publications were obtained and independently reviewed by Dr. Chen and Dr. Li for relevance and satisfaction of inclusion criteria. Disagreements were resolved by discussion. Given the rarity of publications on the treatment, all study designs from level I to level IV were included.

The same 2 reviewers extracted the data into prearranged summary tables. Data included study size, patient demographics, AO/OTA (Arbeitsgemeinschaft für Osteosynthesefragen/Orthopaedic Trauma Association) fracture type either reported or assessed by description and imaging (33A, extra-articular; 33B, partial articular with 1 intact condyle; 33C, complete articular with both condyles involved), baseline comorbidity, implant used and fracture treatment (if separate from arthroplasty), postoperative regimen, respective outcomes, and complication rates.

Results

We identified 728 articles: 389 through Medline, 294 through EMBASE, and 45 through the Cochrane Library (Figure 1).

Figure 1.
After duplicates were removed, 476 articles remained. After titles and abstracts were reviewed, 22 articles met the screening criteria. Five series included patients with TKA-treated acute DFF but did not report their specific outcomes (these were described separately).
Table 1.

The current evidence regarding primary TKA for acute DFF is primarily level IV (Table 1). Only 1 level III study16 compared TKA with ORIF. Three case series11,19,24 met our inclusion criteria (Table 1, Table 2). In addition, 5 case series involved patients who met our criteria, but these studies did not separately report results for DFFs and proximal tibia fractures,9,20-22 or separately for acute fractures and nonunions or ORIF failures.8

Table 2.
These studies were considered level IV and were tabulated separately (Table 3). Specific patient characteristics and management strategies varied significantly between studies, though many studies augmented 33A fractures with internal fixation, whereas 33C fractures more often underwent resection and placement of highly constrained implants. Of 117 acute DFFs reviewed, 20% were 33A fractures, 7% were 33B fractures, and 73% were 33C fractures (Table 1). Of the studies that specified, there were 8 cases of rheumatoid arthritis and 18 cases of osteoarthritis (Table 2).
Table 3.


Modular, hinged, and tumor-type arthroplasty designs accounted for 83% of the treatments included in this review. Trade names are listed in Table 4. Authors who used these implants took a more aggressive approach, often resecting the entire femoral epiphyseal-metaphyseal area, menisci, and collateral ligaments.9,13,15,16,18 The majority of patients who underwent resection had 33C fractures (Tables 1, 3).
Table 4.
Figures 2A-2D show an aggressive resection example.Authors who used less constrained arthroplasty designs focused on bone preservation, augmentation with graft, and internal fixation.7,20 In and colleagues25 thought that if the cruciate and collateral ligaments are found to be intact, then resecting these ligaments and performing the deep cuts necessary for linked prostheses are too aggressive. Their internal fixation methods included use of cannulated screws, Dall-Miles cabling (Stryker), and plate osteosynthesis. Choi and colleagues19 took a similar approach but also used stem extensions in 6 of 8 fractures assessed to be unstable (Figures 3A-3H).
Figure 2.
Yoshino and colleagues7 used posterior-stabilized implants with femoral stem extensions (Figures 4A-4C).
Figure 3.
Intraoperative use of an external fixator to align and stabilize a comminuted fracture before insertion of an intramedullary guide and during femoral cutting has also been described.19 All 33B and many 33A fractures were treated in this fashion.

The majority of authors who treated fractures with resection and modular implants allowed their patients full weight-bearing soon after surgery (Table 1),11,12,15-18,24 whereas authors who treated their patients partly with fracture fixation often had to delay weight-bearing (Table 1).
Figure 4.
Overall, results were encouraging, with most studies finding between 90° and 135° of flexion to near full extension after each type of treatment. At follow-up, most survivors achieved full weight-bearing and were capable of walking up and down stairs.

Cement use was universally described in the literature. Some authors avoided placing cement in the fracture site (to reduce the risk of nonunion),7,19 whereas others used bone cement to fill metaphyseal defects that remained after fracture resection and implantation.11,24Complication rates were modest, and there were no reports specifically on implant loosening or fracture nonunion.7,10,12-19 The majority of complications were recorded in 2 studies that used megaprostheses in sicker populations: Bell and colleagues11 noted debilitating illnesses in all their patients, and Appleton and colleagues24 included 9 nonambulatory patients and 36 patients who required 2 assistants to ambulate. All deaths were attributed to medical comorbidities and disseminated malignancy. Contrarily, studies by Pearse and colleagues16 and Choi and colleagues19 included previously ambulatory patients and reported no deaths or complications (Table 2). Likewise, in studies that combined results of DFFs and proximal tibia fractures, death and complication rates varied from 7% to 31% (Table 3).

 

 

Discussion

DFFs in the elderly historically were difficult to treat. Reported outcomes are largely favorable, but, even with newer plate designs, catastrophic failures still occur in the absence of bony union.26,27 After ORIF, patients’ weight-bearing is often restricted for 12 weeks or longer28—a protocol that is undesirable in elderly patients, especially given that the rate of mortality 1 year after these fractures has been found to be as high as 25%.29

Arthroplasty for DFFs—performed either with ORIF, or independently with a constrained implant—is a documented treatment modality, but the evidence is poor, and results have been mixed. Patients who received hinged TKA with major fracture resection had higher complication rates.8,11,22,24 However, the problems were mostly medical, not associated with surgical technique. Appleton and colleagues24 found a higher than expected 1-year mortality rate, 41%, but used an unhealthy baseline population (44% cognitive impairment, 17% nonambulatory before injury).Although Boureau and colleagues22 found a 1-year mortality rate of 30%, only 1 in 10 deaths was attributable to a perioperative complication. Among the remaining cases involving resection and megaprostheses for previously ambulatory patients, only 1 perioperative death was recorded (Table 2).11,12,16,18 Therefore, the risks associated with patients’ baseline health and ambulatory status must be weighed against the benefits of aggressive arthroplasty.

An overwhelming majority of 33C fractures were treated with megaprostheses—a finding perhaps attributable to the higher likelihood that patients with osteoporosis have intra-articular, comminuted injuries. In addition, surgeons may have been more likely to indicate 33C fractures for joint replacement, whereas 33A and 33B patterns were more amenable to fracture fixation.17,18 Interestingly, few type B fractures (0 in primary analysis and only 9 of 67 cases in Table 3) were treated with megaprostheses. In these situations, 1 condyle and ligamentous constraint remain intact, reducing the need for a constrained implant.

There were no reports of atraumatic or aseptic loosening, though use of rotating platforms with linked prostheses helps minimize this complication. Also surprising is the lack of nonunions in any of the reviewed studies, as nonunion is one of the most devastating complications of ORIF. Only 1 superficial and 2 deep infections were reported in all of the literature—representing 1.8% of all cases, which is comparable to the rate for elective primary TKA.30In elderly patients with significant comorbidities, the main surgical goals are to minimize operative time and reduce time to mobility. It is therefore imperative to keep in mind that arthroplasty is elective. However, functional results of primary TKA for DFF may be more encouraging for healthier patients, as many can achieve satisfactory ROM and early weight-bearing. Therefore, TKA for DFF may benefit healthy and ambulatory patients in the setting of intra-articular comminution. Whether this treatment affects mortality rates remains to be seen.

There were several limitations to this study. First, the literature on the topic is scant. Second, exclusion criteria were kept lax to allow for inclusion of all treatments. This came at a cost to internal validity, given the heterogeneous population and differences in comorbidities between studies. Fracture classification was inconsistent as well: Although AO/OTA classification was dominant, descriptive classifications were used in several cases7,10,12 (these descriptions, however, were sufficient for assigning equivalent AO/OTA classes). Details on preoperative functional status and comorbidity status and on postoperative protocols were also limited, though ROM and ambulatory status were provided in most studies. Last, most of these studies were single case reports or case series, so there may be reporting bias in the body of the literature, as reflected in the discrepancies between encouraging case reports and concerning case series with longer follow-up. Such bias can be avoided with larger, controlled sampling and adequate follow-up.

TKA should be considered for acute DFF in patients who have knee arthritis and are able to tolerate the physiological load of the surgery. In the choice of implant design, several factors should be considered, including bone quality, articular involvement, degree of comminution, and ligamentous injury. Unconstrained knee designs should be considered in cases in which the fracture pattern appears stable and the collateral ligaments are intact (eg, 33A and 33BB fractures). Megaprostheses, which may allow for immediate weight-bearing but require considerable bone resection, would be beneficial in 33C fractures and in fractures with ligamentous compromise. However, their complication rates are unclear, and comparative studies are needed to investigate whether the rates are higher for these patients than for patients treated more traditionally.

Am J Orthop. 2017;46(3):E163-E171. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

 

 

References

1. Cass J, Sems SA. Operative versus nonoperative management of distal femur fracture in myelopathic, nonambulatory patients. Orthopedics. 2008;31(11):1091.

2. Eichenholtz SN. Management of long-bone fracture in paraplegic patients. J Bone Joint Surg Am. 1963;45(2):299-310.

3. Thomson AB, Driver R, Kregor PJ, Obremskey WT. Long-term functional outcomes after intra-articular distal femur fractures: ORIF versus retrograde intramedullary nailing. Orthopedics. 2008;31(8):748-750.

4. Rademakers MV, Kerkhoffs GM, Sierevelt IN, Raaymakers EL, Marti RK. Intra-articular fractures of the distal femur: a long-term follow-up study of surgically treated patients. J Orthop Trauma. 2004;18(4):213-219.

5. Schenker ML, Mauck RL, Ahn J, Mehta S. Pathogenesis and prevention of posttraumatic osteoarthritis after intra-articular fracture. J Am Acad Orthop Surg. 2014;22(1):20-28.

6. Papadopoulos EC, Parvizi J, Lai CH, Lewallen DG. Total knee arthroplasty following prior distal femoral fracture. Knee. 2002;9(4):267-274.

7. Yoshino N, Takai S, Watanabe Y, Fujiwara H, Ohshima Y, Hirasawa Y. Primary total knee arthroplasty for supracondylar/condylar femoral fracture in osteoarthritic knees. J Arthroplasty. 2001;16(4):471-475.

8. Rosen AL, Strauss E. Primary total knee arthroplasty for complex distal femur fractures in elderly patients. Clin Orthop Relat Res. 2004;(425):101-105.

9. Malviya A, Reed MR, Partington PF. Acute primary total knee arthroplasty for peri-articular knee fractures in patients over 65 years of age. Injury. 2011;42(11):1368-1371.

10. Wolfgang GL. Primary total knee arthroplasty for intercondylar fracture of the femur in a rheumatoid arthritic patient. A case report. Clin Orthop Relat Res. 1982;(171):80-82.

11. Bell KM, Johnstone AJ, Court-Brown CM, Hughes SP. Primary knee arthroplasty for distal femoral fractures in elderly patients. J Bone Joint Surg Br. 1992;74(3):400-402.

12. Shah A, Asirvatham R, Sudlow RA. Primary resection total knee arthroplasty for complicated fracture of the distal femur with an arthritic knee joint. Contemp Orthop. 1993;26(5):463-467.

13. Freedman EL, Hak DJ, Johnson EE, Eckardt JJ. Total knee replacement including a modular distal femoral component in elderly patients with acute fracture or nonunion. J Orthop Trauma. 1995;9(3):231-237.

14. Patterson RH, Earll M. Repair of supracondylar femur fracture and unilateral knee replacement at the same surgery. J Orthop Trauma. 1999;13(5):388-390.

15. Nau T, Pflegerl E, Erhart J, Vecsei V. Primary total knee arthroplasty for periarticular fractures. J Arthroplasty. 2003;18(8):968-971.

16. Pearse EO, Klass B, Bendall SP, Railton GT. Stanmore total knee replacement versus internal fixation for supracondylar fractures of the distal femur in elderly patients. Injury. 2005;36(1):163-168.

17. Mounasamy V, Ma SY, Schoderbek RJ, Mihalko WM, Saleh KJ, Brown TE. Primary total knee arthroplasty with condylar allograft and MCL reconstruction for a comminuted medial condyle fracture in an arthritic knee—a case report. Knee. 2006;13(5):400-403.

18. Mounasamy V, Cui Q, Brown TE, Saleh KJ, Mihalko WM. Primary total knee arthroplasty for a complex distal femur fracture in the elderly: a case report. Eur J Orthop Surg Traumatol. 2007;17(5):491-494.

19. Choi NY, Sohn JM, Cho SG, Kim SC, In Y. Primary total knee arthroplasty for simple distal femoral fractures in elderly patients with knee osteoarthritis. Knee Surg Relat Res. 2013;25(3):141-146.

20. Parratte S, Bonnevialle P, Pietu G, Saragaglia D, Cherrier B, Lafosse JM. Primary total knee arthroplasty in the management of epiphyseal fracture around the knee. Orthop Traumatol Surg Res. 2011;97(6 suppl):S87-S94.

21. Benazzo F, Rossi SM, Ghiara M, Zanardi A, Perticarini L, Combi A. Total knee replacement in acute and chronic traumatic events. Injury. 2014;45(suppl 6):S98-S104.

22. Boureau F, Benad K, Putman S, Dereudre G, Kern G, Chantelot C. Does primary total knee arthroplasty for acute knee joint fracture maintain autonomy in the elderly? A retrospective study of 21 cases. Orthop Traumatol Surg Res. 2015;101(8):947-951.

23. Bishop JA, Suarez P, Diponio L, Ota D, Curtin CM. Surgical versus nonsurgical treatment of femur fractures in people with spinal cord injury: an administrative analysis of risks. Arch Phys Med Rehabil. 2013;94(12):2357-2364.

24. Appleton P, Moran M, Houshian S, Robinson CM. Distal femoral fractures treated by hinged total knee replacement in elderly patients. J Bone Joint Surg Br. 2006;88(8):1065-1070.

25. In Y, Koh HS, Kim SJ. Cruciate-retaining stemmed total knee arthroplasty for supracondylar-intercondylar femoral fractures in elderly patients: a report of three cases. J Arthroplasty. 2006;21(7):1074-1079.

26. Kregor PJ, Stannard JA, Zlowodzki M, Cole PA. Treatment of distal femur fractures using the less invasive stabilization system: surgical experience and early clinical results in 103 fractures. J Orthop Trauma. 2004;18(8):509-520.

27. Vallier HA, Hennessey TA, Sontich JK, Patterson BM. Failure of LCP condylar plate fixation in the distal part of the femur. A report of six cases. J Bone Joint Surg Am. 2006;88(4):846-853.

28. Gwathmey FW Jr, Jones-Quaidoo SM, Kahler D, Hurwitz S, Cui Q. Distal femoral fractures: current concepts. J Am Acad Orthop Surg. 2010;18(10):597-607.

29. Streubel PN, Ricci WM, Wong A, Gardner MJ. Mortality after distal femur fractures in elderly patients. Clin Orthop Relat Res. 2011;469(4):1188-1196.

30. Peersman G, Laskin R, Davis J, Peterson M. Infection in total knee replacement: a retrospective review of 6489 total knee replacements. Clin Orthop Relat Res. 2001;(392):15-23.

References

1. Cass J, Sems SA. Operative versus nonoperative management of distal femur fracture in myelopathic, nonambulatory patients. Orthopedics. 2008;31(11):1091.

2. Eichenholtz SN. Management of long-bone fracture in paraplegic patients. J Bone Joint Surg Am. 1963;45(2):299-310.

3. Thomson AB, Driver R, Kregor PJ, Obremskey WT. Long-term functional outcomes after intra-articular distal femur fractures: ORIF versus retrograde intramedullary nailing. Orthopedics. 2008;31(8):748-750.

4. Rademakers MV, Kerkhoffs GM, Sierevelt IN, Raaymakers EL, Marti RK. Intra-articular fractures of the distal femur: a long-term follow-up study of surgically treated patients. J Orthop Trauma. 2004;18(4):213-219.

5. Schenker ML, Mauck RL, Ahn J, Mehta S. Pathogenesis and prevention of posttraumatic osteoarthritis after intra-articular fracture. J Am Acad Orthop Surg. 2014;22(1):20-28.

6. Papadopoulos EC, Parvizi J, Lai CH, Lewallen DG. Total knee arthroplasty following prior distal femoral fracture. Knee. 2002;9(4):267-274.

7. Yoshino N, Takai S, Watanabe Y, Fujiwara H, Ohshima Y, Hirasawa Y. Primary total knee arthroplasty for supracondylar/condylar femoral fracture in osteoarthritic knees. J Arthroplasty. 2001;16(4):471-475.

8. Rosen AL, Strauss E. Primary total knee arthroplasty for complex distal femur fractures in elderly patients. Clin Orthop Relat Res. 2004;(425):101-105.

9. Malviya A, Reed MR, Partington PF. Acute primary total knee arthroplasty for peri-articular knee fractures in patients over 65 years of age. Injury. 2011;42(11):1368-1371.

10. Wolfgang GL. Primary total knee arthroplasty for intercondylar fracture of the femur in a rheumatoid arthritic patient. A case report. Clin Orthop Relat Res. 1982;(171):80-82.

11. Bell KM, Johnstone AJ, Court-Brown CM, Hughes SP. Primary knee arthroplasty for distal femoral fractures in elderly patients. J Bone Joint Surg Br. 1992;74(3):400-402.

12. Shah A, Asirvatham R, Sudlow RA. Primary resection total knee arthroplasty for complicated fracture of the distal femur with an arthritic knee joint. Contemp Orthop. 1993;26(5):463-467.

13. Freedman EL, Hak DJ, Johnson EE, Eckardt JJ. Total knee replacement including a modular distal femoral component in elderly patients with acute fracture or nonunion. J Orthop Trauma. 1995;9(3):231-237.

14. Patterson RH, Earll M. Repair of supracondylar femur fracture and unilateral knee replacement at the same surgery. J Orthop Trauma. 1999;13(5):388-390.

15. Nau T, Pflegerl E, Erhart J, Vecsei V. Primary total knee arthroplasty for periarticular fractures. J Arthroplasty. 2003;18(8):968-971.

16. Pearse EO, Klass B, Bendall SP, Railton GT. Stanmore total knee replacement versus internal fixation for supracondylar fractures of the distal femur in elderly patients. Injury. 2005;36(1):163-168.

17. Mounasamy V, Ma SY, Schoderbek RJ, Mihalko WM, Saleh KJ, Brown TE. Primary total knee arthroplasty with condylar allograft and MCL reconstruction for a comminuted medial condyle fracture in an arthritic knee—a case report. Knee. 2006;13(5):400-403.

18. Mounasamy V, Cui Q, Brown TE, Saleh KJ, Mihalko WM. Primary total knee arthroplasty for a complex distal femur fracture in the elderly: a case report. Eur J Orthop Surg Traumatol. 2007;17(5):491-494.

19. Choi NY, Sohn JM, Cho SG, Kim SC, In Y. Primary total knee arthroplasty for simple distal femoral fractures in elderly patients with knee osteoarthritis. Knee Surg Relat Res. 2013;25(3):141-146.

20. Parratte S, Bonnevialle P, Pietu G, Saragaglia D, Cherrier B, Lafosse JM. Primary total knee arthroplasty in the management of epiphyseal fracture around the knee. Orthop Traumatol Surg Res. 2011;97(6 suppl):S87-S94.

21. Benazzo F, Rossi SM, Ghiara M, Zanardi A, Perticarini L, Combi A. Total knee replacement in acute and chronic traumatic events. Injury. 2014;45(suppl 6):S98-S104.

22. Boureau F, Benad K, Putman S, Dereudre G, Kern G, Chantelot C. Does primary total knee arthroplasty for acute knee joint fracture maintain autonomy in the elderly? A retrospective study of 21 cases. Orthop Traumatol Surg Res. 2015;101(8):947-951.

23. Bishop JA, Suarez P, Diponio L, Ota D, Curtin CM. Surgical versus nonsurgical treatment of femur fractures in people with spinal cord injury: an administrative analysis of risks. Arch Phys Med Rehabil. 2013;94(12):2357-2364.

24. Appleton P, Moran M, Houshian S, Robinson CM. Distal femoral fractures treated by hinged total knee replacement in elderly patients. J Bone Joint Surg Br. 2006;88(8):1065-1070.

25. In Y, Koh HS, Kim SJ. Cruciate-retaining stemmed total knee arthroplasty for supracondylar-intercondylar femoral fractures in elderly patients: a report of three cases. J Arthroplasty. 2006;21(7):1074-1079.

26. Kregor PJ, Stannard JA, Zlowodzki M, Cole PA. Treatment of distal femur fractures using the less invasive stabilization system: surgical experience and early clinical results in 103 fractures. J Orthop Trauma. 2004;18(8):509-520.

27. Vallier HA, Hennessey TA, Sontich JK, Patterson BM. Failure of LCP condylar plate fixation in the distal part of the femur. A report of six cases. J Bone Joint Surg Am. 2006;88(4):846-853.

28. Gwathmey FW Jr, Jones-Quaidoo SM, Kahler D, Hurwitz S, Cui Q. Distal femoral fractures: current concepts. J Am Acad Orthop Surg. 2010;18(10):597-607.

29. Streubel PN, Ricci WM, Wong A, Gardner MJ. Mortality after distal femur fractures in elderly patients. Clin Orthop Relat Res. 2011;469(4):1188-1196.

30. Peersman G, Laskin R, Davis J, Peterson M. Infection in total knee replacement: a retrospective review of 6489 total knee replacements. Clin Orthop Relat Res. 2001;(392):15-23.

Issue
The American Journal of Orthopedics - 46(3)
Issue
The American Journal of Orthopedics - 46(3)
Page Number
E163-E171
Page Number
E163-E171
Publications
Publications
Topics
Article Type
Display Headline
Primary Total Knee Arthroplasty for Distal Femur Fractures: A Systematic Review of Indications, Implants, Techniques, and Results
Display Headline
Primary Total Knee Arthroplasty for Distal Femur Fractures: A Systematic Review of Indications, Implants, Techniques, and Results
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Acute Intraprosthetic Dissociation of a Dual-Mobility Hip in the United States

Article Type
Changed
Thu, 09/19/2019 - 13:21
Display Headline
Acute Intraprosthetic Dissociation of a Dual-Mobility Hip in the United States

Take-Home Points

  • AIPD of DM-THA is defined by dissociation within 1 year of implantation resulting from component impingement or closed reduction maneuvers.
  • This is a distinct entity from “late” IPD (>1 year) from implantation as this is associated most often with polyethylene wear, component loosening, and arthrofibrosis.
  • A history of DM dislocation followed by subjective “clunking,” instability, and a series of more frequent dislocations should raise concern for AIPD.
  • Classic radiographic findings of AIPD include eccentric hip reduction and soft tissue radiolucency (ie, halo sign) from dissociated polyethylene component.
  • Treating practitioners of AIPD should consider closed reduction with general anesthesia and sedation in the operating room to limit risk of dissociation.

Dual-mobility (DM) components were invented in the 1970s and have been used in primary and revision total hip arthroplasty (THA) in Europe ever since.1 However, DM components are most commonly used in the treatment of recurrent hip instability, and early results have been promising.2 In DM-THAs, a smaller (22-mm or 28-mm) metal femoral head snap-fits into a larger polyethylene ball (inner articulation), which articulates with a highly polished metal shell (outer articulation), which is either implanted directly in the acetabulum or placed in an uncemented acetabular cup. The 2 articulations used in these devices theoretically increase hip range of motion (ROM) and increase the inferior head displacement distance (jump distance) required for dislocation.3

However, this DM articulation with increased ROM may also cause chronic impingement of the femoral component neck or Morse taper against the outer polyethylene bearing, resulting in polyethylene wear and late intraprosthetic dissociation (IPD) (separation of inner articulation between femoral head and polyethylene liner). In 2004, Lecuire and colleagues4 reported 7 cases of IPD occurring a mean of 10 years after implantation during the period 1989 to 1997. In 2013, Philippot and colleagues5 reported that 81 of 1960 primary THAs developed IPD a mean of 9 years after implantation. These IPD cases were attributed to polyethylene wear or outer articulation blockage caused by arthrofibrosis or heterotopic ossification. Reports of acute IPD (AIPD), however, are rare. In 2011, Stigbrand and Ullmark6 reported 3 cases in which the DM prosthesis dislocated within 1 year after implantation. It was suggested that the inner metal head dissociated from the larger polyethylene component after attempted closed reduction for dislocation (separation of larger polyethylene component from acetabulum or acetabular liner).

DM components were unavailable to surgeons in the United States until 2011. The first US Food and Drug Administration (FDA)-approved DM device was the MDM (Modular Dual Mobility, Stryker). To our knowledge, 2 cases of AIPD with this prosthesis have been reported.7, 8 As with the cases in Europe, closed reduction was the suspected cause, but there was no explanation for the initial dislocation event.

In this article, we present the case of a nondemented man who developed AIPD of a THA with the MDM component and a 28-mm femoral head with a skirted neck (StelKast). His operative findings suggest a poor head-to-neck ratio caused by a larger diameter femoral neck or a skirted prosthesis, or a forceful reduction maneuver, may predispose DM components to AIPD. The patient provided written informed consent for print and electronic publication of this case report.

Case Report

In 2012, a 63-year-old man with a history of drug abuse underwent left primary THA. Seven posterior dislocations and 3 years later, the acetabular component was revised to the MDM prosthesis; the well-fixed StelKast femoral component was retained (Figure 1).

Figure 1.

Within 3 months after revision surgery, the left hip dislocated 3 times in 1 week, when the patient bent over to retrieve an object on the ground. The first 2 dislocations were treated with closed reduction under conscious sedation at an outside emergency department.

Figure 2.
Shortly after, the patient, with complaints of left hip pain and clunking, was seen by a physician assistant, but the treating team did not notice the eccentric reduction on radiographs. The third dislocation was treated with closed reduction under conscious sedation in the emergency department at our institution (Figure 2). Postreduction radiographs still showed the eccentric reduction, and a radiolucent halo was visible superior to the greater trochanter (Figure 3).
Figure 3.


With the patient’s erythrocyte sedimentation rate and C-reactive protein level both normal, a second revision was performed. During surgery, the polyethylene head was found beneath the gluteus maximus (Figure 4).
Figure 4.
Gross inspection revealed a small amount of eccentric polyethylene wear and metal debris of the inner articulation (Figure 5). As the abductor muscles were intact, it was decided to proceed with revision to a larger DM component and to downsize the femoral head to a skirtless component (Table, Figure 6).

 

 

Discussion

Recurrent dislocation and instability accounts for 22.5% of THA revisions in the United States.9 Until 2011, options for managing recurrent dislocation in the United States included modular component exchange, component revision for malposition, and use of constrained components.10

Figure 5.
However, the decreased motion of constrained components may produce excess stress that eventually results in failure.11-13

In 1974, Bousquet first reported use of the DM prosthesis in primary THA; the prosthesis allowed increased stability without sacrificing motion or fixation.1 However, longer-term studies of DM components disclosed a new complication, IPD. In 2004, Lecuire and colleagues4 reported 7 cases of IPD occurring a mean of 10 years after implantation of the Bousquet prosthesis.

Table.
Philippot and colleagues5 reported that 81 of 1960 primary THAs with DM components developed IPD a mean of 9 years after implantation. They described 3 types of IPD based on mechanism of injury: type I, caused by wear of the inner articulation without arthrofibrosis or cup loosening (n = 26); type II, resulting from blocked outer articulation motion, caused by arthrofibrosis, nonunion, calcification, or heterotopic ossification (n = 41); and type III, associated with acetabular component loosening (n = 14). IPD occurred an average of 11 years (type I), 8 years (type II), and 9 years (type III) after implantation.

AIPD, which occurs within 1 year after implantation, has been reported much less often than late IPD. Stigbrand and Ullmark6 reported 3 cases of AIPD that developed within 7 months after implantation of Amplitude and Advantage (Zimmer Biomet) DM prostheses.
Figure 6.
The authors proposed that AIPD is related to incomplete coupling of the metal head and the inner polyethylene liner or to shearing of the large polyethylene component on the acetabular rim during a closed reduction maneuver. According to their description, the femoral head in the acetabulum had an “eccentric” radiographic appearance. The authors recommended administering muscle relaxants during closed reduction to avoid dissociation of the liner during the reduction.

This unusual complication apparently is not confined to a specific implant or region. Since the MDM component was introduced in the United States, 2 more cases of AIPD have been identified (Table). Banzhof and colleagues7 reported the case of a 68-year-old woman who, 2 months after the MDM was placed for recurrent instability, dislocated the component while rising from a seated position. Her IPD most likely resulted from a closed reduction. The affected hip eventually required closed reduction in the operating room. Postreduction radiographs showed the characteristic eccentric appearance; a halo, also visible in the soft tissues, corresponded with the dissociated radiolucent polyethylene liner. The authors attributed the early failure to an eccentrically seated metal liner that separated the locking mechanism. The MDM component was revised to a conventional THA, with the femoral head upsized and length added.

Ward and colleagues8 reported the case of an 87-year-old woman who had a conventional THA revised to an MDM component for recurrent instability. Two months after surgery, this patient, who had dementia, experienced 2 posterior dislocations while rising from a chair. Closed reduction in the emergency department seemed successful, but later she presented to the surgeon’s office with symptoms of instability and clunking, complaints similar to our patient’s. Radiographs showed an eccentric reduction caused by IPD, and the MDM component was revised to a constrained liner. Adding a MDM component to a retained DePuy (DePuy Synthes) femoral stem and head is considered “off-label use,” which, the authors proposed, may have been related to the AIPD in their patient’s case. However, one manufacturer’s femoral component and head are often mated with another manufacturer’s acetabular component to allow for a less complex revision. Our recommendation for surgeons is that, before proceeding with this treatment option, they investigate each component’s exact dimensions to ensure there are no subtle size differences that could cause problems. For example, a 28-mm head diameter that is actually 28.2 mm may affect mating properties, with the inner polyethylene articulation causing AIPD to develop.

Other cases of earlier IPD have been described, but they do not fit the APID definition given in this article. Riviere and colleagues14 reported the case of a 42-year-old man who, because of a previous adverse reaction to metal debris, underwent revision to a DM polyethylene ball in a retained BHR (Birmingham Hip Resurfacing) acetabular shell (Birmingham Hip, Smith & Nephew). Unfortunately, IPD occurred 14 months after surgery. Banka and colleagues15 reported the case of a 70-year-old woman who underwent revision to a DM cup for recurrent instability, but they did not specify the length of time between implantation and IPD and did not offer an explanation for the complication. Finally, Odland and Sierra16 reported the case of a 77-year-old man, with previous intertrochanteric and pelvic fractures, who underwent revision to a DM cup with retention of a Waldemar femoral component (Waldemar Link). He spontaneously developed IPD with ambulation 2 years after surgery.

Certainly, our patient’s presentation course is similar to other patients’. Within 3 months after revision to the MDM component, his left hip dislocated 3 times in 1 week. We contend his AIPD resulted from closed reduction, with the polyethylene dislodged from the femoral head with contact on the acetabulum. A larger or skirted neck may increase impingement during normal activity and thereby widen the polyethylene opening excessively and/or reduce the polyethylene ball ROM to impinge during the relocation maneuver. In this case, dissociation was noted only after the third dislocation. Pathognomonic eccentric positioning of the head in the acetabulum and, less commonly, the halo sign were evident on postreduction radiographs. Optimal treatment for AIPD of a DM component is controversial. Choices are limited to a constrained liner or, if possible, repeat DM with larger components. For recurrent dislocation, our patient underwent revision to an MDM component, but a femoral head with a skirted neck was used in an attempt to increase soft-tissue tension. During the second revision, minor eccentric wear of the inner articulation of the polyethylene component (consistent with impingement) was noted, and wear was visible on inspection of the outer articulation. We think his AIPD resulted from femoral neck impingement of the skirted head against the polyethylene ball.

AIPD is a discrete entity, with sudden failure of a DM component within 1 year after implantation. AIPD is characterized by dissociation of the femoral head from the inner articulation, resulting from impingement or closed reduction. More studies are needed to determine which patients with DM components are at highest risk and which treatment is most appropriate. We recommend taking extra care when reducing hips with this articulation and adopting a low threshold for general anesthesia use in the presence of paralysis.

Am J Orthop. 2017;46(3):E154-E159. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

 

 

References

1. Farizon F, de Lavison R, Azoulai JJ, Bousquet G. Results with a cementless alumina-coated cup with dual mobility. A twelve-year follow-up study. Int Orthop. 1998;22(4):219-224.

2. Lachiewicz PF, Watters TS. The use of dual-mobility components in total hip arthroplasty. J Am Acad Orthop Surg. 2012;20(8):481-486.

3. De Martino I, Triantafyllopoulos GK, Sculco PK, Sculco TP. Dual mobility cups in total hip arthroplasty. World J Orthop. 2014;5(3):180-187.

4. Lecuire F, Benareau I, Rubini J, Basso M. Intra-prosthetic dislocation of the Bousquet dual mobility socket [in French]. Rev Chir Orthop Reparatrice Appar Mot. 2004;90(3):249-255.

5. Philippot R, Boyer B, Farizon F. Intraprosthetic dislocation: a specific complication of the dual-mobility system. Clin Orthop Relat Res. 2013;471(3):965-970.

6. Stigbrand H, Ullmark G. Component dissociation after closed reduction of dual mobility sockets—a report of three cases. Hip Int. 2011;21(2):263-266.

7. Banzhof JA, Robbins CE, Ven AV, Talmo CT, Bono JV. Femoral head dislodgement complicating use of a dual mobility prosthesis for recurrent instability. J Arthroplasty. 2013;28(3):543.e1-e3.

8. Ward JP, McCardel BR, Hallstrom BR. Complete dissociation of the polyethylene component in a newly available dual-mobility bearing used in total hip arthroplasty: a case report. JBJS Case Connect. 2013;3(3):e94.

9. Bozic KJ, Kurtz SM, Lau E, Ong K, Vail TP, Berry DJ. The epidemiology of revision total hip arthroplasty in the United States. J Bone Joint Surg Am. 2009;91(1):128-133.

10. Parvizi J, Picinic E, Sharkey PF. Revision total hip arthroplasty for instability: surgical techniques and principles. J Bone Joint Surg Am. 2008;90(5):1134-1142.

11. Guyen O, Lewallen DG, Cabanela ME. Modes of failure of Osteonics constrained tripolar implants: a retrospective analysis of forty-three failed implants. J Bone Joint Surg Am. 2008;90(7):1553-1560.

12. Lachiewicz PF, Kelley SS. The use of constrained components in total hip arthroplasty. J Am Acad Orthop Surg. 2002;10(4):233-238.

13. Williams JT Jr, Ragland PS, Clarke S. Constrained components for the unstable hip following total hip arthroplasty: a literature review. Int Orthop. 2007;31(3):273-277.

14. Riviere C, Lavigne M, Alghamdi A, Vendittoli PA. Early failure of metal-on-metal large-diameter head total hip arthroplasty revised with a dual-mobility bearing: a case report. JBJS Case Connect. 2013;3(3):e95.

15. Banka TR, Ast MP, Parks ML. Early intraprosthetic dislocation in a revision dual-mobility hip prosthesis. Orthopedics. 2014;37(4):e395-e397.

16. Odland AN, Sierra RJ. Intraprosthetic dislocation of a contemporary dual-mobility design used during conversion THA. Orthopedics. 2014;37(12):e1124-e1128.

Article PDF
Author and Disclosure Information

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Acknowledgment: The authors thank Stephen Perlman for assisting with the literature search.

Issue
The American Journal of Orthopedics - 46(3)
Publications
Topics
Page Number
E154-E159
Sections
Author and Disclosure Information

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Acknowledgment: The authors thank Stephen Perlman for assisting with the literature search.

Author and Disclosure Information

Authors’ Disclosure Statement: The authors report no actual or potential conflict of interest in relation to this article.

Acknowledgment: The authors thank Stephen Perlman for assisting with the literature search.

Article PDF
Article PDF

Take-Home Points

  • AIPD of DM-THA is defined by dissociation within 1 year of implantation resulting from component impingement or closed reduction maneuvers.
  • This is a distinct entity from “late” IPD (>1 year) from implantation as this is associated most often with polyethylene wear, component loosening, and arthrofibrosis.
  • A history of DM dislocation followed by subjective “clunking,” instability, and a series of more frequent dislocations should raise concern for AIPD.
  • Classic radiographic findings of AIPD include eccentric hip reduction and soft tissue radiolucency (ie, halo sign) from dissociated polyethylene component.
  • Treating practitioners of AIPD should consider closed reduction with general anesthesia and sedation in the operating room to limit risk of dissociation.

Dual-mobility (DM) components were invented in the 1970s and have been used in primary and revision total hip arthroplasty (THA) in Europe ever since.1 However, DM components are most commonly used in the treatment of recurrent hip instability, and early results have been promising.2 In DM-THAs, a smaller (22-mm or 28-mm) metal femoral head snap-fits into a larger polyethylene ball (inner articulation), which articulates with a highly polished metal shell (outer articulation), which is either implanted directly in the acetabulum or placed in an uncemented acetabular cup. The 2 articulations used in these devices theoretically increase hip range of motion (ROM) and increase the inferior head displacement distance (jump distance) required for dislocation.3

However, this DM articulation with increased ROM may also cause chronic impingement of the femoral component neck or Morse taper against the outer polyethylene bearing, resulting in polyethylene wear and late intraprosthetic dissociation (IPD) (separation of inner articulation between femoral head and polyethylene liner). In 2004, Lecuire and colleagues4 reported 7 cases of IPD occurring a mean of 10 years after implantation during the period 1989 to 1997. In 2013, Philippot and colleagues5 reported that 81 of 1960 primary THAs developed IPD a mean of 9 years after implantation. These IPD cases were attributed to polyethylene wear or outer articulation blockage caused by arthrofibrosis or heterotopic ossification. Reports of acute IPD (AIPD), however, are rare. In 2011, Stigbrand and Ullmark6 reported 3 cases in which the DM prosthesis dislocated within 1 year after implantation. It was suggested that the inner metal head dissociated from the larger polyethylene component after attempted closed reduction for dislocation (separation of larger polyethylene component from acetabulum or acetabular liner).

DM components were unavailable to surgeons in the United States until 2011. The first US Food and Drug Administration (FDA)-approved DM device was the MDM (Modular Dual Mobility, Stryker). To our knowledge, 2 cases of AIPD with this prosthesis have been reported.7, 8 As with the cases in Europe, closed reduction was the suspected cause, but there was no explanation for the initial dislocation event.

In this article, we present the case of a nondemented man who developed AIPD of a THA with the MDM component and a 28-mm femoral head with a skirted neck (StelKast). His operative findings suggest a poor head-to-neck ratio caused by a larger diameter femoral neck or a skirted prosthesis, or a forceful reduction maneuver, may predispose DM components to AIPD. The patient provided written informed consent for print and electronic publication of this case report.

Case Report

In 2012, a 63-year-old man with a history of drug abuse underwent left primary THA. Seven posterior dislocations and 3 years later, the acetabular component was revised to the MDM prosthesis; the well-fixed StelKast femoral component was retained (Figure 1).

Figure 1.

Within 3 months after revision surgery, the left hip dislocated 3 times in 1 week, when the patient bent over to retrieve an object on the ground. The first 2 dislocations were treated with closed reduction under conscious sedation at an outside emergency department.

Figure 2.
Shortly after, the patient, with complaints of left hip pain and clunking, was seen by a physician assistant, but the treating team did not notice the eccentric reduction on radiographs. The third dislocation was treated with closed reduction under conscious sedation in the emergency department at our institution (Figure 2). Postreduction radiographs still showed the eccentric reduction, and a radiolucent halo was visible superior to the greater trochanter (Figure 3).
Figure 3.


With the patient’s erythrocyte sedimentation rate and C-reactive protein level both normal, a second revision was performed. During surgery, the polyethylene head was found beneath the gluteus maximus (Figure 4).
Figure 4.
Gross inspection revealed a small amount of eccentric polyethylene wear and metal debris of the inner articulation (Figure 5). As the abductor muscles were intact, it was decided to proceed with revision to a larger DM component and to downsize the femoral head to a skirtless component (Table, Figure 6).

 

 

Discussion

Recurrent dislocation and instability accounts for 22.5% of THA revisions in the United States.9 Until 2011, options for managing recurrent dislocation in the United States included modular component exchange, component revision for malposition, and use of constrained components.10

Figure 5.
However, the decreased motion of constrained components may produce excess stress that eventually results in failure.11-13

In 1974, Bousquet first reported use of the DM prosthesis in primary THA; the prosthesis allowed increased stability without sacrificing motion or fixation.1 However, longer-term studies of DM components disclosed a new complication, IPD. In 2004, Lecuire and colleagues4 reported 7 cases of IPD occurring a mean of 10 years after implantation of the Bousquet prosthesis.

Table.
Philippot and colleagues5 reported that 81 of 1960 primary THAs with DM components developed IPD a mean of 9 years after implantation. They described 3 types of IPD based on mechanism of injury: type I, caused by wear of the inner articulation without arthrofibrosis or cup loosening (n = 26); type II, resulting from blocked outer articulation motion, caused by arthrofibrosis, nonunion, calcification, or heterotopic ossification (n = 41); and type III, associated with acetabular component loosening (n = 14). IPD occurred an average of 11 years (type I), 8 years (type II), and 9 years (type III) after implantation.

AIPD, which occurs within 1 year after implantation, has been reported much less often than late IPD. Stigbrand and Ullmark6 reported 3 cases of AIPD that developed within 7 months after implantation of Amplitude and Advantage (Zimmer Biomet) DM prostheses.
Figure 6.
The authors proposed that AIPD is related to incomplete coupling of the metal head and the inner polyethylene liner or to shearing of the large polyethylene component on the acetabular rim during a closed reduction maneuver. According to their description, the femoral head in the acetabulum had an “eccentric” radiographic appearance. The authors recommended administering muscle relaxants during closed reduction to avoid dissociation of the liner during the reduction.

This unusual complication apparently is not confined to a specific implant or region. Since the MDM component was introduced in the United States, 2 more cases of AIPD have been identified (Table). Banzhof and colleagues7 reported the case of a 68-year-old woman who, 2 months after the MDM was placed for recurrent instability, dislocated the component while rising from a seated position. Her IPD most likely resulted from a closed reduction. The affected hip eventually required closed reduction in the operating room. Postreduction radiographs showed the characteristic eccentric appearance; a halo, also visible in the soft tissues, corresponded with the dissociated radiolucent polyethylene liner. The authors attributed the early failure to an eccentrically seated metal liner that separated the locking mechanism. The MDM component was revised to a conventional THA, with the femoral head upsized and length added.

Ward and colleagues8 reported the case of an 87-year-old woman who had a conventional THA revised to an MDM component for recurrent instability. Two months after surgery, this patient, who had dementia, experienced 2 posterior dislocations while rising from a chair. Closed reduction in the emergency department seemed successful, but later she presented to the surgeon’s office with symptoms of instability and clunking, complaints similar to our patient’s. Radiographs showed an eccentric reduction caused by IPD, and the MDM component was revised to a constrained liner. Adding a MDM component to a retained DePuy (DePuy Synthes) femoral stem and head is considered “off-label use,” which, the authors proposed, may have been related to the AIPD in their patient’s case. However, one manufacturer’s femoral component and head are often mated with another manufacturer’s acetabular component to allow for a less complex revision. Our recommendation for surgeons is that, before proceeding with this treatment option, they investigate each component’s exact dimensions to ensure there are no subtle size differences that could cause problems. For example, a 28-mm head diameter that is actually 28.2 mm may affect mating properties, with the inner polyethylene articulation causing AIPD to develop.

Other cases of earlier IPD have been described, but they do not fit the APID definition given in this article. Riviere and colleagues14 reported the case of a 42-year-old man who, because of a previous adverse reaction to metal debris, underwent revision to a DM polyethylene ball in a retained BHR (Birmingham Hip Resurfacing) acetabular shell (Birmingham Hip, Smith & Nephew). Unfortunately, IPD occurred 14 months after surgery. Banka and colleagues15 reported the case of a 70-year-old woman who underwent revision to a DM cup for recurrent instability, but they did not specify the length of time between implantation and IPD and did not offer an explanation for the complication. Finally, Odland and Sierra16 reported the case of a 77-year-old man, with previous intertrochanteric and pelvic fractures, who underwent revision to a DM cup with retention of a Waldemar femoral component (Waldemar Link). He spontaneously developed IPD with ambulation 2 years after surgery.

Certainly, our patient’s presentation course is similar to other patients’. Within 3 months after revision to the MDM component, his left hip dislocated 3 times in 1 week. We contend his AIPD resulted from closed reduction, with the polyethylene dislodged from the femoral head with contact on the acetabulum. A larger or skirted neck may increase impingement during normal activity and thereby widen the polyethylene opening excessively and/or reduce the polyethylene ball ROM to impinge during the relocation maneuver. In this case, dissociation was noted only after the third dislocation. Pathognomonic eccentric positioning of the head in the acetabulum and, less commonly, the halo sign were evident on postreduction radiographs. Optimal treatment for AIPD of a DM component is controversial. Choices are limited to a constrained liner or, if possible, repeat DM with larger components. For recurrent dislocation, our patient underwent revision to an MDM component, but a femoral head with a skirted neck was used in an attempt to increase soft-tissue tension. During the second revision, minor eccentric wear of the inner articulation of the polyethylene component (consistent with impingement) was noted, and wear was visible on inspection of the outer articulation. We think his AIPD resulted from femoral neck impingement of the skirted head against the polyethylene ball.

AIPD is a discrete entity, with sudden failure of a DM component within 1 year after implantation. AIPD is characterized by dissociation of the femoral head from the inner articulation, resulting from impingement or closed reduction. More studies are needed to determine which patients with DM components are at highest risk and which treatment is most appropriate. We recommend taking extra care when reducing hips with this articulation and adopting a low threshold for general anesthesia use in the presence of paralysis.

Am J Orthop. 2017;46(3):E154-E159. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

 

 

Take-Home Points

  • AIPD of DM-THA is defined by dissociation within 1 year of implantation resulting from component impingement or closed reduction maneuvers.
  • This is a distinct entity from “late” IPD (>1 year) from implantation as this is associated most often with polyethylene wear, component loosening, and arthrofibrosis.
  • A history of DM dislocation followed by subjective “clunking,” instability, and a series of more frequent dislocations should raise concern for AIPD.
  • Classic radiographic findings of AIPD include eccentric hip reduction and soft tissue radiolucency (ie, halo sign) from dissociated polyethylene component.
  • Treating practitioners of AIPD should consider closed reduction with general anesthesia and sedation in the operating room to limit risk of dissociation.

Dual-mobility (DM) components were invented in the 1970s and have been used in primary and revision total hip arthroplasty (THA) in Europe ever since.1 However, DM components are most commonly used in the treatment of recurrent hip instability, and early results have been promising.2 In DM-THAs, a smaller (22-mm or 28-mm) metal femoral head snap-fits into a larger polyethylene ball (inner articulation), which articulates with a highly polished metal shell (outer articulation), which is either implanted directly in the acetabulum or placed in an uncemented acetabular cup. The 2 articulations used in these devices theoretically increase hip range of motion (ROM) and increase the inferior head displacement distance (jump distance) required for dislocation.3

However, this DM articulation with increased ROM may also cause chronic impingement of the femoral component neck or Morse taper against the outer polyethylene bearing, resulting in polyethylene wear and late intraprosthetic dissociation (IPD) (separation of inner articulation between femoral head and polyethylene liner). In 2004, Lecuire and colleagues4 reported 7 cases of IPD occurring a mean of 10 years after implantation during the period 1989 to 1997. In 2013, Philippot and colleagues5 reported that 81 of 1960 primary THAs developed IPD a mean of 9 years after implantation. These IPD cases were attributed to polyethylene wear or outer articulation blockage caused by arthrofibrosis or heterotopic ossification. Reports of acute IPD (AIPD), however, are rare. In 2011, Stigbrand and Ullmark6 reported 3 cases in which the DM prosthesis dislocated within 1 year after implantation. It was suggested that the inner metal head dissociated from the larger polyethylene component after attempted closed reduction for dislocation (separation of larger polyethylene component from acetabulum or acetabular liner).

DM components were unavailable to surgeons in the United States until 2011. The first US Food and Drug Administration (FDA)-approved DM device was the MDM (Modular Dual Mobility, Stryker). To our knowledge, 2 cases of AIPD with this prosthesis have been reported.7, 8 As with the cases in Europe, closed reduction was the suspected cause, but there was no explanation for the initial dislocation event.

In this article, we present the case of a nondemented man who developed AIPD of a THA with the MDM component and a 28-mm femoral head with a skirted neck (StelKast). His operative findings suggest a poor head-to-neck ratio caused by a larger diameter femoral neck or a skirted prosthesis, or a forceful reduction maneuver, may predispose DM components to AIPD. The patient provided written informed consent for print and electronic publication of this case report.

Case Report

In 2012, a 63-year-old man with a history of drug abuse underwent left primary THA. Seven posterior dislocations and 3 years later, the acetabular component was revised to the MDM prosthesis; the well-fixed StelKast femoral component was retained (Figure 1).

Figure 1.

Within 3 months after revision surgery, the left hip dislocated 3 times in 1 week, when the patient bent over to retrieve an object on the ground. The first 2 dislocations were treated with closed reduction under conscious sedation at an outside emergency department.

Figure 2.
Shortly after, the patient, with complaints of left hip pain and clunking, was seen by a physician assistant, but the treating team did not notice the eccentric reduction on radiographs. The third dislocation was treated with closed reduction under conscious sedation in the emergency department at our institution (Figure 2). Postreduction radiographs still showed the eccentric reduction, and a radiolucent halo was visible superior to the greater trochanter (Figure 3).
Figure 3.


With the patient’s erythrocyte sedimentation rate and C-reactive protein level both normal, a second revision was performed. During surgery, the polyethylene head was found beneath the gluteus maximus (Figure 4).
Figure 4.
Gross inspection revealed a small amount of eccentric polyethylene wear and metal debris of the inner articulation (Figure 5). As the abductor muscles were intact, it was decided to proceed with revision to a larger DM component and to downsize the femoral head to a skirtless component (Table, Figure 6).

 

 

Discussion

Recurrent dislocation and instability accounts for 22.5% of THA revisions in the United States.9 Until 2011, options for managing recurrent dislocation in the United States included modular component exchange, component revision for malposition, and use of constrained components.10

Figure 5.
However, the decreased motion of constrained components may produce excess stress that eventually results in failure.11-13

In 1974, Bousquet first reported use of the DM prosthesis in primary THA; the prosthesis allowed increased stability without sacrificing motion or fixation.1 However, longer-term studies of DM components disclosed a new complication, IPD. In 2004, Lecuire and colleagues4 reported 7 cases of IPD occurring a mean of 10 years after implantation of the Bousquet prosthesis.

Table.
Philippot and colleagues5 reported that 81 of 1960 primary THAs with DM components developed IPD a mean of 9 years after implantation. They described 3 types of IPD based on mechanism of injury: type I, caused by wear of the inner articulation without arthrofibrosis or cup loosening (n = 26); type II, resulting from blocked outer articulation motion, caused by arthrofibrosis, nonunion, calcification, or heterotopic ossification (n = 41); and type III, associated with acetabular component loosening (n = 14). IPD occurred an average of 11 years (type I), 8 years (type II), and 9 years (type III) after implantation.

AIPD, which occurs within 1 year after implantation, has been reported much less often than late IPD. Stigbrand and Ullmark6 reported 3 cases of AIPD that developed within 7 months after implantation of Amplitude and Advantage (Zimmer Biomet) DM prostheses.
Figure 6.
The authors proposed that AIPD is related to incomplete coupling of the metal head and the inner polyethylene liner or to shearing of the large polyethylene component on the acetabular rim during a closed reduction maneuver. According to their description, the femoral head in the acetabulum had an “eccentric” radiographic appearance. The authors recommended administering muscle relaxants during closed reduction to avoid dissociation of the liner during the reduction.

This unusual complication apparently is not confined to a specific implant or region. Since the MDM component was introduced in the United States, 2 more cases of AIPD have been identified (Table). Banzhof and colleagues7 reported the case of a 68-year-old woman who, 2 months after the MDM was placed for recurrent instability, dislocated the component while rising from a seated position. Her IPD most likely resulted from a closed reduction. The affected hip eventually required closed reduction in the operating room. Postreduction radiographs showed the characteristic eccentric appearance; a halo, also visible in the soft tissues, corresponded with the dissociated radiolucent polyethylene liner. The authors attributed the early failure to an eccentrically seated metal liner that separated the locking mechanism. The MDM component was revised to a conventional THA, with the femoral head upsized and length added.

Ward and colleagues8 reported the case of an 87-year-old woman who had a conventional THA revised to an MDM component for recurrent instability. Two months after surgery, this patient, who had dementia, experienced 2 posterior dislocations while rising from a chair. Closed reduction in the emergency department seemed successful, but later she presented to the surgeon’s office with symptoms of instability and clunking, complaints similar to our patient’s. Radiographs showed an eccentric reduction caused by IPD, and the MDM component was revised to a constrained liner. Adding a MDM component to a retained DePuy (DePuy Synthes) femoral stem and head is considered “off-label use,” which, the authors proposed, may have been related to the AIPD in their patient’s case. However, one manufacturer’s femoral component and head are often mated with another manufacturer’s acetabular component to allow for a less complex revision. Our recommendation for surgeons is that, before proceeding with this treatment option, they investigate each component’s exact dimensions to ensure there are no subtle size differences that could cause problems. For example, a 28-mm head diameter that is actually 28.2 mm may affect mating properties, with the inner polyethylene articulation causing AIPD to develop.

Other cases of earlier IPD have been described, but they do not fit the APID definition given in this article. Riviere and colleagues14 reported the case of a 42-year-old man who, because of a previous adverse reaction to metal debris, underwent revision to a DM polyethylene ball in a retained BHR (Birmingham Hip Resurfacing) acetabular shell (Birmingham Hip, Smith & Nephew). Unfortunately, IPD occurred 14 months after surgery. Banka and colleagues15 reported the case of a 70-year-old woman who underwent revision to a DM cup for recurrent instability, but they did not specify the length of time between implantation and IPD and did not offer an explanation for the complication. Finally, Odland and Sierra16 reported the case of a 77-year-old man, with previous intertrochanteric and pelvic fractures, who underwent revision to a DM cup with retention of a Waldemar femoral component (Waldemar Link). He spontaneously developed IPD with ambulation 2 years after surgery.

Certainly, our patient’s presentation course is similar to other patients’. Within 3 months after revision to the MDM component, his left hip dislocated 3 times in 1 week. We contend his AIPD resulted from closed reduction, with the polyethylene dislodged from the femoral head with contact on the acetabulum. A larger or skirted neck may increase impingement during normal activity and thereby widen the polyethylene opening excessively and/or reduce the polyethylene ball ROM to impinge during the relocation maneuver. In this case, dissociation was noted only after the third dislocation. Pathognomonic eccentric positioning of the head in the acetabulum and, less commonly, the halo sign were evident on postreduction radiographs. Optimal treatment for AIPD of a DM component is controversial. Choices are limited to a constrained liner or, if possible, repeat DM with larger components. For recurrent dislocation, our patient underwent revision to an MDM component, but a femoral head with a skirted neck was used in an attempt to increase soft-tissue tension. During the second revision, minor eccentric wear of the inner articulation of the polyethylene component (consistent with impingement) was noted, and wear was visible on inspection of the outer articulation. We think his AIPD resulted from femoral neck impingement of the skirted head against the polyethylene ball.

AIPD is a discrete entity, with sudden failure of a DM component within 1 year after implantation. AIPD is characterized by dissociation of the femoral head from the inner articulation, resulting from impingement or closed reduction. More studies are needed to determine which patients with DM components are at highest risk and which treatment is most appropriate. We recommend taking extra care when reducing hips with this articulation and adopting a low threshold for general anesthesia use in the presence of paralysis.

Am J Orthop. 2017;46(3):E154-E159. Copyright Frontline Medical Communications Inc. 2017. All rights reserved.

 

 

References

1. Farizon F, de Lavison R, Azoulai JJ, Bousquet G. Results with a cementless alumina-coated cup with dual mobility. A twelve-year follow-up study. Int Orthop. 1998;22(4):219-224.

2. Lachiewicz PF, Watters TS. The use of dual-mobility components in total hip arthroplasty. J Am Acad Orthop Surg. 2012;20(8):481-486.

3. De Martino I, Triantafyllopoulos GK, Sculco PK, Sculco TP. Dual mobility cups in total hip arthroplasty. World J Orthop. 2014;5(3):180-187.

4. Lecuire F, Benareau I, Rubini J, Basso M. Intra-prosthetic dislocation of the Bousquet dual mobility socket [in French]. Rev Chir Orthop Reparatrice Appar Mot. 2004;90(3):249-255.

5. Philippot R, Boyer B, Farizon F. Intraprosthetic dislocation: a specific complication of the dual-mobility system. Clin Orthop Relat Res. 2013;471(3):965-970.

6. Stigbrand H, Ullmark G. Component dissociation after closed reduction of dual mobility sockets—a report of three cases. Hip Int. 2011;21(2):263-266.

7. Banzhof JA, Robbins CE, Ven AV, Talmo CT, Bono JV. Femoral head dislodgement complicating use of a dual mobility prosthesis for recurrent instability. J Arthroplasty. 2013;28(3):543.e1-e3.

8. Ward JP, McCardel BR, Hallstrom BR. Complete dissociation of the polyethylene component in a newly available dual-mobility bearing used in total hip arthroplasty: a case report. JBJS Case Connect. 2013;3(3):e94.

9. Bozic KJ, Kurtz SM, Lau E, Ong K, Vail TP, Berry DJ. The epidemiology of revision total hip arthroplasty in the United States. J Bone Joint Surg Am. 2009;91(1):128-133.

10. Parvizi J, Picinic E, Sharkey PF. Revision total hip arthroplasty for instability: surgical techniques and principles. J Bone Joint Surg Am. 2008;90(5):1134-1142.

11. Guyen O, Lewallen DG, Cabanela ME. Modes of failure of Osteonics constrained tripolar implants: a retrospective analysis of forty-three failed implants. J Bone Joint Surg Am. 2008;90(7):1553-1560.

12. Lachiewicz PF, Kelley SS. The use of constrained components in total hip arthroplasty. J Am Acad Orthop Surg. 2002;10(4):233-238.

13. Williams JT Jr, Ragland PS, Clarke S. Constrained components for the unstable hip following total hip arthroplasty: a literature review. Int Orthop. 2007;31(3):273-277.

14. Riviere C, Lavigne M, Alghamdi A, Vendittoli PA. Early failure of metal-on-metal large-diameter head total hip arthroplasty revised with a dual-mobility bearing: a case report. JBJS Case Connect. 2013;3(3):e95.

15. Banka TR, Ast MP, Parks ML. Early intraprosthetic dislocation in a revision dual-mobility hip prosthesis. Orthopedics. 2014;37(4):e395-e397.

16. Odland AN, Sierra RJ. Intraprosthetic dislocation of a contemporary dual-mobility design used during conversion THA. Orthopedics. 2014;37(12):e1124-e1128.

References

1. Farizon F, de Lavison R, Azoulai JJ, Bousquet G. Results with a cementless alumina-coated cup with dual mobility. A twelve-year follow-up study. Int Orthop. 1998;22(4):219-224.

2. Lachiewicz PF, Watters TS. The use of dual-mobility components in total hip arthroplasty. J Am Acad Orthop Surg. 2012;20(8):481-486.

3. De Martino I, Triantafyllopoulos GK, Sculco PK, Sculco TP. Dual mobility cups in total hip arthroplasty. World J Orthop. 2014;5(3):180-187.

4. Lecuire F, Benareau I, Rubini J, Basso M. Intra-prosthetic dislocation of the Bousquet dual mobility socket [in French]. Rev Chir Orthop Reparatrice Appar Mot. 2004;90(3):249-255.

5. Philippot R, Boyer B, Farizon F. Intraprosthetic dislocation: a specific complication of the dual-mobility system. Clin Orthop Relat Res. 2013;471(3):965-970.

6. Stigbrand H, Ullmark G. Component dissociation after closed reduction of dual mobility sockets—a report of three cases. Hip Int. 2011;21(2):263-266.

7. Banzhof JA, Robbins CE, Ven AV, Talmo CT, Bono JV. Femoral head dislodgement complicating use of a dual mobility prosthesis for recurrent instability. J Arthroplasty. 2013;28(3):543.e1-e3.

8. Ward JP, McCardel BR, Hallstrom BR. Complete dissociation of the polyethylene component in a newly available dual-mobility bearing used in total hip arthroplasty: a case report. JBJS Case Connect. 2013;3(3):e94.

9. Bozic KJ, Kurtz SM, Lau E, Ong K, Vail TP, Berry DJ. The epidemiology of revision total hip arthroplasty in the United States. J Bone Joint Surg Am. 2009;91(1):128-133.

10. Parvizi J, Picinic E, Sharkey PF. Revision total hip arthroplasty for instability: surgical techniques and principles. J Bone Joint Surg Am. 2008;90(5):1134-1142.

11. Guyen O, Lewallen DG, Cabanela ME. Modes of failure of Osteonics constrained tripolar implants: a retrospective analysis of forty-three failed implants. J Bone Joint Surg Am. 2008;90(7):1553-1560.

12. Lachiewicz PF, Kelley SS. The use of constrained components in total hip arthroplasty. J Am Acad Orthop Surg. 2002;10(4):233-238.

13. Williams JT Jr, Ragland PS, Clarke S. Constrained components for the unstable hip following total hip arthroplasty: a literature review. Int Orthop. 2007;31(3):273-277.

14. Riviere C, Lavigne M, Alghamdi A, Vendittoli PA. Early failure of metal-on-metal large-diameter head total hip arthroplasty revised with a dual-mobility bearing: a case report. JBJS Case Connect. 2013;3(3):e95.

15. Banka TR, Ast MP, Parks ML. Early intraprosthetic dislocation in a revision dual-mobility hip prosthesis. Orthopedics. 2014;37(4):e395-e397.

16. Odland AN, Sierra RJ. Intraprosthetic dislocation of a contemporary dual-mobility design used during conversion THA. Orthopedics. 2014;37(12):e1124-e1128.

Issue
The American Journal of Orthopedics - 46(3)
Issue
The American Journal of Orthopedics - 46(3)
Page Number
E154-E159
Page Number
E154-E159
Publications
Publications
Topics
Article Type
Display Headline
Acute Intraprosthetic Dissociation of a Dual-Mobility Hip in the United States
Display Headline
Acute Intraprosthetic Dissociation of a Dual-Mobility Hip in the United States
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media

Is the doctor in?

Article Type
Changed
Fri, 09/14/2018 - 11:59
The growth of telehospitalist programs

 

Within hospital medicine, there has been a recent increase in programs that provide virtual or telehealth hospitalists, primarily to hospitals that are small, remote, and/or understaffed. According to a 2013 Cisco health care customer experience report, the number of telehealth consumers will likely markedly increase to at least 7 million by 2018.1

Since telehospitalist programs are still relatively new, there are many questions about why and how they exist and how they are (and can be) funded. Questions also remain about some limitations of telehospitalist programs for both the “givers” and the “receivers” of the services. I tackle some of these questions in this article.

What is a telehospitalist?

Dr. Danielle Scheurer
A telehospitalist is a hospitalist who provides remote services to patients and providers in need of such services. These services can range from initial encounters, follow-up encounters, post-acute care visits, home visits, consultations, and emergency care.

What are the drivers of telehospitalist programs?

One primary driver of telehealth (and specifically telehospitalist) programs is an ongoing shortage of hospitalists, especially in remote areas and critical access hospitals where coverage issues are especially prominent at night and/or on weekends. In many hospitals, there is also a growing unwillingness on the part of physicians to be routinely on call at night. Although working on call used to be on par with being a physician, many younger-generation physicians are less willing to blur “work and life.” This increases the need for dedicated night coverage in many hospitals.

Another driver for some programs (especially at tertiary care medical centers) is a desire to more thoroughly assess patients prior to transfer to their respective centers (the alternative being a phone conversation with the transferring center about the patient’s status). There is also a growing desire to keep patients local if possible, which is usually better for the patient and the family and can decrease the total cost of their care.

Another catalyst to telehospitalist program growth is the growing cultural comfort level with two-way video interactions, such as Skype and FaceTime. Since videoconferencing has permeated most of our professional and personal lives, telehealth seems familiar and comfortable for both providers and patients. In a recent consumer survey, three out of every four consumers responded that they are very comfortable communicating with providers via technology, as opposed to seeing them in person.1

Another driver for some programs is financial. Depending on the way the program is structured, it can be not only financially feasible but financially beneficial, especially if the program can consolidate coverage across multiple sites (more on this later).

One other driver for some health care systems is the need to cover areas with on-site nurse practitioners and physician assistants. Using a telehospitalist makes it easier to get appropriate and required oversight for this coverage model across time and space.

What are the advantages of being a telehospitalist?

Some of the career advantages of being a telehospitalist include the shift flexibility and convenience. This work allows a hospitalist to serve a shift from anywhere in the world and from the convenience of their home. Some telehospitalists can easily work local night shifts when they live many time zones away (and therefore, don’t actually have to work a night shift). Many programs are designed to have a single hospitalist cover many hospitals over a wide geography, which would be logistically impossible to do in person. This is especially appealing for multihospital systems that cannot afford to have a hospitalist on site at each location.

The earning potential can also be appealing, depending on the number of shifts a hospitalist is willing to work.

What are the limitations of being a telehospitalist?

There are limits to what a telehospitalist can perform, many of which depend on the manner in which the program and the technology are arranged. Telehealth can vary from a cart-based videoconferencing system that is transported into a patient’s room to an independent robot that travels throughout sites. The primary limitation is the need to rely on someone in the patient’s room to act as virtual hands. This usually falls to the bedside nurse and requires a good working relationship and patience on their part. The bedside nurses have to “buy into” the program in advance and may need to have scripting for how to explain the process to the patients.

Another major challenge is interacting with different electronic health record systems. Becoming agile with a single EHR is challenging enough, but maneuvering several of them in a single shift can be extremely trying. Telehospitalists can also be challenged by technology glitches or failures that need troubleshooting both on their end and on-site. Although these problems are rare, there will always be a concern that the patient will not get his or her needs met if the technology fails.

 

 

How does the financing work?

Although this is a rapidly changing landscape, telehospitalists are not currently able to generate much revenue from professional billing. Unlike in-person visits, Medicare will not reimburse professional fees for telehospitalist visits. Although each payer is unique, most other (nonMedicare) payers are also not willing to reimburse for televisits. This may change in the future, however, as Medicare does pay for virtual specialty services such as telestroke. In addition, many states have enacted telemedicine parity laws, which require private payers to pay for all health care services equally, regardless of modality (audio, video, or in person).

For now, the financial case for employing telehospitalists for most programs has to be made using benfits other than the generation of professional fees. For telehospitalist programs that can cover several sites, the cost is substantially less than employing individual on-site hospitalists to do low-volume work. Telehospitalist programs are also, likely, less costly than is locum tenens staffing. For programs that evaluate the need for transfers, a case can be made that keeping a patient in a smaller, low-cost venue, rather than transferring them to a larger, higher-cost venue, can also reduce overall cost for a health care system.

What about licensing and credentialing?

Telehospitalists can be hindered by the need to have a license in several states and to be credentialed in several systems. This can be cumbersome, time-consuming, and expensive. To ease the multistate licensing burden, the Interstate Medical Licensure Compact has been established.2 This is an accelerated licensure process for eligible physicians that improves license portability across states. There are currently 18 states that participate, and the number continues to increase.

For credentialing, most hospitals require initial credentialing and full recredentialing every 2 years. Maintaining credentials at several sites can be extremely time consuming. To ease this burden, some hospitals with telehealth programs have adopted “credentialing by proxy,” which means that one hospital will accept the credentialing process of another facility.

What next?

In summary, there has been and will likely continue to be explosive growth of telehospitalist programs and providers for all the reasons outlined above. Although some barriers to efficient and effective practice do exist, many of those barriers are being overcome quite rapidly. I expect this growth to continue for the betterment of hospitalists, our patients, and the systems in which we work. For a more in-depth look into telemedicine in hospital medicine, view a report created by a work group of SHM's Practice Management Committee.

Dr. Scheurer is a hospitalist and chief quality officer at the Medical University of South Carolina in Charleston. She is physician editor of The Hospitalist. Email her at [email protected].

References

1.Cisco. (2013 March 4). Cisco Study Reveals 74 Percent of Consumers Open to Virtual Doctor Visit. Cisco: The Network. Retrieved from https://newsroom.cisco.com/press-release-content?type=webcontent&articleId=1148539.

2. Interstate Medical Licensure Compact Commission. (2017). Interstate Medical Licensure Compact. Retrieved from http://www.licenseportability.org/index.html.

Publications
Topics
Sections
The growth of telehospitalist programs
The growth of telehospitalist programs

 

Within hospital medicine, there has been a recent increase in programs that provide virtual or telehealth hospitalists, primarily to hospitals that are small, remote, and/or understaffed. According to a 2013 Cisco health care customer experience report, the number of telehealth consumers will likely markedly increase to at least 7 million by 2018.1

Since telehospitalist programs are still relatively new, there are many questions about why and how they exist and how they are (and can be) funded. Questions also remain about some limitations of telehospitalist programs for both the “givers” and the “receivers” of the services. I tackle some of these questions in this article.

What is a telehospitalist?

Dr. Danielle Scheurer
A telehospitalist is a hospitalist who provides remote services to patients and providers in need of such services. These services can range from initial encounters, follow-up encounters, post-acute care visits, home visits, consultations, and emergency care.

What are the drivers of telehospitalist programs?

One primary driver of telehealth (and specifically telehospitalist) programs is an ongoing shortage of hospitalists, especially in remote areas and critical access hospitals where coverage issues are especially prominent at night and/or on weekends. In many hospitals, there is also a growing unwillingness on the part of physicians to be routinely on call at night. Although working on call used to be on par with being a physician, many younger-generation physicians are less willing to blur “work and life.” This increases the need for dedicated night coverage in many hospitals.

Another driver for some programs (especially at tertiary care medical centers) is a desire to more thoroughly assess patients prior to transfer to their respective centers (the alternative being a phone conversation with the transferring center about the patient’s status). There is also a growing desire to keep patients local if possible, which is usually better for the patient and the family and can decrease the total cost of their care.

Another catalyst to telehospitalist program growth is the growing cultural comfort level with two-way video interactions, such as Skype and FaceTime. Since videoconferencing has permeated most of our professional and personal lives, telehealth seems familiar and comfortable for both providers and patients. In a recent consumer survey, three out of every four consumers responded that they are very comfortable communicating with providers via technology, as opposed to seeing them in person.1

Another driver for some programs is financial. Depending on the way the program is structured, it can be not only financially feasible but financially beneficial, especially if the program can consolidate coverage across multiple sites (more on this later).

One other driver for some health care systems is the need to cover areas with on-site nurse practitioners and physician assistants. Using a telehospitalist makes it easier to get appropriate and required oversight for this coverage model across time and space.

What are the advantages of being a telehospitalist?

Some of the career advantages of being a telehospitalist include the shift flexibility and convenience. This work allows a hospitalist to serve a shift from anywhere in the world and from the convenience of their home. Some telehospitalists can easily work local night shifts when they live many time zones away (and therefore, don’t actually have to work a night shift). Many programs are designed to have a single hospitalist cover many hospitals over a wide geography, which would be logistically impossible to do in person. This is especially appealing for multihospital systems that cannot afford to have a hospitalist on site at each location.

The earning potential can also be appealing, depending on the number of shifts a hospitalist is willing to work.

What are the limitations of being a telehospitalist?

There are limits to what a telehospitalist can perform, many of which depend on the manner in which the program and the technology are arranged. Telehealth can vary from a cart-based videoconferencing system that is transported into a patient’s room to an independent robot that travels throughout sites. The primary limitation is the need to rely on someone in the patient’s room to act as virtual hands. This usually falls to the bedside nurse and requires a good working relationship and patience on their part. The bedside nurses have to “buy into” the program in advance and may need to have scripting for how to explain the process to the patients.

Another major challenge is interacting with different electronic health record systems. Becoming agile with a single EHR is challenging enough, but maneuvering several of them in a single shift can be extremely trying. Telehospitalists can also be challenged by technology glitches or failures that need troubleshooting both on their end and on-site. Although these problems are rare, there will always be a concern that the patient will not get his or her needs met if the technology fails.

 

 

How does the financing work?

Although this is a rapidly changing landscape, telehospitalists are not currently able to generate much revenue from professional billing. Unlike in-person visits, Medicare will not reimburse professional fees for telehospitalist visits. Although each payer is unique, most other (nonMedicare) payers are also not willing to reimburse for televisits. This may change in the future, however, as Medicare does pay for virtual specialty services such as telestroke. In addition, many states have enacted telemedicine parity laws, which require private payers to pay for all health care services equally, regardless of modality (audio, video, or in person).

For now, the financial case for employing telehospitalists for most programs has to be made using benfits other than the generation of professional fees. For telehospitalist programs that can cover several sites, the cost is substantially less than employing individual on-site hospitalists to do low-volume work. Telehospitalist programs are also, likely, less costly than is locum tenens staffing. For programs that evaluate the need for transfers, a case can be made that keeping a patient in a smaller, low-cost venue, rather than transferring them to a larger, higher-cost venue, can also reduce overall cost for a health care system.

What about licensing and credentialing?

Telehospitalists can be hindered by the need to have a license in several states and to be credentialed in several systems. This can be cumbersome, time-consuming, and expensive. To ease the multistate licensing burden, the Interstate Medical Licensure Compact has been established.2 This is an accelerated licensure process for eligible physicians that improves license portability across states. There are currently 18 states that participate, and the number continues to increase.

For credentialing, most hospitals require initial credentialing and full recredentialing every 2 years. Maintaining credentials at several sites can be extremely time consuming. To ease this burden, some hospitals with telehealth programs have adopted “credentialing by proxy,” which means that one hospital will accept the credentialing process of another facility.

What next?

In summary, there has been and will likely continue to be explosive growth of telehospitalist programs and providers for all the reasons outlined above. Although some barriers to efficient and effective practice do exist, many of those barriers are being overcome quite rapidly. I expect this growth to continue for the betterment of hospitalists, our patients, and the systems in which we work. For a more in-depth look into telemedicine in hospital medicine, view a report created by a work group of SHM's Practice Management Committee.

Dr. Scheurer is a hospitalist and chief quality officer at the Medical University of South Carolina in Charleston. She is physician editor of The Hospitalist. Email her at [email protected].

References

1.Cisco. (2013 March 4). Cisco Study Reveals 74 Percent of Consumers Open to Virtual Doctor Visit. Cisco: The Network. Retrieved from https://newsroom.cisco.com/press-release-content?type=webcontent&articleId=1148539.

2. Interstate Medical Licensure Compact Commission. (2017). Interstate Medical Licensure Compact. Retrieved from http://www.licenseportability.org/index.html.

 

Within hospital medicine, there has been a recent increase in programs that provide virtual or telehealth hospitalists, primarily to hospitals that are small, remote, and/or understaffed. According to a 2013 Cisco health care customer experience report, the number of telehealth consumers will likely markedly increase to at least 7 million by 2018.1

Since telehospitalist programs are still relatively new, there are many questions about why and how they exist and how they are (and can be) funded. Questions also remain about some limitations of telehospitalist programs for both the “givers” and the “receivers” of the services. I tackle some of these questions in this article.

What is a telehospitalist?

Dr. Danielle Scheurer
A telehospitalist is a hospitalist who provides remote services to patients and providers in need of such services. These services can range from initial encounters, follow-up encounters, post-acute care visits, home visits, consultations, and emergency care.

What are the drivers of telehospitalist programs?

One primary driver of telehealth (and specifically telehospitalist) programs is an ongoing shortage of hospitalists, especially in remote areas and critical access hospitals where coverage issues are especially prominent at night and/or on weekends. In many hospitals, there is also a growing unwillingness on the part of physicians to be routinely on call at night. Although working on call used to be on par with being a physician, many younger-generation physicians are less willing to blur “work and life.” This increases the need for dedicated night coverage in many hospitals.

Another driver for some programs (especially at tertiary care medical centers) is a desire to more thoroughly assess patients prior to transfer to their respective centers (the alternative being a phone conversation with the transferring center about the patient’s status). There is also a growing desire to keep patients local if possible, which is usually better for the patient and the family and can decrease the total cost of their care.

Another catalyst to telehospitalist program growth is the growing cultural comfort level with two-way video interactions, such as Skype and FaceTime. Since videoconferencing has permeated most of our professional and personal lives, telehealth seems familiar and comfortable for both providers and patients. In a recent consumer survey, three out of every four consumers responded that they are very comfortable communicating with providers via technology, as opposed to seeing them in person.1

Another driver for some programs is financial. Depending on the way the program is structured, it can be not only financially feasible but financially beneficial, especially if the program can consolidate coverage across multiple sites (more on this later).

One other driver for some health care systems is the need to cover areas with on-site nurse practitioners and physician assistants. Using a telehospitalist makes it easier to get appropriate and required oversight for this coverage model across time and space.

What are the advantages of being a telehospitalist?

Some of the career advantages of being a telehospitalist include the shift flexibility and convenience. This work allows a hospitalist to serve a shift from anywhere in the world and from the convenience of their home. Some telehospitalists can easily work local night shifts when they live many time zones away (and therefore, don’t actually have to work a night shift). Many programs are designed to have a single hospitalist cover many hospitals over a wide geography, which would be logistically impossible to do in person. This is especially appealing for multihospital systems that cannot afford to have a hospitalist on site at each location.

The earning potential can also be appealing, depending on the number of shifts a hospitalist is willing to work.

What are the limitations of being a telehospitalist?

There are limits to what a telehospitalist can perform, many of which depend on the manner in which the program and the technology are arranged. Telehealth can vary from a cart-based videoconferencing system that is transported into a patient’s room to an independent robot that travels throughout sites. The primary limitation is the need to rely on someone in the patient’s room to act as virtual hands. This usually falls to the bedside nurse and requires a good working relationship and patience on their part. The bedside nurses have to “buy into” the program in advance and may need to have scripting for how to explain the process to the patients.

Another major challenge is interacting with different electronic health record systems. Becoming agile with a single EHR is challenging enough, but maneuvering several of them in a single shift can be extremely trying. Telehospitalists can also be challenged by technology glitches or failures that need troubleshooting both on their end and on-site. Although these problems are rare, there will always be a concern that the patient will not get his or her needs met if the technology fails.

 

 

How does the financing work?

Although this is a rapidly changing landscape, telehospitalists are not currently able to generate much revenue from professional billing. Unlike in-person visits, Medicare will not reimburse professional fees for telehospitalist visits. Although each payer is unique, most other (nonMedicare) payers are also not willing to reimburse for televisits. This may change in the future, however, as Medicare does pay for virtual specialty services such as telestroke. In addition, many states have enacted telemedicine parity laws, which require private payers to pay for all health care services equally, regardless of modality (audio, video, or in person).

For now, the financial case for employing telehospitalists for most programs has to be made using benfits other than the generation of professional fees. For telehospitalist programs that can cover several sites, the cost is substantially less than employing individual on-site hospitalists to do low-volume work. Telehospitalist programs are also, likely, less costly than is locum tenens staffing. For programs that evaluate the need for transfers, a case can be made that keeping a patient in a smaller, low-cost venue, rather than transferring them to a larger, higher-cost venue, can also reduce overall cost for a health care system.

What about licensing and credentialing?

Telehospitalists can be hindered by the need to have a license in several states and to be credentialed in several systems. This can be cumbersome, time-consuming, and expensive. To ease the multistate licensing burden, the Interstate Medical Licensure Compact has been established.2 This is an accelerated licensure process for eligible physicians that improves license portability across states. There are currently 18 states that participate, and the number continues to increase.

For credentialing, most hospitals require initial credentialing and full recredentialing every 2 years. Maintaining credentials at several sites can be extremely time consuming. To ease this burden, some hospitals with telehealth programs have adopted “credentialing by proxy,” which means that one hospital will accept the credentialing process of another facility.

What next?

In summary, there has been and will likely continue to be explosive growth of telehospitalist programs and providers for all the reasons outlined above. Although some barriers to efficient and effective practice do exist, many of those barriers are being overcome quite rapidly. I expect this growth to continue for the betterment of hospitalists, our patients, and the systems in which we work. For a more in-depth look into telemedicine in hospital medicine, view a report created by a work group of SHM's Practice Management Committee.

Dr. Scheurer is a hospitalist and chief quality officer at the Medical University of South Carolina in Charleston. She is physician editor of The Hospitalist. Email her at [email protected].

References

1.Cisco. (2013 March 4). Cisco Study Reveals 74 Percent of Consumers Open to Virtual Doctor Visit. Cisco: The Network. Retrieved from https://newsroom.cisco.com/press-release-content?type=webcontent&articleId=1148539.

2. Interstate Medical Licensure Compact Commission. (2017). Interstate Medical Licensure Compact. Retrieved from http://www.licenseportability.org/index.html.

Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME

Improved Access to Drug Safety Labeling Changes Information

Article Type
Changed
Wed, 01/31/2018 - 13:51

The FDA has made it easier and faster for health care professionals (HCPs) to get up-to-date drug safety information for the more than 18,000 approved drugs via its Drug Safety Labeling Changes (SLCs) database. The FDA Center for Drug Evaluation and Research recently launched a new searchable and downloadable database for SLCs information (http://www.fda.gov/slc). In most cases, the improved website provides supplemental labeling information within days of a safety label change. Now when a physician or other HCP prescribes a medicine using an e-prescribing system, the updated drug safety information displays much faster than it did with the previous safety labeling changes system. Here’s how.

Shortly after FDA approval of the new drug safety information for an existing drug, the information is entered into the safety labeling changes database. Health information technology (IT) vendors that provide clinical and drug information support for hospitals and pharmacies are then alerted to integrate the updated data into their systems as well. Instead of waiting weeks for the monthly release of all safety labeling updates, this information now is accessible within days.

Although SLCs have been available online for many years, previously they were aggregated and posted only monthly. This time frame meant that if a new safety concern was reflected in an approved labeling change early in a month, then the information was not publicly posted until the following month—4 to 5 weeks later. The FDA recognized the need to apply new digital functionalities to shorten the time between an SLC approval and the public availability of the safety information. Between January 2015 and July 2016, FDA made more than 1,500 SLCs (Table).

As health care professionals know, the “labeling” of a medicine includes detailed information provided in the package insert that accompanies the drug whether it’s on the box, inside the product box, or folded and glued to the lid of a bottle. The product labeling includes a summary for the safe and effective use of the drug and is generally intended for use by prescribers and pharmacists.

However, when a drug is approved, not every safety concern or risk potential can be identified or known. Safety information can change multiple times over the lifetime of a drug as the FDA learns about new risks, interactions with other medications, and adverse effects.

After the FDA becomes aware of new safety information, changes to the product labeling may be required. That’s why postmarketing safety oversight is essential to learn more about the effects of medicines when they are used by a large number of people over a long period. If new safety concerns emerge after a medicine is used in a real-world setting, the FDA may require a “Safety Labeling Change.” The FDA’s new, faster connection between updated safety information and safety alerts on the pharmacy computer system can help build improved confidence into each drug prescription.

The new SLCs website contains a database of changed safety information from all sections of the label that addresses a drug’s safety, including:

  • Boxed warning
  • Contraindications
  • Warnings and precautions
  • Adverse reactions
  • Drug interactions
  • Use in specific populations
  • Patient counseling information/patient information/medication guide

Health care providers, health IT vendors, and the public now have access to critical safety data that can impact the health of a patient faster than before.

Providing drug safety labeling changes quickly to health care vendors facilitates having the data further integrated into systems frequently accessed by HCPs. It also carries SLC data downstream for integration into drug information systems and other electronic venues, such as social media, news feeds, and websites, with vast reach among health care professionals, patients, and consumers. Some of these include WebMD, Medscape, American Society of Health-System Pharmacists, PDR.net, Epocrates, First Databank, and Yahoo Health.

The data files are downloadable in a comma-separated values format—a feature that allows information to be gathered faster. There also are hyperlinks to the labeling revisions at Drugs@FDA, and notifications are sent to subscribers via an RSS feed.

The FDA continues to pursue and provide innovative ways to rapidly access important information that protects and advances public health and will work to better identify class labeling changes. The FDA’s primary goal for the redesigned SLC Internet interface is to deliver drug safety labeling changes as quickly and efficiently as possible, to help create and promote better patient health.

Article PDF
Issue
Federal Practitioner - 34(5)
Publications
Topics
Page Number
44-45
Sections
Article PDF
Article PDF
Related Articles

The FDA has made it easier and faster for health care professionals (HCPs) to get up-to-date drug safety information for the more than 18,000 approved drugs via its Drug Safety Labeling Changes (SLCs) database. The FDA Center for Drug Evaluation and Research recently launched a new searchable and downloadable database for SLCs information (http://www.fda.gov/slc). In most cases, the improved website provides supplemental labeling information within days of a safety label change. Now when a physician or other HCP prescribes a medicine using an e-prescribing system, the updated drug safety information displays much faster than it did with the previous safety labeling changes system. Here’s how.

Shortly after FDA approval of the new drug safety information for an existing drug, the information is entered into the safety labeling changes database. Health information technology (IT) vendors that provide clinical and drug information support for hospitals and pharmacies are then alerted to integrate the updated data into their systems as well. Instead of waiting weeks for the monthly release of all safety labeling updates, this information now is accessible within days.

Although SLCs have been available online for many years, previously they were aggregated and posted only monthly. This time frame meant that if a new safety concern was reflected in an approved labeling change early in a month, then the information was not publicly posted until the following month—4 to 5 weeks later. The FDA recognized the need to apply new digital functionalities to shorten the time between an SLC approval and the public availability of the safety information. Between January 2015 and July 2016, FDA made more than 1,500 SLCs (Table).

As health care professionals know, the “labeling” of a medicine includes detailed information provided in the package insert that accompanies the drug whether it’s on the box, inside the product box, or folded and glued to the lid of a bottle. The product labeling includes a summary for the safe and effective use of the drug and is generally intended for use by prescribers and pharmacists.

However, when a drug is approved, not every safety concern or risk potential can be identified or known. Safety information can change multiple times over the lifetime of a drug as the FDA learns about new risks, interactions with other medications, and adverse effects.

After the FDA becomes aware of new safety information, changes to the product labeling may be required. That’s why postmarketing safety oversight is essential to learn more about the effects of medicines when they are used by a large number of people over a long period. If new safety concerns emerge after a medicine is used in a real-world setting, the FDA may require a “Safety Labeling Change.” The FDA’s new, faster connection between updated safety information and safety alerts on the pharmacy computer system can help build improved confidence into each drug prescription.

The new SLCs website contains a database of changed safety information from all sections of the label that addresses a drug’s safety, including:

  • Boxed warning
  • Contraindications
  • Warnings and precautions
  • Adverse reactions
  • Drug interactions
  • Use in specific populations
  • Patient counseling information/patient information/medication guide

Health care providers, health IT vendors, and the public now have access to critical safety data that can impact the health of a patient faster than before.

Providing drug safety labeling changes quickly to health care vendors facilitates having the data further integrated into systems frequently accessed by HCPs. It also carries SLC data downstream for integration into drug information systems and other electronic venues, such as social media, news feeds, and websites, with vast reach among health care professionals, patients, and consumers. Some of these include WebMD, Medscape, American Society of Health-System Pharmacists, PDR.net, Epocrates, First Databank, and Yahoo Health.

The data files are downloadable in a comma-separated values format—a feature that allows information to be gathered faster. There also are hyperlinks to the labeling revisions at Drugs@FDA, and notifications are sent to subscribers via an RSS feed.

The FDA continues to pursue and provide innovative ways to rapidly access important information that protects and advances public health and will work to better identify class labeling changes. The FDA’s primary goal for the redesigned SLC Internet interface is to deliver drug safety labeling changes as quickly and efficiently as possible, to help create and promote better patient health.

The FDA has made it easier and faster for health care professionals (HCPs) to get up-to-date drug safety information for the more than 18,000 approved drugs via its Drug Safety Labeling Changes (SLCs) database. The FDA Center for Drug Evaluation and Research recently launched a new searchable and downloadable database for SLCs information (http://www.fda.gov/slc). In most cases, the improved website provides supplemental labeling information within days of a safety label change. Now when a physician or other HCP prescribes a medicine using an e-prescribing system, the updated drug safety information displays much faster than it did with the previous safety labeling changes system. Here’s how.

Shortly after FDA approval of the new drug safety information for an existing drug, the information is entered into the safety labeling changes database. Health information technology (IT) vendors that provide clinical and drug information support for hospitals and pharmacies are then alerted to integrate the updated data into their systems as well. Instead of waiting weeks for the monthly release of all safety labeling updates, this information now is accessible within days.

Although SLCs have been available online for many years, previously they were aggregated and posted only monthly. This time frame meant that if a new safety concern was reflected in an approved labeling change early in a month, then the information was not publicly posted until the following month—4 to 5 weeks later. The FDA recognized the need to apply new digital functionalities to shorten the time between an SLC approval and the public availability of the safety information. Between January 2015 and July 2016, FDA made more than 1,500 SLCs (Table).

As health care professionals know, the “labeling” of a medicine includes detailed information provided in the package insert that accompanies the drug whether it’s on the box, inside the product box, or folded and glued to the lid of a bottle. The product labeling includes a summary for the safe and effective use of the drug and is generally intended for use by prescribers and pharmacists.

However, when a drug is approved, not every safety concern or risk potential can be identified or known. Safety information can change multiple times over the lifetime of a drug as the FDA learns about new risks, interactions with other medications, and adverse effects.

After the FDA becomes aware of new safety information, changes to the product labeling may be required. That’s why postmarketing safety oversight is essential to learn more about the effects of medicines when they are used by a large number of people over a long period. If new safety concerns emerge after a medicine is used in a real-world setting, the FDA may require a “Safety Labeling Change.” The FDA’s new, faster connection between updated safety information and safety alerts on the pharmacy computer system can help build improved confidence into each drug prescription.

The new SLCs website contains a database of changed safety information from all sections of the label that addresses a drug’s safety, including:

  • Boxed warning
  • Contraindications
  • Warnings and precautions
  • Adverse reactions
  • Drug interactions
  • Use in specific populations
  • Patient counseling information/patient information/medication guide

Health care providers, health IT vendors, and the public now have access to critical safety data that can impact the health of a patient faster than before.

Providing drug safety labeling changes quickly to health care vendors facilitates having the data further integrated into systems frequently accessed by HCPs. It also carries SLC data downstream for integration into drug information systems and other electronic venues, such as social media, news feeds, and websites, with vast reach among health care professionals, patients, and consumers. Some of these include WebMD, Medscape, American Society of Health-System Pharmacists, PDR.net, Epocrates, First Databank, and Yahoo Health.

The data files are downloadable in a comma-separated values format—a feature that allows information to be gathered faster. There also are hyperlinks to the labeling revisions at Drugs@FDA, and notifications are sent to subscribers via an RSS feed.

The FDA continues to pursue and provide innovative ways to rapidly access important information that protects and advances public health and will work to better identify class labeling changes. The FDA’s primary goal for the redesigned SLC Internet interface is to deliver drug safety labeling changes as quickly and efficiently as possible, to help create and promote better patient health.

Issue
Federal Practitioner - 34(5)
Issue
Federal Practitioner - 34(5)
Page Number
44-45
Page Number
44-45
Publications
Publications
Topics
Article Type
Sections
Disallow All Ads
Content Gating
No Gating (article Unlocked/Free)
Alternative CME
Article PDF Media